Category: 3. Business

  • Asian Stocks Set to Extend Gains on Fed Cut Hopes: Markets Wrap

    Asian Stocks Set to Extend Gains on Fed Cut Hopes: Markets Wrap

    (Bloomberg) — Asian stocks were set for a third day of gains, tracking similar advances on Wall Street as weak US consumer data lifted bets of a Federal Reserve interest rate cut next month.

    Equity-index futures pointed to strong starts for Japan and Australia, with a more modest increase for Hong Kong, at Wednesday’s open. In the US, the S&P 500 rose 0.9% and the Nasdaq 100 climbed 0.6% in choppy sessions as Alphabet Inc. threatened Nvidia Corp.’s dominance in the artificial intelligence sector.

    Delayed economic reports out of the US further cemented bets for a Fed cut in December, with traders now pricing in a roughly 90% chance. Retail sales rose modestly in September, suggesting consumer spending is cooling after months of strong demand. While wholesale inflation picked up, consumer confidence in November saw its steepest drop since April.

    “Downbeat economic data is delivering gains to stock and bond bulls alike, as weaker-than-expected retail sales and consumer confidence numbers coincide with accelerating job losses and rising odds of a December Fed cut,” said José Torres, senior economist at Interactive Brokers.

    The latest US economic reports have taken on added weight ahead of the Fed’s December meeting, given the lack of fresh data. Governor Stephen Miran underscored that outlook by reaffirming his belief that the US economy requires substantial interest‑rate reductions. While the Fed typically adjusts rates in 25‑basis‑point increments, it has on occasion moved by 50 basis points or more.

    White House National Economic Council Director Kevin Hassett’s emergence as the frontrunner to replace the Fed chair helped drive Treasury yields down, with the 10‑year slipping to 4% for the first time in a month. The dollar slipped 0.3%.

    Traders bolstered bets on lower rates over the next year, reflecting the view that a Hassett‑led Fed would deliver the aggressive cuts that President Donald Trump has advocated.

    “The argument will be a weaker US dollar, lower front-end rates from May’s meeting onwards and steeper curves,” said Jordan Rochester, a head of macro strategy at Mizuho in London. Hassett is “a credible economist by background, previously working at the Fed as a senior economist, but some may argue his closeness to Trump makes him the patsy.”

    In Asia, recent weakening of the yen is increasing the likelihood of the Bank of Japan raising its benchmark rate next month, according to a former executive director of the central bank. The currency hit a fresh 10-month low against the dollar last week and is fueling inflationary pressure via higher import costs.

    AI Battle

    Alphabet shares jumped 1.6%, moderating earlier gains, after a report that Meta Platforms Inc. was in talks to spend billions on Google’s artificial-intelligence chips. Nvidia shares dropped 2.6%, pulling back from gains it made in Monday’s tech-fueled rally.

    “Nvidia’s dominant position is unlikely to be fundamentally threatened in the short-term, but markets are all about forward expectations, and it certainly seems like Alphabet is poised to snatch market share away from Jensen Huang’s empire,” said Chris Beauchamp, chief market analyst at IG.

    The rally in Alphabet shares is poised to shake up the rankings of the world’s most valuable companies. A potential changing of the guard comes at a time when the AI industry has come under scrutiny, with stretched valuations causing some volatility.

    Elsewhere in commodities, oil fell as signs of progress in peace talks between Ukraine and Russia buoyed expectations that Moscow’s supply will stay online. Gold closed little changed.

    Some of the main moves in markets:

    Stocks

    Nikkei 225 futures rose 1% as of 7:26 a.m. Tokyo time Hang Seng futures rose 0.3% S&P/ASX 200 futures rose 1.1% Currencies

    The Bloomberg Dollar Spot Index fell 0.3% The euro was little changed at $1.1571 The Japanese yen was little changed at 156.00 per dollar The offshore yuan was little changed at 7.0825 per dollar The Australian dollar was little changed at $0.6467 Cryptocurrencies

    Bitcoin was little changed at $87,086.84 Ether was little changed at $2,928.49 Bonds

    Australia’s 10-year yield was little changed at 4.43% This story was produced with the assistance of Bloomberg Automation.

    ©2025 Bloomberg L.P.

    Continue Reading

  • Meatpacker JBS agrees to merge its leather assets with the ones from Viva

    Meatpacker JBS agrees to merge its leather assets with the ones from Viva

    SAO PAULO, Nov 25 (Reuters) – Brazilian meatpacker JBS (Z98.F), opens new tab said on Tuesday it had signed a binding memorandum of understanding with the shareholders of Viva to combine both firms’ assets related to leather production and commercialization.

    In a securities filing, JBS said the new company will be called JBS VIVA and will be owned 50% by JBS and 50% by Viva’s shareholders — Vanz Holding and Viposa.

    Sign up here.

    The company will process more than 20 million leathers per year, with 31 factories and over 11,000 employees, JBS said, adding that the deal still lacks conditions including the signature of definitive agreements.

    JBS will name the chairman and the Chief Financial Officer of JBS VIVA, while Viva’s shareholders will appoint the Chief Executive Officer and the Chief Operating Officer, according to JBS.

    Reporting by Andre Romani, Editing by Natalia Siniawski

    Our Standards: The Thomson Reuters Trust Principles., opens new tab

    Continue Reading

  • Officials find source of leak in Olympic pipeline two weeks after first report | US news

    Officials find source of leak in Olympic pipeline two weeks after first report | US news

    Investigators have identified the source of a leak in the Olympic pipeline two weeks after fuel was first spotted in a ditch near an Everett, Washington, blueberry farm.

    Oil and gas company BP, the operator of the pipeline, shared in a statement that it had determined the leak occurred in a 20in pipeline and not a neighboring 16in pipeline, allowing that pipeline to be restarted.

    “Repair plans for the 20-inch segment are being developed and a timeline for repair and restart will be shared when available,” BP said.

    The news follows announcements by the Washington and Oregon governors, Bob Ferguson and Tina Kotek, respectively, declaring states of emergency due to the disruptions in fuel supplies. The Olympic pipeline carries gasoline, diesel, jet fuel and other petroleum products to both states, including 90% of Oregon’s transportation fuel and much of the Seattle-Tacoma international airport’s jet fuel.

    The leak was first reported on 11 November between the Washington towns of Everett and Snohomish. The state department of ecology determined the leak consisted of a combination of gasoline, jet fuel and diesel. BP shut off two pipelines that ran side by side in the Olympic pipeline system to determine the source of the leak, either a 16in or a 20in pipeline.

    On 16 November, the company restarted the 16in pipeline, but shut if off again after observing “an increase in product observed in a collection point”, it said in a statement.

    Later that week, on 19 November, Ferguson issued a state of emergency in Washington, waiving state regulations to allow commercial vehicle operators to drive longer hours to transport jet fuel to Seattle-Tacoma airport.

    Kotek followed suit in Oregon on Monday, declaring a similar state of emergency and waiver of commercial driving regulations.

    In statements to Reuters on Monday, major airlines operating through Seattle-Tacoma and the airport itself said they had developed contingencies to prevent disruptions to holiday travel.

    “We do not expect disruption to our operations at Seattle-Tacoma international airport through the Thanksgiving travel week,” Alaska Airlines said, adding that it had brought extra fuel into Seattle on inbound flights and additional trucking shipments, and added fuel stops to certain flights.

    Delta Air Lines similarly said it had transported additional fuel to the airport and added refueling stops to some long-haul flights.

    On Monday, BP reported that it had excavated “over 200 feet of pipeline” and expected to “continue overnight operations tonight”. By Tuesday morning, the company had found the source of the leak.

    In updated statements to Reuters on Tuesday, Delta said it “is operating our full Seattle hub schedule and has discontinued fuel stops on select long-haul flights”. Alaska added that it had “discontinued all planned fuel stops but will continue to tanker and truck in additional fuel on a reduced basis as the pipeline increases to normal capacity”.

    Repairs to the 20in pipeline come as Washington state’s ecology department has fined BP $3.8m for a 2023 gasoline spill from the Olympic pipeline. The Olympic pipeline has leaked at least 13 times since 1999, when a leak near Bellingham caused an explosion that killed a teenager and two younger children. According to the Pipeline Safety Trust, a Washington state-based non-profit, the pipeline has leaked three times in 2025.

    “These incidents have caused over $100m in property damage,” Kenneth Clarkson, spokesperson for the Pipeline Safety Trust, said in a statement to the Associated Press. “Olympic Pipeline must explain what has changed and what they’re doing to stop it.”

    Continue Reading

  • Getty Images (US) Inc (and others) v Stability AI LimitedInput: Getty Images v Stability AIOutput: Continued Uncertainty for AI and IP in the UK… | Publications | Insights & Events

    Getty Images (US) Inc (and others) v Stability AI LimitedInput: Getty Images v Stability AIOutput: Continued Uncertainty for AI and IP in the UK… | Publications | Insights & Events

    On 4 November 2025 the UK High Court
    handed down its judgment in the case of
    Getty Images (US) Inc (and others) v Stability
    AI Limited [2025] EWHC 2863 (Ch)
    [High
    Court Judgment Template].

    As one of the first and to date most high-profile intellectual
    property (IP) infringement claims against an AI developer to
    make it all the way to trial in the UK courts, the case was
    originally envisaged as having potential to provide muchneeded
    wide-ranging judicial guidance on the application
    of existing UK IP law in the field of AI. However, as the
    case progressed and the scope of Getty’s claims gradually
    reduced to a shadow of the original, it became apparent that
    this judgment, whilst still of note in respect of a number of
    key issues, would not be the silver bullet which many had
    originally anticipated.

    The Judgment in Brief

    At over 200 pages (alongside an accompanying glossary of
    key technical terms and appendix concerning the context in
    which an average consumer would encounter certain Getty
    registered trade marks) the judgment is long and complex,
    including detailed discussion of the witness and expert
    evidence which the Court considered before reaching its
    findings.

    Read full insight to learn more.

    Continue Reading

  • Journal of Medical Internet Research

    Journal of Medical Internet Research

    The increased life expectancy is one of the greatest achievements of global health systems. However, this increase has led to a substantial rise in age-related neurological disorders, particularly Alzheimer disease and other dementias, necessitating health policies to focus [], not only on survival but also on minimizing health loss due to disability by promoting function and independence []. Estimation suggested an increase from 57 million people living with dementia in 2019 to 153 million by 2050 []. Between 1990 and 2021, the disability-adjusted life years attributed to dementia increased by 168.7% (95% CI 156.3-179.9), driven by population aging []. By 2021, dementia ranked as one of the top 10 neurological conditions globally, with a particularly high burden in people aged 80 years and older []. Mild cognitive impairment (MCI) is a bidirectional transitional stage between dementia and normal cognitive aging, often with unrecognized early symptoms, with 10%-15% of affected individuals progressing to dementia annually []—a risk 3.3 times higher than that of the general older population []. Therefore, MCI represents a critical window for dementia prevention and intervention, with early identification and intervention being essential for reducing dementia incidence. Achieving a cost-effective community intervention has become a critical priority in public health [].

    Exercise intervention and cognitive training (CT) can effectively improve cognitive functions []. Mind-body exercise, such as Taichi and yoga, is a promising exercise type incorporating physical, cognitive, social, and mindfulness components in exercise [] that may have a better cognitive management effect than other types of exercise [].

    Recent evidence suggests that both mind-body exercises and technology-enhanced interventions show promise for MCI prevention []. Systematic reviews demonstrate that Taichi significantly improves cognitive function in older adults with MCI, particularly executive function and memory []. Similarly, virtual reality (VR) technology has emerged as a powerful cognitive rehabilitation tool, offering ecological validity and real-time adaptability that traditional training lacks. VR-based interventions demonstrate significant improvements in memory, attention, and executive function in populations with MCI [,]. However, fewer studies synerized the potential effects of combining traditional mind-body exercise with VR technology using adaptive intervention strategies.

    Systematic reviews and meta-analyses have shown that dual-task intervention combining cognitive function training and physical activity is more effective in improving cognition and physical function than physical activity intervention [,]. This synergistic effect is explained by the guided plasticity facilitation framework, whereby exercise facilitates neuroplasticity by increasing brain-derived neurotrophic factor production and cell proliferation, while cognitive interventions guide this plasticity by enhancing the survival of exercise-induced new cells. Additionally, multicomponent exercise research confirms positive effects on global cognition (effect size=0.32, 95% CI 0.03-0.61), particularly when aerobic exercise is included, supporting the mechanistic basis of the muscle-brain axis in exercise-induced neuroprotection [,].

    The Healthy Ageing Through Internet Counselling in the Elderly (HATICE) trial is a multinational preventive intervention study based on coaching support and is a low-cost and scalable intervention model. HATICE provides remote personalized adaptive intervention through coaching, reduces the burden of cardiovascular disease, and improves cognitive function []. STRONGER 60+ also pays attention to the need for adaptive intervention. This model is based on the multicomponent intervention of The Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER) [,], but focuses more on how to achieve adaptive intervention []. However, current clinical guidelines and expert consensus on multidomain intervention for MCI and dementia prevention provide a range of content and duration that is too wide to make the specific guidance for adaptation in community and clinical practice [,,,], and the current training components and duration of multidomain intervention still depend on experience and experience-based approaches [].

    The dose-response relationship study of CT found that there is an optimal point for the intervention duration, and the optimal duration of the intervention is affected by the age of the participants []. Observational studies have also found that age and comorbidities affect the prevention and progression of MCI [,]. Meanwhile, inappropriate strategy selection might reduce treatment adherence and compromise the critical intervention window []. Therefore, how to achieve adaptive intervention, explore which factors, and formulate adaptive intervention duration (or doses) based on these factors are the key to nonpharmacological prevention of MCI and remain to be developed.

    We used a 2-stage sequential, multiple assignment, randomized trial (SMART) design to develop evidence-based adaptive intervention strategies [] for optimizing cognitive function among older adults with MCI. The SMART design involved CT combined with either offline Taichi (OffTC) or virtual reality Taichi (VRTC). Participants who did not respond adequately to initial treatment (early treatment nonresponders) were rerandomized to receive either treatment intensification or modified treatment components.

    The primary aim was to compare the effectiveness of CT+OffTC versus CT+VRTC versus control on long-term cognitive outcomes. Among early treatment nonresponders, we evaluated whether treatment intensification or component modification produced superior cognitive improvements. We hypothesized that both active interventions would demonstrate greater cognitive benefits compared with control and that treatment intensification would be more effective than component modification among nonresponders. The secondary aims assessed baseline characteristics that predict treatment response and effect modification. We hypothesized that age, baseline cognitive function, and comorbid conditions would influence both response status and treatment effectiveness.

    Study Design and Setting

    The study was conducted in 3 randomly selected districts in Shanghai between April and December 2023. A multistage stratified cluster random sampling method was used for participant recruitment. Based on geographical location and economic development level, 3 districts were randomly selected from the Shanghai municipality. Within each selected district, 2 subdistricts were randomly chosen, and subsequently, 3 communities were randomly selected from each subdistrict using cluster sampling. This sampling approach ensured geographical and socioeconomic diversity in the study population while maintaining feasibility for intervention delivery through existing community health service infrastructure.

    This study was reported according to the Consolidated Standards of Reporting Trials (CONSORT 2025) for randomized trials, detailed in [].

    Treatments

    OffTC intervention implemented the standardized 24-form simplified Taichi exercise regimen, developed by the National Sports Administration. Modifications were implemented for balance-intensive movements (right kick, double ear boxing, left turning kick, left standing stance, and right standing stance) to enhance safety while maintaining form integrity.

    The VRTC intervention used identical 24-form simplified Taichi movements implemented through an immersive technological interface. The VR system incorporated precise arm-length calibration to accurately track upper and lower extremity movement trajectories.

    CT used the Thoven Cognitive Training System (TCSA-BOT, developed by Shanghai Thoven Intelligent Technology Co, Ltd), a comprehensive platform targeting multiple cognitive domains: memory, attention, executive function, logical reasoning, and reaction time. The system was specifically adapted for this research protocol and implemented via a customized WeChat mini-program interface.

    Procedures

    This study used a 24-week SMART design, comprising two 12-week intervention phases. In the first stage, the intervention consisted of 1 hour of weekly CT combined with 1 hour of weekly exercise per week, structured as a dual-task training program incorporating both cognitive and physical elements.

    Participants were initially randomized into 3 groups: control group, CT+OffTC, and CT+VRTC. At week 12, participants were assessed based on 2 questions assessed by a 5-point Likert scale: (1) perceived effectiveness of their current intervention and (2) proficiency in both CT and Taichi exercises. Participants would be marked as nonresponders, if any question scored less than 3 points.

    For the second phase, CT+OffTC responders continued their original intervention protocol. Nonresponders from CT+OffTC were randomly assigned to either CT+VRTC or doubled doses of their original intervention (2CT+2OffTC). Similarly, CT+VRTC responders maintained their initial intervention, while nonresponders were randomly assigned to either CT+OffTC or doubled doses of their original intervention (2CT+2VRTC).

    All interventions were conducted at designated community health centers within the 3 participating districts in Shanghai. OffTC sessions were led by certified Yang-style Taichi instructors with a minimum of 5 years of experience teaching older adults, while VRTC sessions used Meta Quest with Guided Tai Chi software (developer: Cubicle Ninjas) providing immersive virtual environments featuring professionally recorded Yang-style Taichi instruction and real-time movement tracking without requiring external controllers or television displays. Each VRTC session included 5-minute acclimatization periods and integrated safety features. CT was delivered through the TCSA-BOT WeChat mini-program interface, with research staff supervision during weekly 60-minute community center sessions and optional home access via participants’ smartphones. Control group health education consisted of weekly 45-minute group sessions covering general wellness topics, delivered by qualified health educators in the same community facilities, detailed intervention program in and VRTC sample video in .

    Randomization and Masking

    After baseline assessment, participants were randomly assigned to the intervention groups or the control groups. In the first 12 weeks of the 24-week SMART intervention, participants in the intervention group were randomly assigned to receive either weekly 1-hour CT plus 1-hour Taichi or CT plus VRTC. In the 12th week, participants were assessed for intervention mastery, with nonresponders those who are unable to master the content of the intervention, the Likert 5-point scale evaluates the mastery of the intervention content to be less than 3, being rerandomized to alternative or intensified interventions. The control group received only health education and routine care. Randomization was performed with the use of a computer-based code generated by members of the research team at the School of Public Health at the Shanghai Jiao Tong University School of Medicine. Due to the dosage assignment of the intervention being changed during treatment, study participants and staff were aware of treatment allocation, but researchers responsible for data analysis were masked to the allocation groups.

    Participants

    Participants were recruited from 3 randomly selected districts in Shanghai. We screened community-dwelling older adults using the Montreal Cognitive Assessment (MoCA) and Memory Guard score (MGs).

    Study eligibility criteria included: age ≥60 years, MCI diagnosis established through positive screening on the MGs, and meeting the 2018 Chinese guidelines for MCI diagnosis []. Exclusion criteria encompassed: communication disorders, severe impairment in activities of daily living, presence of metal implants, severe psychiatric disorders, illiteracy, cognitive decline attributable to other pathologies, history of neurological diseases, exercise contraindications, color blindness, insufficient education to complete testing protocols, concurrent rehabilitation therapy, or unwillingness to complete the 24-week intervention and follow-up. Additionally, participants with regular exercise experience (eg, Taichi and Baduanjin) within the preceding 3 months were excluded to prevent contamination effects.

    Assessments and Data Sources

    Outcome Measurements

    The primary outcome is cognitive status at 24 weeks (end of the trial), measured by MGs. MoCA was used for baseline cognitive screening and participant categorization.

    The MoCA is a widely validated cognitive screening instrument that assesses multiple cognitive domains including visuospatial and executive functions, naming, memory, attention, language, abstraction, delayed recall, and orientation []. The MoCA uses a 30-point scale, with scores ≤26 indicating possible cognitive impairment []. In our study [], MoCA was administered at baseline to categorize participants into cognitive risk groups.

    MGs is a computerized neuropsychological assessment device that evaluates 6 cognitive domains: orientation, memory, attention, calculation, recall, and language and executive function []. The assessment incorporates both accuracy scores (correct or incorrect responses) and response time data, using machine learning algorithms to provide a comprehensive cognitive evaluation. MGs demonstrates excellent diagnostic performance with an accuracy of 93.75% and an area under the curve of 0.923, achieving high sensitivity (91.67%) and specificity (95.45%) for MCI detection []. The interassessment agreement between MoCA and MGs reached κ=1.0, indicating perfect concordance and validating the reliability of our cognitive measurements. The computerized format allows for standardized administration, objective measurement of cognitive performance, and real-time data collection, making it particularly suitable for detecting cognitive changes in intervention studies.

    Exposures

    The primary exposures in this study were the intervention modalities assigned through the 2-stage SMART design: control, CT+OffTC, and CT+VRTC. Detailed descriptions of each intervention component, delivery methods, and the rerandomization process for nonresponders have been provided in the “Procedures” section above.

    Confounders

    Potential confounding variables were identified based on existing literature on cognitive function interventions and measured at baseline through standardized protocols by trained research staff. Demographic characteristics included age (continuous variable in years, assessed through participant self-report), sex (binary: male or female, obtained from participant demographics), and education level (continuous variable in years). Socioeconomic status was measured through self-reported monthly household income, categorized in Chinese yuan (¥; US $1= ¥7.11).

    Clinical characteristics assessed at baseline included BMI (in kg/m², calculated from measured height and weight using calibrated equipment). Comorbid conditions were assessed as binary variables (yes or no) through self-report, including diabetes (assessed through physician diagnosis or current use of antidiabetic medications), hypertension (assessed through physician diagnosis or current use of antihypertensive medications), cancer, cardiovascular disease, chronic kidney disease, fracture history, respiratory disease, and digestive disease.

    Sample Size

    The intervention was designed to find the optimal adaptive intervention based on individual responses to initial treatment assignments. The sample size of the SMART design relied on the response rate. Response rate indicates whether the intervention is effective. According to the previous studies on physical activity intervention [,] and response rate simulation [,], we hypothesized the response rate of 0.60, the type I error rate of 0.05, a desired half-width of the CI of 0.45, and the required sample size of SMART intervention group is 54 [] based on precision-based sample size calculation.

    Statistical Analysis

    Baseline characteristics were summarized using descriptive statistics, with continuous variables presented as means (SD) and categorical variables as frequencies (percentages). Group comparisons were performed using t test for normally distributed continuous variables and the chi-square test for categorical variables.

    The primary analysis used intention-to-treat principles comparing 24-week memory scores across treatment groups (CT+OffTC, CT+VRTC, and control) using linear regression adjusted for baseline MGs, age, education, sex, hypertension, and diabetes. Pairwise comparisons between groups were conducted with Bonferroni correction for multiple testing. Effect sizes were calculated as Cohen d with 95% CIs.

    Secondary analyses evaluated treatment strategies among early treatment nonresponders (defined as insufficient memory improvement at 12 weeks) using linear regression models comparing treatment intensification versus switching Tai Chi modality, adjusted for 12-week memory scores and baseline characteristics. Dynamic treatment regimen analysis compared embedded treatment sequences using the same covariate-adjusted linear regression approach. Treatment response prediction used linear regression and logistic regression models to identify baseline characteristics associated with 12-week response status. Subgroup analysis was assessed using interaction terms for age group, baseline memory function, hypertension, and diabetes status.

    All statistical tests were 2-sided with a significance threshold of P<.05. Bootstrap resampling (1000 iterations) was used to construct 95% CIs and assess statistical significance.

    Ethical Considerations

    This study was reviewed and approved by the Public Health and Nursing Research Ethics Committee of Shanghai Jiao Tong University School of Medicine (approval number: SJUPN-202008, on November 19, 2020) prior to participant recruitment and registered with the Chinese Clinical Trial Registry on January 27, 2021, ChiCTR2100042748. The study protocol, informed consent procedures, and all study materials received full ethical approval.

    Written informed consent was obtained from all participants prior to enrollment. The consent process included detailed explanations of (1) study purpose, procedures, and duration; (1) potential risks and benefits of participation; (3) voluntary nature of participation and right to withdraw at any time without penalty; (4) data collection, storage, and use procedures; and (5) contact information for study personnel and ethics committee. All participants demonstrated the capacity to provide informed consent through cognitive screening assessments.

    All study data were deidentified using unique participant identification codes, with the linking key stored separately from study data in a secure, password-protected database accessible only to authorized research personnel. Data analysis was conducted using only deidentified datasets.

    Participants received modest compensation for their time and effort, including transportation reimbursement (¥50 [US $7] per visit; currency conversion based on exchange rate as of October 8, 2025) for assessment sessions and a completion incentive (¥200 [US $28] for those who finished the full 24-week intervention period. Compensation was provided regardless of intervention adherence or study outcomes to avoid coercion.

    No images containing identifiable participant information are included in this manuscript or supplementary materials.

    Harms including falls, heat exhaustion, dizziness, and VR-related symptoms were assessed nonsystematically through participant self-report and instructor observation throughout the 24-week intervention period.

    Descriptive Statistics

    Between April and December 2023, a total of 686 individuals were assessed for eligibility, of whom 563 were screened as ineligible due to normal cognitive function. Of the 123 participants who met eligibility criteria, 30 declined participation due to COVID-19 pandemic concerns regarding in-person study visits and health safety protocols, and 1 withdrew prior to randomization, resulting in 92 participants entering Stage I randomization. Participants were allocated to 3 groups: 26 to control, 33 to OffTC+CT, and 33 to VRTC+CT.

    Following Stage I intervention, responders continued their allocated treatments while nonresponders proceeded to Stage II randomization with alternative interventions. In the control group, 20 participants completed the study with 6 withdrawals. Among OffTC+CT participants, 22 responders continued treatment with 21 completing it, while 11 nonresponders were randomized to alternative treatments (6 to 2OffTC+2CT and 5 to VRTC+CT). In the VRTC+CT group, 20 responders completed treatment, while 13 nonresponders underwent Stage II randomization (6 to 2VRTC+2CT and 7 to OffTC+CT) with completion rates of 4/6 and 7/7, respectively ().

    Figure 1. Consolidated Standards of Reporting Trials (CONSORT) flow diagram of participant recruitment, randomization, and retention in a 24-week Sequential Multiple Assignment Randomized Trial examining cognitive training combined with offline Taichi versus virtual reality Taichi for mild cognitive impairment prevention among community-dwelling adults aged ≥60 years in Shanghai, China (April-December 2023).

    The final sample consisted of 81 participants, and the response rate was 64.18% which met the assumption of sample calculation. The study population was predominantly female (65.4% overall), with similar gender distributions across groups (60.0%, 66.7%, and 67.7%, respectively; P=.84), detailed in . All interventions were delivered as intended by certified instructors showing >95% fidelity to standardized procedures.

    Table 1. Baseline demographic, socioeconomic, and clinical characteristics of community-dwelling adults aged ≥60 years with mild cognitive impairment enrolled in a 24-week Sequentiala.
    Variables Overall Control OffTCb+CTc VRTCd+CT P value
    Observation 81 20 30 31
    Female sex, n (%) 53 (65.4) 12 (60.0) 20 (66.7) 21 (67.7) .83
    Education (years), mean (SD) 10.55 (2.94) 11.10 (3.60) 10.63 (3.02) 10.11 (2.39) .50
    Income, CNY (USD), n (%) .53
    < 10,000 (< 1430) 1 (1.2) 0 (0.0) 1 (3.3) 0 (0.0)
    10,001-30,000 (1431-4285) 12 (14.8) 5 (25.0) 5 (16.7) 2 (6.5)
    30,001-60,000 (4286-8570) 49 (60.5) 11 (55.0) 18 (60.0) 20 (64.5)
    60,001-90,000 (8571-12,855) 11 (13.6) 3 (15.0) 4 (13.3) 4 (12.9)
    90,001 120,000 (12,856-17,140) 6 (7.4) 0 (0.0) 2 (6.7) 4 (12.9)
    > 120,001 (> 17,141) 2 (2.5) 1 (5.0) 0 (0.0) 1 (3.2)
    Age (years), mean (SD) 71.38 (6.49) 72.75 (7.53) 72.37 (6.88) 69.55 (5.00) .13
    Diabetes, n (%) 11 (13.6) 4 (20.0) 4 (13.3) 3 (9.7) .57
    Hypertension, n (%) 24 (29.6) 8 (40.0) 8 (26.7) 8 (25.8) .50
    BMI, mean (SD) 24.06 (2.75) 24.87 (2.58) 23.75 (3.03) 23.83 (2.56) .31
    MGse, mean (SD) 27.61 (3.55) 27.99 (3.31) 27.25 (4.31) 27.72 (2.92) .75
    MoCAf, mean (SD) 22.19 (2.15) 21.50 (2.28) 22.20 (2.22) 22.61 (1.93) .19
    Cancer, n (%) 8 (9.9) 2 (10.0) 1 (3.3) 5 (16.1) .24
    Cardiovascular disease, n (%) 9 (11.1) 1 (5.0) 3 (10.0) 5 (16.1) .45
    Chronic kidney disease, n (%) 2 (2.5) 0 (0.0) 1 (3.3) 1 (3.2) .71
    Fracture, n (%) 8 (9.9) 3 (15.0) 2 (6.7) 3 (9.7) .62
    Respiratory disease, n (%) 3 (3.7) 0 (0.0) 3 (10.0) 0 (0.0) .07
    Digestive disease, n (%) 3 (3.7) 0 (0.0) 1 (3.3) 2 (6.5) .48

    aGroupwise comparisons for continuous variables were assessed using either ANOVA or the Kruskal-Wallis test (if not normally distributed), and categorical variables were assessed using the chi-square test.

    bOffTC: offline Taichi.

    cCT: cognitive training.

    dVRTC: virtual reality Taichi.

    eMGs: Memory Guard score.

    fMoCA: Montreal Cognitive Assessment.

    Effect of Interventions

    At 12 weeks, the adjusted mean MGs in the CT+VRTC group were 30.1 (95% CI 28.3-31.9) and 28.1 (95% CI 26.3-29.9) in the CT+OffTC group. The effect of the CT+VRTC group was significantly better than that of the CT+OffTC group, with an increase of 2.03 MGs (95% CI 0.018-3.96; Cohen d=0.558; P=.04; and ).

    Table 2. Treatment effects on Memory Guard scores at 12 weeks and 24 weeks in a Sequential Multiple Assignment Randomized Trial of cognitive training combined with offline Taichi (CT+OffTC) versus virtual reality Taichi (CT+VRTC) versus control among community-dwelling adults aged ≥60 years with mild cognitive impairmenta.
    Timepoint and treatment group Adjusted mean (SE) 95% CI Group comparison Estimate (SE) t-ratio P Value Effect size (95% CI)
    Week 12
    CT+OffTC 28.1 (0.907) 26.3- 29.9 CT+OffTC vs CT+VRTC –2.03 (0.961) –2.117 .04 –0.558 (–1.1 to –0.018)
    CT+VRTC 30.1 (0.900) 28.3-31.9 b
    Week 24
    Control 27.8 (0.903) 26.0 – 29.6 CT+OffTC vs Control 1.49 (1.050) 1.421 .47 0.416 (–0.171 to 1.00)
    CT+OffTC 29.3 (0.803) 27.7 -30.9 CT+VRTC vs Control 5.10 (1.070) 4.778 <.001 1.425 (0.785- 2.06)
    CT+VRTC 32.9 (0.806) 31.3 -34.5 CT+VRTC vs CT+OffTC 3.61 (0.936) 3.857 <.001 1.009 (0.461- 1.56)

    aAdjusted mean Memory Guard scores, pairwise group comparisons, and effect sizes (Cohen d) are presented from linear regression models controlling for baseline Memory Guard scores, age, education, sex, hypertension, and diabetes.

    Table 3. Comparison of treatment effects on Memory Guard scores at 12 weeks and 24 weeks in a Sequential Multiple Assignment Randomized Trial of cognitive training combined with offline Taichi (CT+OffTC) versus virtual reality Taichi (CT+VRTC) versus control among community-dwelling adults aged ≥60 years with mild cognitive impairment.a
    Time point and group comparison Estimate (SE) t-ratio P Value Effect size (95% CI)
    Week 12
    CT+OffTC vs CT+VRTC –2.03 (0.961) –2.117 .04 –0.558 (–1.1 to –0.018)
    Week 24
    CT+OffTC vs Control 1.49 (1.050) 1.421 .47 0.416 (–0.171 to 1.00)
    CT+VRTC vs Control 5.10 (1.070) 4.778 <.001 1.425 (0.785- 2.06)
    CT+VRTC vs CT+OffTC 3.61 (0.936) 3.857 <.001 1.009 (0.461- 1.56)

    aAdjusted mean Memory Guard scores, pairwise group comparisons, and effect sizes (Cohen d) are presented from linear regression models controlling for baseline Memory Guard scores, age, education, sex, hypertension, and diabetes.

    At 24 weeks, the adjusted mean MGs in the CT+VRTC group was the highest, at 32.9 (95% CI 31.3-34.5), followed by the CT+OffTC group, at 29.3 (95% CI 27.7-30.9), and the control group at 27.8 (95% CI 26.0-29.6).

    In pairwise comparisons, the CT+VRTC group showed an improvement of 5.10 (95% CI 2.93-7.27; Cohen d=1.425, 95% CI 0.785-2.060; P<.001) MGs compared with the control group, while the CT+VRTC group showed an improvement of 3.61 (95% CI 1.71-5.51; Cohen d=1.009, 95% CI 0.461-1.560; P<.001) MGs compared with the CT+OffTC group.

    The embedded dynamic treatment plan analysis showed that the efficacy of different adaptive strategies was different (). The VRTC-based plan was always more effective than the control group, among which the responder plan had the highest efficacy, with an efficacy difference of 5.40 (95% CI 3.10-7.70; P<.001) MGs compared with the control group. VRTC intensification showed a significant benefit for nonresponders, with a difference of 5.66 (95% CI 1.14-10.18; P=.01) MGs, while VRTC switch showed a difference of 3.21 (95% CI 0.01-6.41; P=.05) MGs. Among the OffTC-based strategies, only OffTC intensification showed significant improvement compared with the control group, with a difference of 4.24 (95% CI 0.22-8.26; P=.04) MGs.

    Table 4. The comparisons of dynamic treatment regimen strategies on the 24-week Memory Guard score by treatment response and intervention sequencea.
    Strategies Adjusted mean (SE) 95% CI Comparisons P value
    Control 27.6 (1.35) 24.9-30.3 Reference b
    OffTCc Intensify 31.8 (1.89) 28.1-35.6 4.24 (2.01) .04
    OffTC Switch 29.0 (1.70) 25.6-32.4 1.37 (1.83) .45
    OffTC Responder 28.4 (1.13) 26.2-30.7 0.84 (1.14) .46
    VRTCd Intensify 33.3 (2.13) 29.0-37.5 5.66 (2.26) .01
    VRTC Switch 30.8 (1.54) 27.7-33.9 3.21 (1.60) .05
    VRTC Responder 33.0 (1.13) 30.7-35.2 5.40 (1.15) <.001

    aAdjusted means are from models controlling for baseline Memory Guard score, age, and other control variables.

    bNot applicable.

    cOffTC: offline Taichi.

    dVRTC: virtual reality Taichi.

    The results suggest that VRTC-based adaptive interventions provide strong cognitive benefits across response patterns, while OffTC-based approaches show more limited effectiveness, with benefits primarily seen when treatment intensity is increased in nonresponders.

    Factors Predicting the Response Status

    presents the results of response status predictors. For the response status, diabetes was a strong positive predictor (B=17.404, 95% CI 15.913-18.895; P<.001). Baseline MGs, treatment assignment in stage 1, hypertension, and age were not significant. For perceived effectiveness and content mastery, diabetes remained a significant positive predictor as the same. Additionally, lower baseline MGs was associated with higher perceived effectiveness and content mastery.

    Table 5. Association between baseline characteristics and treatment response outcomes at 12 weeks among participants receiving cognitive training combined with Taichi interventionsa.
    (1) Response (2) Perceived effectiveness (3) Master content
    Baseline MGsb
    Unstandardized regression coefficient (95% CI) –0.146 (–0.365 to 0.073) –0.124c (–0.245 to –0.004) –0.115c (–0.225 to –0.004)
    Robust SE –0.604 –0.202 –0.151
    P value .19 .05 .05
    Treatment in stage 1
    Unstandardized regression coefficient (95% CI) –0.061 (–1.331 to 1.208) –0.009 (–0.818 to 0.800) 0.122 (–0.675 to 0.918)
    Robust SE –2.043 –0.599 –0.362
    P value .93 .98 .77
    Age
    Unstandardized regression coefficient (95% CI) 0.001 (–0.120 to 0.122) –0.023 (–0.105 to 0.058) 0.005 (–0.067 to 0.077)
    Robust SE –0.373 –0.163 –0.123
    P value .99 .57 .90
    Hypertension
    Unstandardized regression coefficient (95% CI) –1.209d (–2.606 to 0.188) –0.939d (–1.903 to 0.025) –0.768 (–1.783 to 0.248)
    Robust SE –0.160 –0.044 –0.109
    P value 0.09 .06 .15
    Diabetes
    Unstandardized regression coefficient (95% CI) 17.404e (15.913 to 18.895) 1.788e (0.927 to 2.649) 1.598e (0.705 to 2.492)
    Robust SE 0.360 0.335 0.284
    P value <.001 <.001 .001
    Constant
    Unstandardized regression coefficient (95% CI) 4.926 (–8.173 to 18.026) 8.968c (0.793 to 17.143) 6.561d (–0.550 to 13.672)
    Robust SE 44.669 17.395 13.164
    P value .46 .04 .08
    Observations, n 61 61 61
    R2 f 0.168 0.165

    aValues represent unstandardized regression coefficients with 95% CIs and robust SEs in parentheses. Model (1): Logistic regression; Models (2-3): Ordinary Least Squares regression. Robust SEs in parentheses.

    bMGs: Memory Guard score.

    cP<.05.

    dP<.10.

    eP<.01.

    fNot applicable.

    Subgroup Analysis

    Subgroup analyses revealed significant heterogeneity in treatment effects (), with younger age, lower baseline cognitive function, and the presence of comorbidities benefiting more from interventions.

    Figure 2. Subgroup analyses examining treatment effect heterogeneity on 24-week Memory Guard scores by age group (≤71 vs >71 years), baseline cognitive function (lower vs higher Memory Guard score), and comorbidity status (hypertension and diabetes) in a Sequential Multiple Assignment Randomized Trial comparing control, cognitive training combined with offline Taichi (CT+OffTC), and cognitive training combined with virtual reality Taichi (CT+VRTC) . Adjusted means with 95% CIs from linear regression models controlling for baseline Memory Guard score, age, and other covariates. MGs: Memory Guard score.

    Younger participants (≤71 years) benefit more cognitive improvement from CT+VRTC, achieving 33.3 (95% CI 31.4-35.2) compared with older participants at 31.9 (95% CI 29.4-34.4). Yet, younger patients with MCI were at greater risk of cognitive decline. This result suggested that younger patients with MCI may be more vulnerable to cognitive decline but also benefit more from VR interventions.

    CT+VRTC demonstrated consistent effectiveness across different baseline cognitive levels. However, participants with higher baseline MGs showed greater overall benefit from interventions.

    CT+VRTC maintained stable effectiveness regardless of comorbid conditions. Importantly, participants with comorbid conditions would benefit more cognitive improvement compared with those without comorbidities, suggesting potential synergy between controlling chronic diseases and cognitive function protection.

    Principal Results

    CT+VRTC demonstrated substantial cognitive improvement compared with both control and CT+OffTC groups, with large effect sizes indicating clinically meaningful benefits. Dynamic treatment regimen analysis revealed that VRTC-based adaptive strategies consistently outperformed control across different response patterns, with VRTC responders achieving the highest effectiveness. Subgroup analyses identified significant treatment heterogeneity, with younger participants, those with lower baseline cognitive function, and individuals with comorbid conditions demonstrating enhanced responsiveness to interventions. To our knowledge, this is the first SMART design to develop evidence-based adaptive intervention strategies for MCI prevention in community settings.

    Comparison With Prior Work

    Compared with the FINGER trial [,], our focused population with MCI demonstrated substantially greater cognitive improvements, with CT+VRTC achieving an effect size of 1.425 (95% CI 0.785-2.060; P<.001) compared with control, suggesting that technology-enhanced dual-task training may be effective in individuals with established cognitive impairment. Consistent with emerging digital health interventions, our findings support the superiority of VR-enhanced approaches over traditional exercise modalities [-], with VRTC consistently outperforming OffTC across all treatment strategies. The effects of VRTC suggest unique advantages of VR technology in cognitive intervention.

    The immersive, multisensory environment inherent in VR-based training appears to impose greater demands on sensorimotor integration and movement precision, potentially enhancing cognitive engagement through increased activation of frontoparietal networks []. Furthermore, the enhanced real-time feedback mechanisms in VR environments may facilitate neuroplastic adaptations, particularly in frontocortical regions critical for executive function and motor planning []. In addition, VRTC may strengthen cortical connectivity and increase frontal lobe activation patterns, via upregulation of brain-derived neurotrophic factor in hippocampal and prefrontal regions, enhanced recruitment of dorsal and ventral attentional networks, increased cerebellum-prefrontal functional connectivity, and modulation of default mode network activity to reduce age-related network dedifferentiation [,,]. Individuals who participated in the technology-enhanced dual-task group (CT+VRTC) showed more stable intervention effects regardless of whether they had chronic diseases or not and their baseline cognitive status.

    The enhanced treatment responsiveness observed in participants with comorbidities reflects potential synergistic effects between intervention modalities and chronic disease management. Patients with comorbidities benefited more, which is consistent with the findings of current intervention studies [,]. Hypertension can lead to white matter injury through small vessel disease in the brain, while diabetes is associated with insulin resistance, inflammatory responses, and oxidative stress, all of which contribute to neurodegenerative processes []. Therefore, cognitive impairment in these patients may result from multiple interrelated factors. Intervention strategies may exert beneficial effects by improving vascular function, reducing inflammation, or enhancing neuroprotective pathways [,].

    Our predictive modeling showed that diabetes consistently predicted enhanced response, while hypertension showed opposite effects. These findings suggest that patients with comorbidity may benefit more from intervention. The immersive cognitive-motor training may help counteract diabetes-induced neuroinflammation and vascular dysfunction through enhanced neuroplasticity, while the engaging nature of VR technology may improve treatment adherence and motivation—factors that are often compromised in patients with chronic metabolic disorders []. These predictive results were aligned with subgroup analysis that patients with comorbidity may benefit more from intervention modality with high confidence.

    Beyond the benefits observed of individual intervention components, our study showed the importance of adaptive treatment strategies. Traditional cognitive interventions have predominantly used fixed protocols with predetermined duration and intensity, failing to account for individual variability in treatment response. The coaching-based intervention represented an early attempt at adaptive intervention [-], using remote support to prevent cognitive function decline. However, these adaptations remained largely experience-driven rather than systematically evidence-based. Our study addressed limitations of subjective feelings as response indicators by examining the agreement between subjective response assessment and objective assessment criteria at 12 weeks. Subjective assessment used patient-reported measures of intervention mastery and perceived effectiveness to guide treatment decisions, and objective assessment used improvement of MGs at 12 weeks. The moderate agreement (κ=0.42) between subjective response assessment and objective cognitive improvement validated our classification approach, demonstrating that participants showing both subjective benefit and objective gains could continue original protocols while nonresponders required treatment modification.

    Dynamic treatment regimen analysis revealed that adaptive strategies consistently outperformed static approaches, with treatment intensification proving more effective than modality switching for nonresponders in both intervention groups. This approach represents a potential shift from an experience-based approach toward precise management, providing clinicians with evidence-based decision rules that may improve treatment efficiency and optimize resource allocation in community-based MCI prevention programs.

    Further research is needed to advance this field. First, investigation of motivation and adherence factors is essential to identify predictors of intervention engagement and develop evidence-based strategies for enhancing long-term adherence to VR-based cognitive training, particularly examining how individual characteristics and intervention design features influence sustained participation and treatment response. Second, optimization of SMART design parameters is warranted, including validation of alternative response assessment time points and development of objective response criteria that integrate cognitive, physiological, and behavioral indicators to reduce reliance on subjective measures. Third, multicenter trials in diverse populations and health care settings are needed to establish the generalizability and implementation feasibility of VR-enhanced adaptive interventions, particularly examining effectiveness across different MCI subtypes and socioeconomic contexts.

    Innovations and Clinical Implications

    This study advances MCI prevention research through several methodological and practical innovations. First, we developed the first SMART-based adaptive intervention framework for cognitive health, moving beyond fixed-protocol designs that dominate current practice. The 12-week objective response assessment criteria—integrating subjective mastery evaluation with cognitive performance metrics—provide clinicians with standardized decision rules for treatment modification, replacing experience-dependent judgments that vary across providers and settings. Second, our finding that treatment intensification outperforms modality switching for nonresponders establishes a specific clinical pathway: rather than abandoning ineffective interventions, clinicians should increase intervention dose before switching modalities. Third, the identification of diabetes as a positive predictor and hypertension as a negative predictor of treatment response enables risk stratification and personalized intervention selection prior to treatment initiation.

    The clinical utility of these findings extends beyond research settings. Community health workers can implement the standardized decision rules without specialized neuropsychological training, as the response assessment requires only basic cognitive screening tools already deployed in Chinese primary care. The VR-Taichi protocol leverages culturally familiar mind-body exercises, addressing adherence barriers that limit Western-developed interventions in Asian populations. Our subgroup analyses identify priority populations for targeted screening: adults aged 60-71 years, individuals with MoCA scores indicating lower baseline function, and patients with metabolic comorbidities. These groups demonstrated 15%-25% greater cognitive improvement compared with their counterparts, suggesting that resource-limited settings should prioritize these populations for intensive intervention.

    This precision medicine approach addresses a critical gap in current dementia prevention guidelines, which provide broad recommendations without specifying how to tailor interventions to individual characteristics or when to modify treatment based on early response. Our findings support policy development for technology-integrated cognitive health programs within urban community health systems, with potential adaptation to other metropolitan areas possessing similar digital infrastructure and hierarchical medical delivery systems.

    Implementation Feasibility and Public Health Applications

    Our findings support the feasibility of implementing standardized VR-based protocols within existing community health service centers, leveraging Shanghai’s robust digital infrastructure and established hierarchical medical system. The demonstrated effectiveness of adaptive treatment strategies, particularly the superior performance of VRTC-based interventions across diverse participant characteristics, suggests that technology-enhanced approaches can be successfully integrated into routine community-based MCI prevention programs. The identification of diabetes as a positive predictor and hypertension as a negative predictor of treatment response provides practical guidance for community health workers in optimizing intervention selection and resource allocation.

    The scalability of our intervention model is supported by several practical considerations specific to urban community settings. First, the Taichi protocol used in VRTC is culturally familiar to Chinese older populations, facilitating acceptance and adherence. Second, the objective response assessment criteria developed through our SMART design provide clear decision rules that can be implemented by trained community health personnel without requiring specialized clinical expertise. Third, the significant effect sizes observed suggest that even with some implementation variability expected in real-world settings, meaningful cognitive benefits are likely to be maintained. These findings support the development of evidence-based public service recommendations for integrating VR technology into Shanghai’s community health infrastructure, with potential for adaptation to other metropolitan areas with similar health care delivery systems and technological capabilities.

    Limitations

    Our study has several limitations that should be considered. The sample was predominantly recruited from urban communities in Shanghai, which may limit generalizability to rural populations or different health care settings. Additionally, while our follow-up period was adequate for demonstrating intervention effects, longer-term outcomes remain to be evaluated. Subjective evaluation of intervention content mastery as a criterion for assessing participants’ responsiveness is not comprehensive, especially for aerobic exercises such as Taichi. Although robustness analysis showed no significant difference between the 2 response definitions, we still recommended that future evaluations incorporate physiological, cognitive, psychological, and behavioral indicators to fully capture participants’ responses to the intervention. Assessment based on VO₂ max, Geriatric Depression Scale (GDS), and quality-of-life measures is suggested as a robust approach for evaluating the effectiveness of nonsubjective interventions.

    Conclusion

    This study represents the SMART design to develop evidence-based adaptive intervention strategies for MCI prevention in community settings, demonstrating that technology-enhanced dual-task training with VR significantly outperforms traditional exercise modalities and control conditions. The CT+VRTC intervention achieved substantial cognitive improvements (Cohen d=1.425, 95% CI 0.785-2.060; P<.001) that exceeded those reported in previous multidomain trials, with adaptive treatment strategies consistently outperforming static approaches across diverse participant characteristics. Our predictive modeling revealed that individuals with diabetes showed enhanced treatment responsiveness across all outcome measures, while those with hypertension demonstrated reduced response, highlighting the critical importance of comorbidity-informed treatment selection. The systematic approach to treatment adaptation provides clinicians with evidence-based decision rules for optimizing intervention delivery in real-world settings. These findings establish a foundation for precision-based cognitive health management, potentially bridging the gap between traditional multidomain approaches and personalized medicine strategies while offering a scalable, cost-effective solution for community-based MCI prevention programs.

    These findings establish a foundation for precision-based cognitive health management, potentially bridging the gap between traditional multidomain approaches and personalized medicine strategies while offering a scalable, cost-effective solution for community-based MCI prevention programs. The demonstrated large effect sizes and adaptive treatment protocols provide evidence-based recommendations for implementing VR-enhanced cognitive interventions within Shanghai’s existing community health service infrastructure, supporting the development of technology-integrated public health strategies for urban older populations.

    This study is supported by the Yunnan Key Research Program (grant 202402AD080004). This study is funded by The Shanghai Public Health System Construction 3‐Year Action Plan (grant no. GWVI-11.1-48). The funder had no involvement in the study design, data collection, analysis, interpretation, or the writing of the manuscript.

    The datasets generated or analyzed during this study are available from the corresponding author on reasonable request.

    All authors contributed to the article and approved the submitted version. After unmasking of the outcome data, all authors had full access to all the data in the study and had final responsibility for the decision to submit for publication. DB and CY verified the raw data in the study. GL, CS, and DB conceptualized the study. CS proposed the experimental design and algorithm framework. CS and CY implemented the model design and evaluation. CY was responsible for using the data analysis software in the study. All authors validated the findings of the study. All authors contributed to the formal analysis of the data. DB and CY conducted the investigation. DB, CS, and GL provided the resources necessary for the study. DB and XH curated the data. CY and DB wrote the original draft of the manuscript. All authors reviewed and edited the manuscript. CY created the visualizations of the study findings. CS and GL supervised the research activities. GL managed the project administration. GL contributed to funding acquisition. CS (shihchenshu@hotmail.com) and GL contributed equally as co-corresponding authors.

    None declared.

    Edited by A Mavragani, S Brini; submitted 18.Jun.2025; peer-reviewed by Z Zhang, LF Belo, A Hu; comments to author 14.Sep.2025; accepted 17.Oct.2025; published 25.Nov.2025.

    ©Chenhao Yu, Dongsheng Bian, Jiatao Zhang, Xiao Han, Chenshu Shi, Guohong Li. Originally published in the Journal of Medical Internet Research (https://www.jmir.org), 25.Nov.2025.

    This is an open-access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work, first published in the Journal of Medical Internet Research (ISSN 1438-8871), is properly cited. The complete bibliographic information, a link to the original publication on https://www.jmir.org/, as well as this copyright and license information must be included.

    Continue Reading

  • TRADING DAY Fed optimism, Thanksgiving week – Reuters

    1. TRADING DAY Fed optimism, Thanksgiving week  Reuters
    2. Wall Street braces for a choppy holiday week amid AI fears  qz.com
    3. Wall Street heads into Thanksgiving week with questions over AI trade, Fed rate cut  CNBC
    4. Three Things to Know & Watch – Week of 11/24/2025  AdvisorHub
    5. History Says Thanksgiving Gains Could Be in Store for Bulls  Schaeffer’s Investment Research

    Continue Reading

  • IMFINZI® approved in the US as first and only perioperative immunotherapy for patients with early gastric and gastroesophageal cancers

    AstraZeneca’s IMFINZI® (durvalumab) in combination with standard-of-care FLOT chemotherapy (fluorouracil, leucovorin, oxaliplatin, and docetaxel) has been approved in the US for the treatment of adult patients with resectable, early-stage and locally advanced (Stages II, III, IVA) gastric and gastroesophageal junction (GEJ) cancers. The approved regimen includes neoadjuvant IMFINZI in combination with chemotherapy before surgery, followed by adjuvant IMFINZI in combination with chemotherapy, then IMFINZI monotherapy.

    The approval follows Priority Review by the Food and Drug Administration (FDA) and is based on event-free survival (EFS) and overall survival (OS) data from the MATTERHORN Phase III trial. The EFS results were presented during the Plenary Session at the 2025 American Society of Clinical Oncology (ASCO) Annual Meeting and simultaneously published in The New England Journal of Medicine. OS results from MATTERHORN were presented in a Proffered Paper session at the European Society for Medical Oncology (ESMO) Congress 2025.

    Gastric cancer is the fifth leading cause of cancer death globally, with nearly one million people diagnosed each year.1 In 2024, there were roughly 6,500 drug-treated patients in the US in early-stage and locally advanced gastric or GEJ cancer.2

    Dave Fredrickson, Executive Vice President, Oncology Haematology Business Unit, AstraZeneca, said: “This approval ushers in a new clinical paradigm for patients with early gastric and gastroesophageal junction cancers, with IMFINZI plus FLOT delivering a durable survival benefit that increases over time. As the third US approval for a perioperative IMFINZI-based regimen, this milestone further validates the perioperative approach and underscores our focus on bringing novel treatments to early-stage cancers where cure is the goal.”

    Yelena Y. Janjigian, MD, Chief Attending Physician of the Gastrointestinal Medical Oncology Service, Memorial Sloan Kettering Cancer Center (MSK), New York and principal investigator in the MATTERHORN trial, said: “Today’s approval marks the first immunotherapy regimen approved in the neoadjuvant setting for gastric and gastroesophageal junction cancers—with durvalumab demonstrating a clear overall survival benefit and opening an entirely new chapter in the treatment of early-stage disease. Nearly seven in 10 patients were alive at three years following treatment with the durvalumab-based perioperative regimen. This survival benefit, observed regardless of PD-L1 status, establishes a new standard of care in this curative-intent setting.”

    Aki Smith, Founder and Executive Director, Hope for Stomach Cancer, said: “From personal experience as a caregiver to my father, I know that for too long patients diagnosed with early gastric or gastroesophageal junction cancer have faced a high risk of their cancer returning, even after undergoing surgery and therapy intended to cure it. Today’s approval represents a major step forward in improving outcomes and offering renewed hope to those affected by this devastating disease.”

    In a planned interim analysis, patients treated with the IMFINZI-based perioperative regimen showed a 29% reduction in the risk of disease progression, recurrence or death versus chemotherapy alone (based on an EFS hazard ratio [HR] of 0.71; 95% confidence interval [CI] 0.58-0.86; P<0.001). Estimated median EFS was not yet reached for the IMFINZI arm versus 32.8 months for the comparator arm. An estimated 78.2% of patients treated with the IMFINZI-based perioperative regimen were event-free at one year, compared to 74.0% in the comparator arm; the estimated 24-month EFS rate was 67.4% versus 58.5%, respectively.

    In the final OS analysis, results showed the IMFINZI and FLOT perioperative regimen reduced the risk of death by 22% compared with chemotherapy alone (based on a HR of 0.78; 95% CI 0.63-0.96; P=0.021). An estimated 69% of patients treated with the IMFINZI-based regimen were alive at three years compared with 62% in the FLOT-only arm. With longer follow-up, the OS curves showed continued separation, signaling a greater magnitude of benefit over time for the IMFINZI-based regimen. An OS benefit was observed regardless of PD-L1 status.

    The safety profile for IMFINZI and FLOT chemotherapy was consistent with the known profiles of each medicine, and the percentage of patients that completed surgery was similar compared to chemotherapy alone. Grade 3 or higher adverse events due to any cause were similar between the two arms (71.6% for IMFINZI and FLOT arm; 71.2% for FLOT-only arm).

    The US regulatory submission was reviewed under Project Orbis, which provides a framework for concurrent submission and review of oncology medicines among participating international partners. As part of Project Orbis, the IMFINZI and FLOT perioperative regimen is also under review by regulatory authorities in Australia, Canada, and Switzerland for the same indication. Regulatory applications are also under review in the European Union (EU), Japan and several other countries.

    IMPORTANT SAFETY INFORMATION

    There are no contraindications for IMFINZI® (durvalumab).

    Immune-Mediated Adverse Reactions

    Important immune-mediated adverse reactions listed under Warnings and Precautions may not include all possible severe and fatal immune-mediated reactions. Immune-mediated adverse reactions, which may be severe or fatal, can occur in any organ system or tissue. Immune-mediated adverse reactions can occur at any time after starting treatment or after discontinuation. Monitor patients closely for symptoms and signs that may be clinical manifestations of underlying immune-mediated adverse reactions. Evaluate liver enzymes, creatinine, and thyroid function at baseline and periodically during treatment. In cases of suspected immune-mediated adverse reactions, initiate appropriate workup to exclude alternative etiologies, including infection. Institute medical management promptly, including specialty consultation as appropriate. Withhold or permanently discontinue IMFINZI depending on severity. See USPI Dosing and Administration for specific details. In general, if IMFINZI requires interruption or discontinuation, administer systemic corticosteroid therapy (1 mg to 2 mg/kg/day prednisone or equivalent) until improvement to Grade 1 or less. Upon improvement to Grade 1 or less, initiate corticosteroid taper and continue to taper over at least 1 month. Consider administration of other systemic immunosuppressants in patients whose immune-mediated adverse reactions are not controlled with corticosteroid therapy.

    Immune-Mediated Pneumonitis

    IMFINZI can cause immune-mediated pneumonitis. The incidence of pneumonitis is higher in patients who have received prior thoracic radiation. In patients who did not receive recent prior radiation, the incidence of immune-mediated pneumonitis was 2.4% (34/1414), including fatal (<0.1%), and Grade 3-4 (0.4%) adverse reactions. The frequency and severity of immune-mediated pneumonitis in patients who did not receive definitive chemoradiation prior to IMFINZI were similar in patients who received IMFINZI as a single agent or with ES-SCLC or BTC when given in combination with chemotherapy.

    Immune-Mediated Colitis

    IMFINZI can cause immune-mediated colitis that is frequently associated with diarrhea. Cytomegalovirus (CMV) infection/reactivation has been reported in patients with corticosteroid-refractory immune-mediated colitis. In cases of corticosteroid-refractory colitis, consider repeating infectious workup to exclude alternative etiologies. Immune-mediated colitis occurred in 2% (37/1889) of patients receiving IMFINZI, including Grade 4 (<0.1%) and Grade 3 (0.4%) adverse reactions.

    Immune-Mediated Hepatitis

    IMFINZI can cause immune-mediated hepatitis. Immune-mediated hepatitis occurred in 2.8% (52/1889) of patients receiving IMFINZI, including fatal (0.2%), Grade 4 (0.3%) and Grade 3 (1.4%) adverse reactions.

    Immune-Mediated Endocrinopathies

    • Adrenal Insufficiency: IMFINZI can cause primary or secondary adrenal insufficiency. For Grade 2 or higher adrenal insufficiency, initiate symptomatic treatment, including hormone replacement as clinically indicated. Immune-mediated adrenal insufficiency occurred in 0.5% (9/1889) of patients receiving IMFINZI, including Grade 3 (<0.1%) adverse reactions.
    • Hypophysitis: IMFINZI can cause immune-mediated hypophysitis. Hypophysitis can present with acute symptoms associated with mass effect such as headache, photophobia, or visual field cuts. Hypophysitis can cause hypopituitarism. Initiate symptomatic treatment including hormone replacement as clinically indicated. Grade 3 hypophysitis/hypopituitarism occurred in <0.1% (1/1889) of patients who received IMFINZI.
    • Thyroid Disorders (Thyroiditis, Hyperthyroidism, and Hypothyroidism): IMFINZI can cause immune-mediated thyroid disorders. Thyroiditis can present with or without endocrinopathy. Hypothyroidism can follow hyperthyroidism. Initiate hormone replacement therapy for hypothyroidism or institute medical management of hyperthyroidism as clinically indicated.
      • Thyroiditis: Immune-mediated thyroiditis occurred in 0.5% (9/1889) of patients receiving IMFINZI, including Grade 3 (<0.1%) adverse reactions.
      • Hyperthyroidism: Immune-mediated hyperthyroidism occurred in 2.1% (39/1889) of patients receiving IMFINZI.
      • Hypothyroidism: Immune-mediated hypothyroidism occurred in 8.3% (156/1889) of patients receiving IMFINZI, including Grade 3 (<0.1%) adverse reactions.
    • Type 1 Diabetes Mellitus, which can present with diabetic ketoacidosis: Monitor patients for hyperglycemia or other signs and symptoms of diabetes. Initiate treatment with insulin as clinically indicated. Grade 3 immune-mediated Type 1 diabetes mellitus occurred in <0.1% (1/1889) of patients receiving IMFINZI.

    Immune-Mediated Nephritis with Renal Dysfunction

    IMFINZI can cause immune-mediated nephritis. Immune-mediated nephritis occurred in 0.5% (10/1889) of patients receiving IMFINZI, including Grade 3 (<0.1%) adverse reactions.

    Immune-Mediated Dermatology Reactions

    IMFINZI can cause immune-mediated rash or dermatitis. Exfoliative dermatitis, including Stevens-Johnson Syndrome (SJS), drug rash with eosinophilia and systemic symptoms (DRESS), and toxic epidermal necrolysis (TEN), has occurred with PD-1/L-1 and CTLA-4 blocking antibodies. Topical emollients and/or topical corticosteroids may be adequate to treat mild to moderate non-exfoliative rashes. Immune-mediated rash or dermatitis occurred in 1.8% (34/1889) of patients receiving IMFINZI, including Grade 3 (0.4%) adverse reactions.  

    Other Immune-Mediated Adverse Reactions

    The following clinically significant, immune-mediated adverse reactions occurred at an incidence of less than 1% each in patients who received IMFINZI or were reported with the use of other immune-checkpoint inhibitors.

    • Cardiac/vascular: Myocarditis, pericarditis, vasculitis.
    • Nervous system: Meningitis, encephalitis, myelitis and demyelination, myasthenic syndrome/myasthenia gravis (including exacerbation), Guillain-Barré syndrome, nerve paresis, autoimmune neuropathy.
    • Ocular: Uveitis, iritis, and other ocular inflammatory toxicities can occur. Some cases can be associated with retinal detachment. Various grades of visual impairment to include blindness can occur. If uveitis occurs in combination with other immune-mediated adverse reactions, consider a Vogt-Koyanagi-Harada-like syndrome, as this may require treatment with systemic steroids to reduce the risk of permanent vision loss.
    • Gastrointestinal: Pancreatitis including increases in serum amylase and lipase levels, gastritis, duodenitis.
    • Musculoskeletal and connective tissue disorders: Myositis/polymyositis, rhabdomyolysis and associated sequelae including renal failure, arthritis, polymyalgia rheumatic.
    • Endocrine: Hypoparathyroidism.
    • Other (hematologic/immune): Hemolytic anemia, aplastic anemia, hemophagocytic lymphohistiocytosis, systemic inflammatory response syndrome, histiocytic necrotizing lymphadenitis (Kikuchi lymphadenitis), sarcoidosis, immune thrombocytopenia, solid organ transplant rejection, other transplant (including corneal graft) rejection.

    Infusion-Related Reactions

    IMFINZI can cause severe or life-threatening infusion-related reactions. Monitor for signs and symptoms of infusion-related reactions. Interrupt, slow the rate of, or permanently discontinue IMFINZI based on the severity. See USPI Dosing and Administration for specific details. For Grade 1 or 2 infusion-related reactions, consider using pre-medications with subsequent doses. Infusion-related reactions occurred in 2.2% (42/1889) of patients receiving IMFINZI, including Grade 3 (0.3%) adverse reactions.

    Complications of Allogeneic HSCT after IMFINZI

    Fatal and other serious complications can occur in patients who receive allogeneic hematopoietic stem cell transplantation (HSCT) before or after being treated with a PD-1/L-1 blocking antibody. Transplant-related complications include hyperacute graft-versus-host disease (GVHD), acute GVHD, chronic GVHD, hepatic veno-occlusive disease (VOD) after reduced intensity conditioning, and steroid-requiring febrile syndrome (without an identified infectious cause). These complications may occur despite intervening therapy between PD-1/L-1 blockade and allogeneic HSCT. Follow patients closely for evidence of transplant-related complications and intervene promptly. Consider the benefit versus risks of treatment with a PD-1/L-1 blocking antibody prior to or after an allogeneic HSCT.

    Embryo-Fetal Toxicity

    Based on its mechanism of action and data from animal studies, IMFINZI can cause fetal harm when administered to a pregnant woman. Advise pregnant women of the potential risk to a fetus. In females of reproductive potential, verify pregnancy status prior to initiating IMFINZI and advise them to use effective contraception during treatment with IMFINZI and for 3 months after the last dose of IMFINZI.

    Lactation

    There is no information regarding the presence of IMFINZI in human milk; however, because of the potential for serious adverse reactions in breastfed infants from IMFINZI, advise women not to breastfeed during treatment and for 3 months after the last dose.

    Adverse Reactions

    • In patients with resectable GC/GEJC, the most common adverse reactions in the overall study (occurring in ≥20% of patients) were diarrhea, nausea, peripheral neuropathy, fatigue, alopecia, decreased appetite, rash, abdominal pain, vomiting, musculoskeletal pain, pyrexia, and stomatitis.
    • In patients with resectable GC/GEJC in the neoadjuvant phase of the MATTERHORN study receiving IMFINZI in combination with FLOT chemotherapy (n=475), permanent discontinuation of IMFINZI due to an adverse reaction occurred in 2.5% of patients. Serious adverse reactions occurred in 21% of patients; the most frequent (≥2%) serious adverse reaction was diarrhea (2.5%). Deaths occurred in 1.9% of patients; deaths ≥2 patients included septic shock (0.6%) and acute coronary syndrome (0.4%). Of the 475 patients in the IMFINZI + FLOT chemotherapy treatment arm and 469 patients in the placebo + FLOT chemotherapy treatment arm who received neoadjuvant treatment, 0.6% and 0.4% of patients, respectively, did not receive surgery due to adverse reactions, and 2.3% and 2.6% of patients, respectively, had a delay in surgery due to ARs.
    • In patients with resectable GC/GEJC in the adjuvant phase of the MATTERHORN study receiving IMFINZI in combination with FLOT chemotherapy (n=365), permanent discontinuation of IMFINZI due to an adverse reaction occurred in 7% of patients. Serious adverse reactions occurred in 29% of patients; the most frequent (≥2%) serious adverse reaction was pneumonia (2.5%). Deaths occurred in 2.2% of patients; deaths ≥2 patients included gastrointestinal perforation (0.5%) and COVID-19 (0.5%).
    • In patients with resectable GC/GEJC in the adjuvant phase of the MATTERHORN study receiving IMFINZI alone (n=345), permanent discontinuation of IMFINZI due to an adverse reaction occurred in 6% of patients. Serious adverse reactions occurred in 14% of patients. Deaths occurred in 1.7% of patients; deaths ≥2 patients included gastrointestinal perforation (0.6%) and COVID-19 (0.6%).

    The safety and effectiveness of IMFINZI has not been established in pediatric patients.

    Indication:

    IMFINZI in combination with fluorouracil, leucovorin, oxaliplatin and docetaxel (FLOT) as neoadjuvant and adjuvant treatment, followed by single agent IMFINZI, is indicated for the treatment of adult patients with resectable gastric or gastroesophageal junction adenocarcinoma (GC/GEJC).

    Please see additional Important Safety Information throughout and Full Prescribing Information including Medication Guide for IMFINZI.

    You may report side effects related to AstraZeneca products.

    Notes

    Gastric and gastroesophageal junction cancers

    Gastric (stomach) cancer is the fifth most common cancer worldwide and the fifth-highest leading cause of cancer mortality.1 Nearly one million new patients were diagnosed with gastric cancer in 2022, with approximately 660,000 deaths reported globally.1 In many regions, its incidence has been increasing in patients younger than 50 years old, along with other gastrointestinal (GI) malignancies.3 In 2024, there were roughly 43,000 drug-treated patients in the US, EU and Japan in early-stage and locally advanced gastric or GEJ cancer.2 Approximately 62,000 patients in these regions are expected to be newly diagnosed in this setting by 2030.4

    GEJ cancer is a type of gastric cancer that arises from and spans the area where the esophagus connects to the stomach.5

    Disease recurrence is common in patients with resectable gastric cancer despite undergoing surgery with curative intent and treatment with neoadjuvant/adjuvant chemotherapy.6 Approximately one in four patients with gastric cancer who undergo surgery develop recurrent disease within one year, and the five-year survival rate remains poor, with less than half of patients alive at five years.6-7

    MATTERHORN

    MATTERHORN is a randomized, double-blind, placebo-controlled, multi-center, global Phase III trial evaluating IMFINZI as perioperative treatment for patients with resectable Stage II-IVA gastric and GEJ cancers. Perioperative therapy includes treatment before and after surgery, also known as neoadjuvant/adjuvant therapy. In the trial, 948 patients were randomized to receive a 1500mg fixed dose of IMFINZI plus FLOT chemotherapy or placebo plus FLOT chemotherapy every four weeks for two cycles prior to surgery. This was followed by IMFINZI or placebo every four weeks for up to 12 cycles after surgery (including two cycles of IMFINZI or placebo plus FLOT chemotherapy and 10 additional cycles of IMFINZI or placebo monotherapy).

    In the MATTERHORN trial, the primary endpoint is EFS, defined as time from randomization until the date of one of the following events (whichever occurred first): RECIST (version 1.1, per blinded independent central review assessment) progression that precludes surgery or requires non-protocol therapy during the neoadjuvant period; RECIST progression/recurrence during the adjuvant period; non-RECIST progression that precludes surgery or requires non-protocol therapy during the neoadjuvant period or discovered during surgery; progression/recurrence confirmed by biopsy post-surgery; or death due to any cause. Key secondary endpoints include pathologic complete response rate, defined as the proportion of patients who have no detectable cancer cells in resected tumor tissue following neoadjuvant therapy, and OS. The trial enrolled participants in 176 centers in 20 countries, including in the US, Canada, Europe, South America and Asia.

    IMFINZI

    IMFINZI® (durvalumab) is a human monoclonal antibody that binds to the PD-L1 protein and blocks the interaction of PD-L1 with the PD-1 and CD80 proteins, countering the tumor’s immune-evading tactics and releasing the inhibition of immune responses.

    In GI cancer, IMFINZI is approved in combination with chemotherapy in locally advanced or metastatic biliary tract cancer (BTC) and in combination with tremelimumab-actl in unresectable hepatocellular carcinoma (HCC). IMFINZI is also approved as a monotherapy in unresectable HCC in Japan and the EU.

    In addition to its indications in GI cancers, IMFINZI is the global standard of care based on OS in the curative-intent setting of unresectable, Stage III non-small cell lung cancer (NSCLC) in patients whose disease has not progressed after chemoradiotherapy (CRT). Additionally, IMFINZI is approved as a perioperative treatment in combination with neoadjuvant chemotherapy in resectable NSCLC, and in combination with a short course of tremelimumab-actl and chemotherapy for the treatment of metastatic NSCLC. IMFINZI is also approved for limited-stage small cell lung cancer (SCLC) in patients whose disease has not progressed following concurrent platinum-based CRT; and in combination with chemotherapy for the treatment of extensive-stage SCLC.

    Perioperative IMFINZI in combination with neoadjuvant chemotherapy is approved in the US, EU, Japan and other countries for patients with muscle-invasive bladder cancer based on results from the NIAGARA Phase III trial. Additionally, in May 2025, IMFINZI added to Bacillus Calmette-Guérin induction and maintenance therapy met the primary endpoint of disease-free survival for patients with high-risk non-muscle-invasive bladder cancer in the POTOMAC Phase III trial.

    IMFINZI in combination with chemotherapy followed by IMFINZI monotherapy is approved as a 1st-line treatment for primary advanced or recurrent endometrial cancer (mismatch repair deficient disease only in the US and EU). IMFINZI in combination with chemotherapy followed by olaparib and IMFINZI is approved for patients with mismatch repair proficient advanced or recurrent endometrial cancer in the EU and Japan.

    Since the first approval in May 2017, more than 414,000 patients have been treated with IMFINZI. As part of a broad development program, IMFINZI is being tested as a single treatment and in combinations with other anti-cancer treatments for patients with NSCLC, bladder cancer, breast cancer, ovarian cancer and several GI cancers.

    AstraZeneca in GI cancers

    AstraZeneca has a broad development program for the treatment of GI cancers across several medicines and a variety of tumor types and stages of disease. In 2022, GI cancers collectively represented approximately 5 million new cancer cases leading to approximately 3.3 million deaths.8

    Within this program, the Company is committed to improving outcomes in gastric, liver, biliary tract, esophageal, pancreatic, and colorectal cancers.

    In addition to its indications in BTC and HCC, IMFINZI is being assessed in combinations, including with tremelimumab-actl, in liver, esophageal and gastric cancers in an extensive development program spanning early to late-stage disease across settings.

    Fam-trastuzumab deruxtecan-nxki, a HER2-directed antibody drug conjugate (ADC), is approved in the US and several other countries for HER2-positive advanced gastric cancer. Fam-trastuzumab deruxtecan-nxki is jointly developed and commercialized by AstraZeneca and Daiichi Sankyo.

    Olaparib, a first-in-class PARP inhibitor, is approved in the US and several other countries for the treatment of BRCA-mutated metastatic pancreatic cancer. Olaparib is developed and commercialized in collaboration with Merck & Co., Inc (MSD outside the US and Canada).

    The Company is also assessing rilvegostomig (AZD2936), a PD-1/TIGIT bispecific antibody, in combination with chemotherapy as an adjuvant therapy in BTC, in combination with bevacizumab with or without tremelimumab-actl as a 1st-line treatment in patients with advanced HCC, and as a 1st-line treatment in patients with HER2-negative, locally advanced unresectable or metastatic gastric and GEJ cancers. Rilvegostomig is also being evaluated in combination with Fam-trastuzumab deruxtecan-nxki in previously untreated, HER2-expressing, locally advanced or metastatic BTC.

    AstraZeneca is advancing multiple modalities that provide complementary mechanisms for targeting Claudin 18.2, a promising therapeutic target in gastric cancer. These include sonesitatug vedotin, a potential first-in-class ADC licensed from KYM Biosciences Inc., currently in Phase III development; AZD5863, a novel Claudin 18.2/CD3 T-cell engager bispecific antibody licensed from Harbour Biomed in Phase I development; and AZD4360, an antibody drug conjugate, currently being evaluated in a Phase I/II trial in patients with advanced solid tumors.

    In early development, AstraZeneca is developing C-CAR031 / AZD7003, a Glypican 3 (GPC3) armored CAR T, in HCC. C-CAR031 / AZD7003is being co-developed with AbelZeta in China where it is under evaluation in an IIT.

    AstraZeneca in immuno-oncology (IO)

    AstraZeneca is a pioneer in introducing the concept of immunotherapy into dedicated clinical areas of high unmet medical need. The Company has a comprehensive and diverse IO portfolio and pipeline anchored in immunotherapies designed to overcome evasion of the anti-tumor immune response and stimulate the body’s immune system to attack tumors.

    AstraZeneca strives to redefine cancer care and help transform outcomes for patients with IMFINZI as a monotherapy and in combination with tremelimumab-actl as well as other novel immunotherapies and modalities. The Company is also investigating next-generation immunotherapies like bispecific antibodies and therapeutics that harness different aspects of immunity to target cancer, including cell therapy and T-cell engagers.

    AstraZeneca is pursuing an innovative clinical strategy to bring IO-based therapies that deliver long-term survival to new settings across a wide range of cancer types. The Company is focused on exploring novel combination approaches to help prevent treatment resistance and drive longer immune responses. With an extensive clinical program, the Company also champions the use of IO treatment in earlier disease stages, where there is the greatest potential for cure.

    AstraZeneca in oncology

    AstraZeneca is leading a revolution in oncology with the ambition to provide cures for cancer in every form, following the science to understand cancer and all its complexities to discover, develop and deliver life-changing medicines to patients.

    The Company’s focus is on some of the most challenging cancers. It is through persistent innovation that AstraZeneca has built one of the most diverse portfolios and pipelines in the industry, with the potential to catalyze changes in the practice of medicine and transform the patient experience.

    AstraZeneca has the vision to redefine cancer care and, one day, eliminate cancer as a cause of death.

    AstraZeneca

    AstraZeneca is a global, science-led biopharmaceutical company that focuses on the discovery, development, and commercialization of prescription medicines in Oncology, Rare Diseases, and BioPharmaceuticals, including Cardiovascular, Renal & Metabolism, and Respiratory & Immunology. Based in Cambridge, UK, AstraZeneca’s innovative medicines are sold in more than 125 countries and used by millions of patients worldwide. Please visit www.astrazeneca-us.com and follow the Company on social media @AstraZeneca.

    Media Inquiries

    Fiona Cookson

    Lauren-Jei McCarthy

     

    +1 212 814 3923

    +1 347 918 7001 

     

     

    US Media Mailbox: usmediateam@astrazeneca.com           

    References

    1. World Health Organization. International Agency for Research on Cancer. Stomach fact sheet. Available at: https://gco.iarc.who.int/media/globocan/factsheets/cancers/7-stomach-fact-sheet.pdf. Accessed November 2025.

    2. AstraZeneca PLC. Investor relations epidemiology spreadsheet. Available at: https://www.astrazeneca.com/investor-relations.html. Accessed November 2025.

    3. Li Y, et al. Global burden of young-onset gastric cancer: a systematic trend analysis of the global burden of disease study 2019. Gastric Cancer. 2024;27(4):684-700.

    4. Kantar Health, validated with SEER stage at diagnosis and Cabasag et al. And Kuzuu et al. 2021.

    5. National Cancer Institute. Gastroesophageal junction. Available at: https://www.cancer.gov/publications/dictionaries/cancer-terms/def/gastroesophageal-junction. Accessed November 2025.

    6. Li Y, et al. Postoperative recurrence of gastric cancer depends on whether the chemotherapy cycle was more than 9 cycles. Medicine. 2022;101(5):e28620.

    7. Ilic M, Ilic I. Epidemiology of stomach cancer. World J Gastroenterol. 2022;28(12):1187-1203.

    8. World Health Organization. World cancer fact sheet. Available at: https://gco.iarc.who.int/media/globocan/factsheets/populations/900-world-fact-sheet.pdf. Accessed November 2025.

    MSK Disclosure: Dr. Janjigian provides consulting and advisory services to AstraZeneca.

    Continue Reading

  • Journal of Medical Internet Research

    Journal of Medical Internet Research

    Idiopathic normal pressure hydrocephalus (iNPH) is a neurological disease characterized by cognitive impairment, gait disturbance, and urinary incontinence [], with ventricular enlargement and the DESH sign (disproportionately enlarged subarachnoid hydrocephalus) shown on brain scans []. It is a common cause of dementia in older people [] and can be treated effectively with cerebrospinal fluid (CSF) shunts [,].

    However, the clinical manifestations of iNPH lack specificity, with only 50% of patients exhibiting the full triad []. Therefore, patients with iNPH are usually misdiagnosed as other neurodegenerative diseases, such as Alzheimer disease or Parkinson disease []. Unlike these conditions, the clinical symptoms of iNPH can be reversed through CSF shunting []. However, only 39% to 81% of patients with shunts show improvement 3 to 6 months after the shunt insertion []. The CSF drainage test is an important tool for evaluating patients with possible iNPH before shunt surgery [,,]. The tap test and continuous external lumbar drainage (ELD) are 2 commonly used methods []. Previous studies have shown that the tap test has a sensitivity of only 26%, whereas the continuous ELD has a sensitivity of 50% and a specificity of 80% [,]. As a result, some medical centers conduct continuous ELD directly to avoid repeated lumbar punctures and reduce the risk of misdiagnosis [-]. On the other hand, current evaluation methods for CSF drainage tests also have relatively low sensitivities. Previous studies using the Mini-Mental State Examination (MMSE), 10-m walking test (TMWT), and timed up and go (TUG) test indicate a sensitivity of 26% for tap test [,]. This implies that current assessment methods are inadequate in providing reliable guidance for clinical practice because traditional cognitive tests can cause large learning effects because of the short time intervals between tests [] and video-based gait analysis may be influenced by the subjective opinions of raters [] and lacks quantitative measures, such as stride length, step height, and gait velocity [,]. These shortcomings of traditional tests highlight the need for a more objective and quantitative evaluation method.

    In recent years, digital neuropsychological evaluation equipment has emerged, which was designed to overcome the major limitations of paper and pencil tests, such as low sensitivity, subjectivity, and test-retest reliability []. They also provide randomized test paradigms and automated recording of variables such as reaction time, thereby improving evaluation efficacy []. A recent study indicated that computerized neuropsychological tests could detect cognitive impairment and postshunt improvements in patients with iNPH []. Additionally, 3-dimensional gait analysis aids in the diagnosis of iNPH by offering multidimensional gait parameters [,]. There is growing evidence demonstrating that motor and cognitive impairment in iNPH may result from interconnected neural network impairments [,], and recent research based on traditional cognitive and gait tests found that the improvement of different symptoms after ELD may follow different temporal trajectories []. Therefore, combining cognitive and gait assessments may provide a more comprehensive and complementary picture of functional response after ELD. Within the expanding field of digital medicine, the integration of digital cognitive and gait tests into diagnostic workflow has been widely investigated and implemented in other cognitive disorders [,]. However, an evidence gap remains regarding the predictive value of these digital tests in the context of ELD in patients with iNPH.

    Therefore, this study aimed to (1) evaluate the improvement of cognitive and gait parameters after ELD through the application of digital tests and (2) investigate the predictive value of digital cognitive and motor assessments for shunt outcomes in patients with iNPH.

    Study Design

    Patients with iNPH were enrolled from an ongoing prospective cohort study at West China Hospital of Sichuan University between May 2022 and November 2023. The inclusion criteria included (1) at least one symptom of cognitive decline, gait disturbance, and urinary incontinence; (2) ventricular enlargement (Evans index >0.3), focally dilated sulci, or the DESH sign shown on brain magnetic resonance imaging; (3) CSF opening pressure ≤200 mm H2O; and (4) informed consent. We excluded patients with (1) gait disturbance, cognitive impairment, or urinary incontinence due to other neurological diseases (cerebral hemorrhage, brain trauma, brain tumor, and intracranial infection); (2) inability to complete quantitative motor or neuropsychological tests; and (3) refusal to undergo or a negative response to continuous ELD. After enrollment, all participants underwent comprehensive baseline clinical assessments and continuous ELD. Detailed methodologies were described in the following sections. The participant flow throughout the study is summarized in . This cohort study was conducted in accordance with the STROBE (Strengthening the Reporting of Observational Studies in Epidemiology) reporting guidelines (Checklist 1).

    Ethical Considerations

    This study was approved by the Ethics Committee of the West China Hospital, Sichuan University (No. 2022‐538). To ensure confidentiality, all data were anonymized before analysis and stored securely, with access restricted to the research team. Participants were not compensated financially but volunteered after being informed of the study’s potential benefits.

    Demographic Characteristics

    We systematically collected demographic information of the enrolled patients, including age, sex, years of education, medical history, and conducted a comprehensive neurological examination. The Japanese iNPH grading scale (iNPHGS) was used to quantify the severity of symptoms of iNPH [,]. Each symptom was scored from 0 to 4, with a total score of 12; higher scores indicating more severe clinical symptoms. The modified Rankin Scale (mRS) was used to assess the patients’ daily living function [], scored from 0 to 5, with higher scores indicating a more severe inability to perform daily living.

    Neuropsychological Assessments

    All neuropsychological tests were conducted by 2 trained neuropsychologists. Traditional neuropsychological assessment uses the MMSE to evaluate global cognitive function []. The BrainFit computerized neuropsychological assessment device (Beijing CAS-Ruiyi Information Technology Co, Ltd, China) was used to provide computerized neuropsychological tests, including the grammatical reasoning test, the one-back test, the trail-making test (TMT), and the Stroop color-word test (SCWT). The reliability of the BrainFit system was validated among Chinese populations in previous studies [,]. The grammatical reasoning test is designed to evaluate the language comprehension and reasoning ability of the participants []. In 60 seconds, they had to determine if the way the shapes were arranged on the screen matched the textual description. The shapes were a circle and a square with a particular positional relationship, and in the middle of the screen was a sentence that described this relationship. If the sentence and shape combination matched, participants clicked “correct”; if not, they clicked “incorrect.” A new question would display within a second of each click []. The score for this test was calculated as the number of correct responses minus the number of errors. The one-back test is based on the “N-back” working memory paradigm []. Within 60 seconds, participants were required to click the “same” or “different” button to indicate whether they thought the current displayed number matched the previous one. A new number appeared within 1 second after each click. The score was calculated as “the number of correct responses minus the number of incorrect responses” []. The Trail Making Test primarily assessed the participant’s attention and reaction speed, requiring the participant to sequentially connect 25 randomly arranged numbers on a page, with the test time recorded []. The Stroop Color-Word Test was used to evaluate executive function, psychomotor speed, and cognitive flexibility. During this study, participants were asked to quickly name the color of words that describe different colors, with test time and the number of correct responses recorded [].

    Gait Analysis

    Traditional gait analysis includes the TMWT and the 5-m TUG test [,]. The TMWT requires the patient to walk on a 10-m-long path while recording a video. The total time and the number of steps were recorded. The TUG test requires the patient to walk on a 5-m path from one end to the other, turn 180 degrees, and then walk back to the beginning. Each test recorded the overall time, number of steps, and number of turns necessary. Each of these tests was repeated 3 times, and the average of the 3 results was recorded.

    Quantitative gait analysis was evaluated using a 3-dimensional gait analysis system ReadyGo (Beijing CAS-Ruiyi Information Technology Co, Ltd), the accuracy and sensitivity of the system have been validated in previous studies [,,]. The ReadyGo system uses a single-camera setup to record 3-dimensional motion by using deep learning for precise positioning of skeletal points. For the single-gait test, participants were instructed to walk at their habitual pace on a 3 m walkway. Step width, stride length, step height, gait velocity, and turning time were the analyzed parameters. Step width was defined as the average width between the left and right feet in each image frame. A single-foot stride length was the distance between 2 landings from the same foot; the overall stride length was the average of the left and right sides. The step height was the height at which a foot could swing without touching the ground. The average step heights of the left and right feet were used for the final analysis. Gait velocity was defined as the distance between the start and end points divided by test time [].

    Continuous ELD Test

    Patients with possible iNPH underwent lumbar puncture and continuous ELD after obtaining informed consent. For 3 to 5 days, the daily CSF drainage volume was controlled between 100 and 150 mL [,,,]. The patients underwent traditional cognitive and gait assessments at baseline and every day after ELD, with extra digital neuropsychological and gait assessments conducted at baseline and on the third day following ELD. The criteria for a positive response to the traditional evaluation method of ELD included (1) improvement of ≥20% in either time or number of steps in the 10-m walking test or ≥10% improvement in both time and number of steps, (2) improvement of ≥10% in the TUG test time, or (3) improvement of ≥3 points in the MMSE score [,,].

    Postoperative Follow-Up

    Patients diagnosed with probable iNPH after ELD underwent lumboperitoneal shunt placement after informed consent was obtained. These patients were followed up regularly at 3 months, 6 months, and 1 year postoperatively. Follow-up was conducted through in-person visits or telephone interviews with the patients and their long-term caregivers. During the in-person visits, comprehensive neurological examinations and brain imaging were performed. Objective assessments of cognitive function, gait, urinary function, daily living abilities, subjective symptom improvements reported by the patients and caregivers, and postoperative complications were documented. Telephone interviews primarily assessed whether the patients and caregivers reported symptom improvement and identified any possible complications. Patients who showed an improvement of ≥1 point in mRS score compared to baseline or an improvement of ≥1 point in any iNPHGS score were defined as definite iNPH (shunt responders) [,]. Those who did not report symptom improvements during the follow-up period were classified as nonresponders and underwent pressure readjustment and long-term follow-up.

    Statistical Analyses

    The raw scores of grammatical reasoning, the one-back test, TMT, SCWT correct number, and reaction time were converted into standardized Z-scores based on previously reported norms for the Chinese population, facilitating comparisons across tests [,]. The z-scores for the Stroop C correct number, grammatical reasoning test, and one-back test were calculated using the formula (raw score−mean)/SD. Z-scores for TMT and Stroop C reaction time were derived by subtracting the raw score from the mean and dividing by the SD. Lower z-scores indicated more significant impairments in the corresponding cognitive domain. The cognitive Z-score for every patient was calculated by taking the mean of the Z-scores from the tests mentioned earlier [,]. The improvement rate of the cognitive Z-score was calculated as follows: (post-ELD parameter−pre-ELD parameter)/pre-ELD parameter×100%. For quantitative gait analysis, the improvement rates in gait velocity, stride length, and step height post-ELD were calculated as (post-ELD parameter − pre-ELD parameter)/pre-ELD parameter×100%. The improvement rates of the step width and turning time post-ELD were calculated as follows: (pre-ELD parameter−post-ELD parameter)/pre-ELD parameter×100%. The gait improvement rate was calculated as averages of all 5 gait parameters, and the combined improvement rate was averaged based on the improvement rate of the cognitive Z-score and gait improvement rate []. In this pilot study, gait and cognitive improvement were assigned equal weight in the combined improvement rate to reflect global functional improvement after ELD, as the gait and cognitive impairment are core symptoms in iNPH and are considered equally important in current clinical assessments such as iNPHGS [,].

    For continuous variables, normally distributed data are shown as mean (SD), whereas nonnormally distributed data are presented as median (IQR). Group comparisons were performed using the Student t test for normally distributed data and the Mann-Whitney U test for skewed data, whereas comparisons of proportions were performed using the χ2 test. Group comparisons before and after the ELD were performed using paired t tests for normally distributed data and Wilcoxon signed-rank tests for nonnormally distributed data. To address the limited sample size, we used Firth penalized logistic regression to evaluate the predictive value of improvement rates from each evaluation method for shunt response, and variables demonstrating significant difference in group comparison were included in subsequent multivariate models. This method effectively reduced the small-sample bias and provided more reliable coefficient estimates []. The results of Firth logistic regression models were reported by odds ratios (ORs) and receiver operating characteristic curves. Optimal cutoff values were determined based on the Youden index, and diagnostic metrics, including area under the curve (AUC), sensitivity, specificity, positive predictive value, and negative predictive value, were calculated. The DeLong test was used for AUC comparison between traditional tests and digital tests. Permutation tests with 5000 iterations on all Firth regression models and internal validation of the diagnostic metrics with 2000 bootstrap resamples were used to assess the model performance and further enhance the robustness of the statistical inference []. A P value of .05 was considered statistically significant. All statistical analyses were performed using STATA SE 16.0 (StataCorp, TX, USA) and R version 4.3.0 (R Foundation).

    Demographic and Clinical Characteristics of Patients With Probable iNPH

    A total of 127 patients diagnosed with possible NPH were consecutively enrolled in this study (). After excluding 7 patients who refused lumbar puncture, 13 patients who developed secondary NPH, and 30 patients who were unable to cooperate with the quantitative motor and cognitive tests, 77 patients with possible iNPH have finally received continuous ELD. Of them, 70 patients were ELD responders and were diagnosed as probable iNPH by clinical neurologists, whereas 7 were negative for ELD tests. Among the 70 patients with probable iNPH, 31 refused shunt surgery and were ultimately diagnosed with probable iNPH, and 39 (55.7%) underwent lumboperitoneal shunt surgery.

    Figure 1. Study flowchart. iNPH: idiopathic normal pressure hydrocephalus; MRI: magnetic resonance imaging.

    presents the baseline characteristics of the 70 patients with probable iNPH. The median (IQR) age was 74 (69-80) years, and 70% (49/70) of the patients were male. The prevalence of cognitive impairment, gait disturbances, and urinary incontinence was 92.86% (65/70), 95.71% (67/70), and 54.29% (38/70), respectively, with 51.43% presenting with the full Hakim triad (). We compared the baseline characteristics between patients with iNPH who received shunt surgery and those who did not, and the shunted group exhibited a significantly more severe DESH score on brain magnetic resonance imaging (P=.007), whereas the 2 groups did not differ in demographics, symptom severity, or improvement rates after ELD. This suggests that the 2 groups were largely comparable at baseline.

    Table 1. Baseline characteristics of patients with probable iNPH by the shunt status.
    Variables All (n=70) No shunt (n=31) Shunted (n=39) P value
    Demographics
    Age (y), median (IQR) 74.00 (69.00 to 80.00) 74.00 (69.00 to 80.00) 75.00 (71.00 to 78.00) .64
    Sex (male), n (%) 49 (70.00) 19 (61.29) 30 (76.92) .16
    Education level (y), median (IQR) 12.00 (9.00 to 15.00) 12.00 (9.00 to 12.00) 9.00 (9.00 to 15.00) .45
    Hyperlipidemia, n (%) 28 (40.00) 16 (51.61) 12 (30.77) .08
    Hypertension, n (%) 35 (50.00) 14 (45.16) 21 (53.85) .47
    Diabetes, n (%) 22 (31.43) 8 (25.81) 14 (35.90) .37
    Triad, n (%) 36 (51.43) 12 (38.71) 24 (61.54) .06
    Cognitive decline, n (%) 65 (92.86) 28 (90.32) 37 (94.87) .46
    Gait disturbance, n (%) 67 (95.71) 30 (96.77) 37 (94.87) .70
    Urinary incontinence, n (%) 38 (54.29) 14 (45.16) 24 (61.54) .17
    mRS (scores), median (IQR) 2.00 (2.00 to 3.00) 2.00 (2.00 to 3.00) 3.00 (2.00 to 3.00) .34
    iNPHGS (scores), mean (SD) 5.27 (2.22) 4.87 (2.03) 5.59 (2.32) .18
    Evans index, median (IQR) 0.33 (0.31 to 0.35) 0.32 (0.30 to 0.34) 0.33 (0.32 to 0.36) .10
    DESH score, median (IQR) 6.00 (5.00 to 7.00) 5.00 (4.00 to 7.00) 6.00 (6.00 to 7.00) .007
    Baseline neuropsychological tests
    MMSE(score), median (IQR) 21.00 (14.00 to 25.00) 19.00 (13.00 to 24.00) 21.00 (15.00 to 25.00) .65
    Grammatical reasoning (score), median (IQR) 1.00 (0.00 to 3.00) 1.00 (0.00 to 2.00) 1.00 (0.00 to 3.00) .53
    One-back test (score), median (IQR) 5.00 (2.00 to 9.00) 6.00 (2.00 to 9.00) 5.00 (2.00 to 9.00) .95
    Trail-making test (s), median (IQR) 140.00 (103.00 to 150.00) 140.00 (103.00 to 150.00) 140.00 (122.00 to 150.00) .98
    Stroop color-word test (score), median (IQR) 36.00 (14.00 to 45.00) 37.00 (34.00 to 46.00) 34.00 (14.00 to 42.00) .28
    SCWT reaction time (s), median (IQR) 150.00 (116.56 to 186.65) 154.56 (142.25 to 222.46) 148.92 (106.41 to 183.66) .10
    Cognitive z-score, mean (SD) −2.35 (1.19) −2.46 (1.27) −2.26 (1.12) .51
    Baseline gait tests
    5-m TUG (s), median (IQR) 18.00 (15.00 to 27.00) 22.00 (16.00 to 30.00) 17.00 (15.00 to 22.00) .13
    TMWT (s), median (IQR) 15.00 (12.00 to 22.00) 17.00 (13.00 to 25.00) 15.00 (12.00 to 18.00) .13
    TMWT steps, median (IQR) 26.00 (21.00 to 38.00) 30.00 (23.00 to 42.00) 25.00 (19.00 to 33.00) .06
    Step width (m), median (IQR) 0.16 (0.14 to 0.17) 0.15 (0.14 to 0.17) 0.16 (0.15 to 0.16) .68
    Stride length (m), mean (SD) 1.30 (0.50) 1.28 (0.49) 1.31 (0.50) .81
    Step height (m), mean (SD) 0.08 (0.03) 0.08 (0.02) 0.08 (0.03) .38
    Gait velocity (m/s), mean (SD) 0.68 (0.22) 0.66 (0.25) 0.69 (0.20) .70
    Turning time (s), median (IQR) 2.22 (1.57 to 3.40) 2.22 (1.57 to 2.95) 2.28 (1.73 to 3.57) .47
    Improvement rate after ELD
    Cognitive improvement (%), median (IQR) 19.5 (−15.9 to 41.9) 27.9 (−5.3 to 47.6) 2.0 (−17.3 to 32.1) .14
    Gait improvement (%), median (IQR) 5.1 (−3.5 to 15.1) 5.5 (−3.5 to 12.6) 5.1 (−0.4 to 14.8) .99
    Combined improvement (%), median (IQR) 12.4 (−6.5 to 26.7) 16.7 (5.8 to 30.1) 3.2 (−7.9 to 21.6) .12

    aiNPHGS: iNPH grading scale.

    bThe Mann-Whitney U test was used for group comparisons.

    cmRS: modified Rankin scale.

    dThe Student t test was used for group comparisons.

    eDESH: disproportionately enlarged subarachnoid hydrocephalus.

    fStatistically significant.

    gMMSE: Mini-Mental State Examination.

    hSCWT: Stroop color-word test.

    iTUG: timed up and go test.

    jTMWT: 10-m walking test.

    k ELD: external lumbar drainage.

    As shown in and in , among the 70 patients with probable iNPH, traditional tests, including MMSE (P<.001), TUG (P=.004), TMWT time (P=.016), and steps (P<.001), showed significant improvement 3 days after ELD. For digital tests, one-back test (P=.01), SCWT correct numbers (P=.009), and composite cognitive z-scores (P=.025) improved significantly 3 days after ELD. Quantitative gait analyses also showed significant improvement in stride length (P=.01), step height (P=.048), gait velocity (P=.019), and reduced turning time (P<.001) after 3-day ELD compared to baseline.

    Figure 2. Changes in cognitive and gait parameters after 3-d ELD. (A) Mini-Mental Status Examination (MMSE); (B) 5-m timed up and go test (5m-TUG); (C) 10-m walking test (TMWT) time; (D) 10-m walking test (TMWT); (E) one-back test; (F) stroop color-word test (SCWT); (G) stride length; (H) step height, (I) gait velocity, and (J) turning time. ELD: external lumbar drainage.

    Predictive Value of Digital Neuropsychological and Gait Tests for Shunt Outcome in Patients With iNPH

    A total of 39 patients with probable iNPH underwent lumboperitoneal shunt placement; after a median follow-up interval of 116 days, 34 (89.7%) showed at least a 1-point improvement in mRS or iNPHGS, leading to a clinical diagnosis of definite iNPH or being classified as “shunt responders,” 4 patients only reported subjective symptomatic improvement but not measurable through mRS or iNPHGS, and 1 patient did not report any improvement at the time of final follow-up and was managed with pressure readjustment and continued follow-up.

    First, we compared the clinical characteristics between shunt responders and nonresponders (). The results demonstrated a higher proportion of male sex (P=.036), lower Evan index (P=.035), and significantly higher improvement rate of gait after ELD (P=.04) for shunt responders, whereas they did not differ significantly in age, educational level, or vascular risk factors. Multivariate Firth logistic regression model adjusted for sex and Evan index showed a significant association between a higher improvement rate of digital gait analysis and a lower risk of unfavorable shunt outcome (adjusted OR 0.90, 95% CI 0.78‐0.99; P=.03). In addition, an association between a higher combined improvement rate of digital neuropsychological and gait analysis and a lower risk of unfavorable shunt outcome was also observed (adjusted OR 0.98, 95% CI 0.95‐1.00; P=.03; ).

    Table 2. Clinical characteristics of iNPH patients who received lumboperitoneal shunt.
    Variables All (n=39) Subjective improvement and nonresponders (n=5) Objective responders (n=34) P value
    Age (y), median (IQR) 75.00 (71.00 to 78.00) 67.00 (66.00 to 73.00) 76.00 (71.00 to 81.00) .10
    Sex, male, n (%) 30 (76.92) 2 (40.00) 28 (82.35) .04
    Education level (y), median (IQR) 9.00 (9.00 to 15.00) 9.00 (9.00 to 12.00) 9.00 (9.00 to 15.00) .49
    Hyperlipidemia, n (%) 12 (30.77) 2 (40.00) 10 (29.41) .63
    Hypertension, n (%) 21 (53.85) 2 (40.00) 19 (55.88) .51
    Diabetes, n (%) 14 (35.90) 3 (60.00) 11 (32.35) .23
    Evans index, median (IQR) 0.33 (0.32 to 0.36) 0.38 (0.34 to 0.38) 0.33 (0.31 to 0.34) .04
    DESH score, mean (SD) 6.31 (1.32) 5.80 (0.98) 6.38 (1.35) .37
    Cognitive improvement rate after ELD, %, median (IQR) 2.01 (−17.35 to 32.07) −10.56 (−31.43 to 28.94) 2.01 (−17.35 to 33.55) .47
    Gait improvement rate after ELD, %, median (IQR) 5.07 (−0.38 to 14.77) −12.32 (−14.83 to −2.27) 5.24 (1.63 to 17.05) .04
    Combined improvement rate after ELD, %, median (IQR) 3.17 (−7.93 to 21.58) −12.70 (−16.85 to 14.34) 3.17 (−6.47 to 26.72) .28

    aiNPH: idiopathic normal pressure hydrocephalus.

    bStatistically significant.

    cDESH: disproportionately enlarged subarachnoid hydrocephalus.

    dELD: external lumbar drainage.

    As shown in and , traditional tests in ELD yielded poor diagnostic performance, with an AUC of 0.55 (95% CI 0.30‐0.81), sensitivity of 40% (95% CI 12%‐77%), specificity of 71% (95% CI 54%‐83%), PPV of 17% (95% CI 5%‐45%), and NPV of 89% (95% CI 72%‐96%). In contrast, the combined digital cognitive and gait approach yielded an AUC of 0.92 (95% CI 0.83‐1.00), sensitivity of 100% (95% CI 57%‐100%), specificity of 79% (95% CI 63%‐90%), PPV of 42% (95% CI 19%‐68%), and NPV of 100% (95% CI 88%‐100%). The bootstrap-derived cutoffs showed moderate dispersion, whereas the predictive performances of all digital tests remain relatively stable (lower limits of all AUCs >0.81). The DeLong test demonstrated that combining digital cognitive and gait tests showed better predictive performance of shunt outcome compared to traditional tests (Z=2.43; P=.015).

    Furthermore, the calibration curve demonstrated that the combined improvement model showed good overall calibration quality (, Spiegelhalter P=.62), with a low average calibration error of 3.0%, despite slight overconfidence (calibration slope=1.299).

    Table 3. The predictive efficacy of traditional tests, digital neuropsychological, and gait tests. All CIs were calculated using 2000 bootstrap resamples.
    Cutoff
    (95% CI)
    AUC
    (95% CI)
    Sensitivity (95% CI) Specificity (95% CI) PPV
    (95% CI)
    NPV
    (95% CI)
    Traditional tests 0.159 (0.06‐0.29) 0.553 (0.301‐0.805) 0.400 (0.118‐0.769) 0.706 (0.538‐0.832) 0.167 (0.047‐0.448) 0.889 (0.719‐0.961)
    Digital tests
     Gait improvement rate after ELD 0.138 (0.10‐0.60) 0.929 (0.830‐1.000) 1.000 (0.566‐1.000) 0.765 (0.600‐0.876) 0.385 (0.177‐0.645) 1.000 (0.871‐1.000)
     Cognitive improvement rate after ELD 0.125 (0.08‐0.60) 0.912 (0.812‐1.000) 1.000 (0.566‐1.000) 0.794 (0.632‐0.897) 0.417 (0.193‐0.680) 1.000 (0.875‐1.000)
     Combined improvement rate after ELD 0.132 (0.08‐0.62) 0.924 (0.829‐1.000) 1.000 (0.566‐1.000) 0.794 (0.632‐0.897) 0.417 (0.193‐0.680) 1.000 (0.875‐1.000)

    a AUC: area under the curve.

    bPPV: positive predictive value.

    cNPV: negative predictive value.

    dELD: external lumbar drainage.

    Figure 3. Receiver operating characteristic curve analysis comparing the predictive value of traditional tests, digital neuropsychological and gait tests in differentiating shunt-responders from non-responders. AUC: area under the curve.

    Principal Findings

    In this study, we investigated the predictive value of digital neuropsychological and gait analyses during ELD for shunt outcome in patients with iNPH. Our findings revealed that while both traditional and digital cognitive and gait assessments improved significantly after 3 days of ELD, the digital tests outperformed traditional testing in terms of predicting shunt outcomes.

    Our study found that patients with probable iNPH showed gait improvement after ELD, which was primarily manifested as increased gait speed, longer stride length, higher step height, and shorter turning time. This is consistent with the findings of prior investigations using quantitative gait analysis []. Electronic walkways, wearable sensors, and accelerometers are all commonly used for quantitative gait analysis [,]. A prospective study using electronic walkways discovered that patients with iNPH had increased stride length and gait speed following a tap test []. Another research study has revealed that 72 hours following the tap test, patients with probable iNPH had longer stride lengths, shorter double support times, and faster cadences []. A recent study based on 3-dimensional gait analysis indicated that patients with probable iNPH may show improvements in spatiotemporal parameters, such as step length, gait speed, and cadence 24 hours after the tap test, with improvement rates ranging from 4.9% to 10.5% []. Besides, a preliminary study using a video-based method qualitatively assessed gait changes after the tap test and found significant improvements in step height [], whereas earlier studies based on inertial sensors and electronic walkways may not provide precise quantitative assessments of step height. Our vision-based system provided richer spatiotemporal gait parameters than previous methods, without the need for wearable sensors. Therefore, it can provide improvements in step height following ELD, which partially complements the findings of previous studies.

    Cognitive impairment is the second most common symptom in patients with iNPH, which may be partly due to mechanical stress to the brain and the existence of Alzheimer disease co-pathologies [,]. Frontal lobe dysfunction is thought to be the classic cognitive profile in patients with iNPH []. In our study, we observed significant improvements in one-back tests and Stroop color-word tests post-ELD, indicating improvements in executive function, attention, and working memory, all of which are associated with improved frontal lobe functions. Patients with iNPH often show significant improvements in executive subfunction after CSF drainage [], with prior research showing improvements in verbal fluency, frontal assessment battery, trail-making test, and Stroop color-word tests [-]. Therefore, executive subfunction assessment is emphasized in evaluations during CSF drainage to distinguish iNPH from its mimics [,]. Previous studies have developed an executive function battery for patients with iNPH based on tests such as TMT-A, Stroop color-word tests, and digit symbol substitution tests [], and this battery demonstrates a sensitivity of 80% in predicting shunt responders with a specificity of 100%, indicating that evaluating executive function, attention, and reaction time before and after CSF drainage may be crucial in identifying shunt responders [].

    Despite the widespread use of the CSF drainage test for diagnosing and predicting shunt prognosis in patients with iNPH, earlier research indicated that the sensitivity of a single-tap test might be as low as 26%, whereas continuous ELD had a sensitivity of approximately 50% [,]. Although the tap test response would be influenced by morphological features and CSF dynamic changes [], the method used to evaluate the CSF drainage test itself may be crucial for determining its overall sensitivity [,]. In the latest Japanese iNPH management guidelines, MMSE, TMWT, and TUG tests were recommended to assess changes in patients’ gait and cognitive function []. However, using these methods for assessment may result in very low sensitivity and negative predictive value []. A previous large-scale European multicenter study indicated that the negative predictive value of a single lumbar puncture drainage could be as low as 18% [], with other literature reports generally ranging from 18% to 50% []. In our cohort, traditional cognitive and gait tests exhibited a sensitivity of only 40% and a specificity of 70%, which demonstrated that traditional tests may potentially lead to misdiagnosis of shunt responders. Therefore, traditional assessment methods may be insufficient to guide the selection of shunt candidates [].

    In recent years, digital neuropsychological evaluation equipment has emerged that provides randomized test paradigms and automated recording of variables such as reaction time, thereby improving test efficacy []. Preliminary research has used digital cognitive and gait evaluation tools for patients with iNPH, establishing computerized neuropsychological tests as reliable techniques for diagnosing cognitive impairments and postoperative cognitive improvements in patients with iNPH []. In contrast, a recent meta-analysis suggested that quantitative gait analysis can detect gait improvements in patients with iNPH at baseline, after the tap test, and postshunt []. These sensor-based technologies and digital systems possess high scalability, offering continuous and high-precision monitoring of cognitive and motor functions []. Our results further emphasized the predictive value of these digital assessment tools during ELD for patients with iNPH, which also showed better performance in predicting shunt responses. Therefore, the use of digital evaluation tools may aid in the selection of potential shunt candidates. Integrating these digital data into telehealth platforms would enable remote and continuous health care for patients with geographic barriers, reduce hospital visits, and enhance patient outcomes []. However, the results of our study are still preliminary; research gaps for digital assessments still exist in modest sample size, clinical standardization, and multicenter validation. Future research should explore more data-driven weighting schemes and conduct external validation as more data become available.

    Strengths and Limitations

    The main advantages of our study are the prospective cohort study design, which collected detailed clinical data, and comprehensive neuropsychological and gait assessment data from the patients. Additionally, we are the first to combine computerized neuropsychological assessment and 3-dimensional gait analysis to evaluate symptom changes during ELD in patients with iNPH. The limitations of the study are as follows: (1) Nearly half of the patients with positive ELD responses refused shunt surgery, and the relatively small sample size of shunted patients may constrain the statistical power, despite our use of Firth penalized regression models. Therefore, the relatively high AUC and sensitivity values should be interpreted with caution. Future multi-center studies with larger samples are needed to validate our preliminary findings. (2) Cut-off values were calculated statistically based on the Youden index. Given the relatively small sample size, these data-derived thresholds should be interpreted as preliminary estimations that require external validation in future studies with larger cohorts. (3) The high rate of shunt refusal may inevitably introduce selection bias, although the baseline characteristics between patients who accepted and refused shunts were largely comparable, except for DESH score (). (4) Several patients were excluded because of severe illness; therefore, these results may be primarily applicable to patients who have iNPH with mild-to-moderate symptoms. (5) In this study, quantitative assessments were only completed on the third day after ELD. Although previous studies indicate that a 3-day continuous ELD is sufficient to improve cognitive and gait function in patients with iNPH [,], a recent longitudinal study suggests that patients with iNPH would exhibit delayed response up to 2 weeks after ELD []. Therefore, further research is needed to determine the best evaluation time point. (6) Due to the relatively short follow-up interval, the long-term predictive value of these digital tests for shunt response was unclear, which may be affected by various prognostic factors []. (7) Although our findings on digital assessments are promising, the generalizability across different health systems and the cost-effectiveness of these digital assessments are underexplored.

    Conclusions

    Digital neuropsychological and motor assessments could identify gait and cognitive improvements after continuous ELD. Moreover, the digital tests showed better predictive performance for shunt outcome compared to traditional tests in our pilot study. Further validation of our findings in a multicenter setting is required to establish the optimal cutoff values for these digital evaluation tests.

    The authors would like to express their gratitude to all patients and their families who participated in this study. They also extend our acknowledgments to all members of the West China Hospital Multidisciplinary Team of Hydrocephalus for their contribution.

    This study was supported by the STI2030-Major Projects Youth Scientist Program (2022ZD0213600), National Natural Science Foundation of China (U23A20422 and 82071203), and the Young Scientists Fund (82201608).

    All data are available from the corresponding author upon request.

    Writing – review & editing: ZH, NH, HG, FY, SF, LQ, RW, XY, SW, QL, YL, DZ, LZ, JH, QC

    None declared.

    Edited by Javad Sarvestan; submitted 03.Jun.2025; peer-reviewed by Andrea Bianconi, Efstratios-Stylianos Pyrgelis; accepted 29.Oct.2025; published 25.Nov.2025.

    © Hanlin Cai, Keru Huang, Zilong Hao, Na Hu, Hui Gao, Feng Yang, Shiyu Feng, Linyuan Qin, Ruihan Wang, Xiyue Yang, Shan Wang, Qian Liao, Yi Liu, Dong Zhou, Liangxue Zhou, Jiaojiang He, Qin Chen. Originally published in the Journal of Medical Internet Research (https://www.jmir.org), 25.Nov.2025.

    This is an open-access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work, first published in the Journal of Medical Internet Research (ISSN 1438-8871), is properly cited. The complete bibliographic information, a link to the original publication on https://www.jmir.org/, as well as this copyright and license information must be included.

    Continue Reading

  • Journal of Medical Internet Research

    Journal of Medical Internet Research

    There is a critical need for scalable, cost-effective interventions to address high rates of physical inactivity and related chronic diseases, especially in underserved populations considered to be at risk. In fact, the World Health Organization estimates that further inaction could lead to 500 million new global cases of preventable noncommunicable diseases, with associated direct health care costs of US $520 billion, by 2030 []. Internet-based physical activity (PA) interventions have great potential for widespread dissemination and have already been shown to improve PA levels [-]. However, most of this work was conducted in the general population and not extended to those most at risk, including underrepresented minority populations.

    Hispanic adults report the highest rates of inactivity outside of work (32.1% vs 23% for non-Hispanic White individuals) and experience disproportionate rates of associated conditions (eg, obesity and diabetes) [,]. In fact, 79% of Latinas are overweight or obese, compared to 64% of non-Hispanic White women []. Thus, in the Pasos Hacia La Salud study, we tested a web-based, PA intervention in Spanish with 205 Latinas and found significantly greater increase in weekly minutes of moderate-to-vigorous PA (MVPA) compared to a control condition [,]. Moreover, the costs associated with providing the intervention were low at 6 months (US $17 per person per month) and further decreased (US $12 per person per month) at 12 months []. While the Pasos program was shown to be effective and low cost, most intervention participants (69%) did not meet national PA guidelines at 6 months [,,], and longer-term maintenance was not evaluated.

    To help sedentary Latinas achieve and maintain health-enhancing levels of PA, the intervention was refined by adding SMS text messages and further targeting key social cognitive theory [] variables (eg, self-efficacy, enjoyment, and social support). Subsequently, in the Pasos Hacia La Salud II study, the new technology- and theory-enhanced version was tested and compared with the original Pasos Hacia La Salud intervention in a randomized controlled trial (N=205) []. There were no significant between-group differences in PA at 6 or 12 months; however, the enhanced intervention arm had higher PA levels than the original intervention arm at 18 and 24 months [].

    While both programs have been shown to be beneficial, the enhanced intervention appears to have an advantage in terms of long-term behavior change. However, these enhancements (eg, SMS text messages, discussion board, and additional staff contacts) likely come at additional expense (compared to the original arm). An evaluation of intervention costs is needed to determine which program is feasible and the best fit for the clinical and community setting and resources. Thus, to inform future dissemination and implementation efforts, this study examined additional costs associated with intervention enhancements and how they influence cost-effectiveness.

    Overview

    Pasos Hacia La Salud II was a fully powered randomized trial of 2 PA interventions: the original web-based PA intervention (original) and an enhanced arm that included additional elements to support increasing activity (enhanced). Participants were adult Latinas aged 18 to 65 years who were underactive (engaging in <60 min of MVPA per week), could read and write in Spanish, had access to technology to use the study website and receive SMS text messages, and were healthy enough for unsupervised exercise. A full description of the study protocol, measures, and participants has been published previously [,].

    Intervention Description

    In the original arm of the web-based intervention, participants attended a baseline session led by trained bilingual interventionists who explained MVPA and helped participants set incremental goals. Participants also completed goal-setting sessions over the phone at 1 and 9 months and at follow-up visits (6 and 12 mo). Participants were encouraged to accumulate MVPA on most days of the week and encouraged to achieve the goal of 150 minutes of MVPA by 6 months. All participants were given a pedometer (Accusplit Inc) and were encouraged to use it to self-monitor daily activity.

    The interventionist also oriented participants to the study website, which participants could access throughout the intervention period (24 months). The website included tools to help participants increase their MVPA, such as self-monitoring, goal setting, social support, instruction about how to be physically active, problem solving, motivational stage-matched PA manuals, and individually tailored feedback. Further details about the tailored intervention content are described elsewhere []. Participants were encouraged to regularly access the website, log their PA on the website, and receive a tip of the week. Participants were also prompted to complete online surveys to customize their intervention content. Most web-based content was automated and did not require staff time. However, staff regularly checked the message boards to answer questions as necessary and were available to help participants with any technology issues.

    In addition to the intervention components described earlier, the enhanced intervention included (1) SMS text messages; (2) additional phone calls at 2, 3, 15, and 21 months; (3) additional in-person visits at 18 and 24 months; and (4) additional data-driven content and website features that encouraged user participation. Participants were also provided with nonmonetary incentives for their website use. Participants earned points by logging into the website and using certain features, such as the goal-setting calendar. Once participants earned a given number of points, they were sent prizes, such as water bottles, phone cases, and T-shirts.

    To further enhance social support for PA, a discussion board was used to facilitate meetups among participants. Research staff posted details about free and low-cost PA events in the area so that participants could meet, discuss coordinating attendance with others, overcome barriers to PA, and motivate each other to attend these events.

    Ethical Considerations

    The study was reviewed and approved by the Brown University Human Research Protections Program (1708001868), and all participants provided written informed consent in the language of their choosing (English or Spanish). To protect confidentiality, all data were deidentified for storage and analysis. To thank them for their time, participants received compensation for participating in study activities. Participants received US $25 for completing each of their assessment visits (at baseline and 6, 12, 18, and 24 months) with an additional US $50 bonus at 24 months if they completed all visits. In addition, all participants received incentives for filling out monthly questionnaires (US $10 per month) to generate the tailored website content. Participants also received reimbursement costs related to travel and childcare costs. Participants in the enhanced arm also received prizes for engaging with website content (as mentioned earlier). This trial has been registered at ClinicalTrials.gov (NCT03491592),

    PA Measures

    The main outcomes used to determine cost-effectiveness were the change in minutes of MVPA from baseline to 12 and 24 months, as measured by ActiGraph accelerometers and self-report, and the percentage of participants meeting national guidelines at 12 and 24 months. Self-reported activity was measured using the 7-day PA recall (PAR) interview, an interviewer-administered measure that has shown good reliability and validity in adult populations, including in Spanish populations [-]. Adherence to national guidelines was defined as engaging in ≥150 minutes per week of MVPA, as measured by the 7-day PAR [].

    Costs

    Overview

    Cost calculations followed the model used in our previous publications []. Costs were calculated from a payer perspective to estimate the amount it would cost to deliver the intervention in a clinical or community setting. This included all costs associated with personnel, materials, and use and maintenance of technology to deliver the intervention, sourced from actual costs incurred during the trial.

    Costs associated with research, such as recruitment, baseline and follow-up measurement visits, obtaining consent, or compensation for research participation, were not included. We also did not include costs associated with intervention development. Thus, both research and development costs are not relevant to the future implementation of the existing intervention in community or clinical settings. Excluding these costs is consistent with published guidelines on cost-effectiveness analyses [].

    Personnel Time

    Staff time logs were used to determine the time needed to complete each activity associated with intervention delivery. This included training time (for both the trainer and the trainee); time to compile study materials; time for study visits and phone calls (depending on study condition); and time for study maintenance activities, such as checking the online message board, resending pedometers to participants who lost them, calling the website developer with technical questions, or (in the enhanced group) offering SMS text message support. Study maintenance activities were conducted during a set time each week, rather than calculated as a per-participant activity.

    Costs for personnel were calculated by determining the total time needed to deliver the intervention per participant in each condition and multiplying by actual staff salary rates, including benefits. Staff delivering the intervention were entry-level research staff with an undergraduate degree (earning a yearly salary of US $54,080 plus 31% in benefits, for a total hourly cost of US $34.05). Training staff were master’s-level behavioral scientists (earning a yearly salary of US $68,640, plus 31% in benefits, resulting in a total hourly cost of US $43.20). Overhead was estimated as an additional 10% of all personnel costs to account for the use of shared space.

    Technology

    Web hosting was US $75 per month for both study arms. The automated SMS text messaging was supported through the study website and thus did not incur any additional costs.

    Materials

    Costs for materials were based on actual costs incurred during intervention delivery. All participants in both study arms received an Acusplit pedometer to record their daily steps for US $20 each. Participants in both arms also received a study binder at their baseline visit with information about exercise, tip sheets, and logs for using their pedometers. The total cost for the binders and printed materials was US $4.95 for each participant.

    Total cost for prizes in the enhanced group was calculated based on the actual cost of the items and the number of each item that was distributed. As not all participants earned all prizes, we calculated an average cost per participant for prizes to allow for an estimate of the actual cost of delivering prizes at varying rates based on participant engagement. Prize costs ranged from US $1.11 each for water bottles to US $12.50 for a yoga mat. Total cost for prizes in the enhanced arm was US $2038.

    A computer was also purchased for the study staff to conduct baseline study visits, check the study website message boards, etc. The cost of the computer was US $1200.

    Analysis

    To allow for comparison, we used the same analysis approach as in the parent study []. Costs were calculated from a payer perspective to estimate the cost of implementing the intervention in a community or clinical setting. Total costs were calculated separately for each study arm and included the total of personnel time (including benefits and use of shared space), materials, and web hosting. Total costs for each arm were divided by the number of participants in each study arm to calculate the cost per person to deliver each intervention. Costs were calculated in US dollars for 2024.

    Cost-effectiveness was calculated as the cost per additional minute of MVPA per person over the course of the study. Total increase in minutes was calculated using linear interpolation of minutes across measurement time points (baseline and 6, 12, 18, and 24 months) and subtracting baseline minutes. Main outcomes for costs and cost-effectiveness were calculated at 12 months (main study outcome) and cumulatively at 24 months. The cost of intervention delivery per person was divided by the cumulative increased minutes per person over the course of the study to estimate the cost per additional minute per person. This was done both for self-reported activity (7-day PAR) and objectively measured activity (ActiGraph accelerometer). Similarly, the cost of each intervention was divided by the number of participants in that group who met national guidelines at each time point to estimate the cost of moving 1 person from being inactive to successfully meeting guidelines for activity at 12 and 24 months.

    Incremental cost-effectiveness ratios (ICERs) were calculated to determine the additional cost per minute of MVPA in the enhanced group beyond that achieved by the original group, as well as the cost per additional individual meeting guidelines in the enhanced group compared with the original group. ICERs were calculated by dividing the difference in the costs between the 2 study arms by the difference in cumulative minutes between the 2 arms and, separately, by the difference in the number of people meeting guidelines. For ICERs for guidelines, CIs were computed using the nonparametric bootstrap with 1000 replications.

    Overview

    Participants (N=195) had a mean age of 43.3 (SD 10.29) years and were primarily of Dominican (80/195, 41%) and Colombian (33/195, 16.9%) descent. Approximately half of the participants (92/195, 47.2%) had a high school education or less. A full description of baseline characteristics and a CONSORT (Consolidated Standards of Reporting Trials) diagram have been published previously [].

    PA Changes

    As published previously [], participants in the enhanced group (103/195, 52.8%) increased self-reported weekly MVPA from 57.9 minutes at baseline to 115 minutes at 6 months, 106.8 minutes at 12 months, and 135.3 minutes at 24 months, while the original group (92/195, 47.2%) increased self-reported weekly MVPA from 55.2 minutes at baseline to 88 minutes at 6 months, 111.7 minutes at 12 months, and 93.9 minutes at 24 months []. ActiGraph-measured weekly MVPA in the enhanced group changed from 19.7 minutes at baseline to 47 minutes at 6 months, 44.5 minutes at 12 months, and 47.4 minutes at 24 months. In the original group, ActiGraph-measured weekly MVPA changed from 20.6 minutes at baseline to 43 minutes at 6 months, 55.9 minutes at 12 months, and 31.2 minutes at 24 months.

    None of the participants met national MVPA guidelines at baseline. At 12 months, 28.2% (29/103) and 29% (27/92) of participants met guidelines according to self-report in the enhanced and original conditions, respectively. At 24 months, this increased to 36.9% (38/103) and 31% (29/92) in the enhanced and original groups, respectively.

    Costs

    Total costs associated with delivering the interventions are shown in . Total cost of delivering the original intervention at 12 months was US $15,741, or US $171 per person (US $14 per person per month). The enhanced intervention cost US $20,435 to deliver over the first 12 months, or US $198 per person (US $16 per person per month).

    Table 1. Costs of delivering the enhanced and original interventions.
    Costs Original intervention (n=92) Enhanced intervention (n=103)
    12 mo 24 mo (cumulative) 12 mo 24 mo (cumulative)
    Personnel, US $
    Training 510 510 510 510
    Intervention delivery 10,387 10,723 13,495 21,845
    Website, US $
    Hosting 900 1800 900 1800
    Technical support 449 899 449 899
    Materials, US $
    Computer 1200 1200 1200 1200
    Pedometers 1840 1840 2060 2060
    Paper, binders, etc 455 455 510 510
    Prizes a 1311 2038
    Total costs, US $ 15,741 17,428 20,435 30,862
    Average cost per participant, US $ 171 189 198 300
    Average cost per participant per month, US $ 14 8 16 13

    aNot applicable.

    At 24 months, cumulative costs for the original intervention increased slightly to US $17,428, or US $189 per person (US $8 per person per month). However, the cumulative costs for the enhanced intervention increased to US $30,862 at 24 months, or US $300 per person (US $12 per person per month). The largest source of the difference was personnel cost for intervention delivery between 12 and 24 months ( and ).

    Table 2. Staff time needed for intervention activities.
    Time Original intervention (n=92) Enhanced intervention (n=103)
    12 mo 24 mo (cumulative) 12 mo 24 mo (cumulative)
    Training, min
    Trainee 360 360 360 360
    Trainer 360 360 360 360
    Intervention activities per person, min
    Assembling materials 5 5 5 5
    Baseline goal setting 60 60 70 70
    1-wk call 10 10 15 15
    1-mo call 25 25 30 30
    2-mo call a 10 10
    3-mo call 10 10
    6-mo goal setting 25 25 30 30
    9-mo call 25 25 30 30
    12-mo goal setting 25 30
    15-mo call 30
    21-mo call 30
    18-mo goal setting 30
    Study maintenance activities (not per person), min
    Technical support 720 1440 720 1440
    Message board and injury check 360 720 360 720
    Resending pedometers 180 360 180 360
    SMS text message support 480 960

    aNot applicable.

    Cost-Effectiveness

    Cost per increased minute of activity is shown in . For self-reported activity, each additional minute gained over 12 months in the enhanced group cost US $0.09, compared with US $0.11 per minute in the original group. Incremental costs of increased minutes in the enhanced group were US $0.05 per minute beyond those reported by the original group.

    As increases in MVPA measured by the ActiGraph were smaller, these also cost more, with each increased minute costing US $0.19 in the enhanced group and US $0.16 in the original group. As increases in the original group were larger than those in the enhanced group, ICERs could not be calculated.

    Costs for cumulative increases over the 24-month period were lower. Each self-reported increased minute cost US $0.06 in the enhanced group and US $0.05 in the original group. Incremental increases in the enhanced group beyond the original group were US $0.08 per additional minute of self-reported MVPA. ActiGraph-recorded minutes over the 24-month period were US $0.12 in the enhanced group compared to US $0.10 in the original group, with incremental minutes costing US $0.20 each in the enhanced group beyond that in the original group.

    Cost per person meeting guidelines was normalized per 100 people in each arm. This was higher in the enhanced arm at 12 months (US $705) than in the original arm (US $503). These rose to US $812 and US $601 at 24 months, respectively. ICERs at 24 months were US $1837 (95% CI US $730.89-US $2673.89) per additional person meeting guidelines in the enhanced arm beyond those in the original arm.

    Table 3. Costs of increases in physical activity.
    Original group Enhanced group
    7-d PARa ActiGraph 7-d PAR ActiGraph
    Total increase in MVPAb per person, min
    Baseline to 12 mo 1584 1038 2120 1038
    Total 24 mo 3491 1921 4935 1921
    Costs per minute increase in MVPA,US $
    Baseline to 12 mo 0.11 0.16 0.09 0.16
    Total 24 mo 0.05 0.10 0.06 0.10
    Incremental cost per minute of increase inMVPA,US$
    Baseline to 12 mo c 0.05
    Total 24 mo 0.08
    Costs per person meeting guidelines, US $
    12 mo 503 705
    24 mo 601 812
    Incremental cost per person meeting guidelines, US $
    12 mo N/Ad
    24 mo 1837 (730.89-2673.89)

    aPAR: physical activity recall.

    bMVPA: moderate-to-vigorous physical activity.

    cNot applicable.

    dNot available. As the original arm outperformed the enhanced arm in these metrics, it was not possible to calculate the incremental cost-effectiveness ratios.

    Principal Findings

    Analyses showed that the technology- and theory-enhanced PA intervention for Latinas was more costly than the original intervention but still markedly less costly than most medical interventions. The enhanced intervention cost US $300 per person for a 24-month program, or approximately US $12 per person per month. The original intervention cost US $189 per person, or approximately US $8 per person per month. The largest expense by far was personnel time, which accounted for approximately 72% and 64% of costs in the enhanced and original groups, respectively. Most of the increased cost in the enhanced intervention was attributable to additional personnel time for making additional monthly calls and providing SMS text message support. The prizes also contributed to higher costs in the enhanced group.

    Costs and activity gains were similar throughout the first year, thus cost-effectiveness was also similar, with minutes gained in the enhanced group costing US $0.09 for each participant compared to US $0.11 for each participant in the original group (US $0.19 and US $0.16 by ActiGraph, respectively). During the second year in the program, the enhanced group continued to increase their MVPA, while gains in the original group declined. However, the original group had few costs incurred in the second year, while the enhanced group continued to deliver maintenance doses of intervention. Cost-effectiveness thus remained similar over the full 24 months, with each additional minute costing just US $0.05 in the original group and US $0.06 in the enhanced group (US $0.10 and US $0.12 by ActiGraph, respectively). ICERs showed that, beyond the cost of the original intervention, each additional minute in the enhanced intervention cost just US $0.08 (US $0.20 for ActiGraph-measured minutes). While more individuals in the enhanced group met guidelines at 24 months, the cost of meeting guidelines was also higher, costing US $812 per person compared to US $601 in the original arm.

    While absolute minutes of PA varied between self-report and objective measures, as is commonly seen between these 2 measures [,], the overall pattern of results between the 2 were similar. While accelerometry is considered the gold standard, there is a large body of research showing the benefits associated with self-reported activity, which largely informed the development of national guidelines [,]. Self-reported MVPA allows for subjective interpretation of intensity, which may not align with universally applied cut points for accelerometers, particularly for participants who are overweight, obese, or inactive. Both the 7-day PAR and ActiGraph showed sustained increases in activity in the enhanced group over the 24-month study period, corroborating better maintenance of activity gains.

    Comparison With Prior Work

    These results suggest that paying for more intervention yields commensurate increases in activity. Given the enormous benefits of PA [,-], particularly if it is maintained over time [,], the additional cost of more intensive interventions that yield greater increase in PA over time is likely preferable for implementation sites when feasible. Although we could not find other studies that framed the incremental cost of an intervention in terms of additional people meeting PA guidelines, meeting PA guidelines is recognized as an important research metric []. One health economics study concluded that PA interventions that cost less than US $2900 over 2 years to help persons meet PA guidelines can be considered cost-effective []. Our finding that the enhanced group spent US $1837 per additional person meeting guidelines compared to the original group at 24 months is well below this threshold, and therefore, the intervention can be considered cost-effective.

    This amount also seems a relatively small price to pay compared to the cost of managing chronic diseases associated with inactivity. The cost of managing diabetes, for example, was US $237 billion in the United States in 2017, with insulin alone costing approximately US $5000 per user annually [,]. An analysis of health care use in Australia found that individuals meeting activity guidelines were about one-third less likely to visit the emergency room or be hospitalized, half as likely to use outpatient services, and incurred approximately Aus $1400 (US $920) less in annual health care costs []. Multiple studies have shown that PA programs are not only low cost but ultimately cost saving, saving considerably more in health care costs than the programs cost [,]. One evaluation of a PA intervention for older community-dwelling adults found that, for those not initially meeting activity guidelines, the program saved US $143 to US $164 per participant over 6 months beyond the cost of the intervention []. Paying for PA programs can therefore be seen as an investment not only in the health and well-being of the participants but also in health care, particularly for populations at high risk. Given the higher costs and higher yields of the enhanced intervention, implementation of the enhanced version may be most appropriate with clinical populations managing chronic disease. As the original intervention still yielded substantial increases in activity, it may be more appropriate for community settings focusing on prevention and overall well-being.

    Compared to the parent study, the original intervention in this study was slightly more expensive, costing US $14 per person per month at 12 months versus US $12; this was due to increased personnel costs due to wage increases []. Cost to implement the interventions will thus be largely dependent on staff salaries, which could vary broadly. Clinical sites could deliver the intervention via medical assistants or nursing staff, which would likely increase costs; conversely, using volunteers or automating components to reduce staff time could substantially lower costs.

    Limitations

    This study has several limitations. Cost and cost-effectiveness analyses were based on aggregated costs data, not individual cost data, thus results should be interpreted as overall estimates for delivering the intervention rather than individual effectiveness. The costs were also limited to a payer perspective and did not include health care or societal costs or quality-adjusted life years. However, these approaches allow for direct comparability with the previous study, which used the same analytic approach. Moreover, some costs, such as overhead for shared space, could only be estimated and would vary considerably based on the implementation site. We were unable to determine the effectiveness of each intervention component; therefore, it was not possible to determine the added cost-effectiveness of individual intervention components. Key differences between the conditions were the SMS text messaging and the additional calls and in-person visits. The cost of these components varied greatly, with SMS text messaging being almost free and highly scalable and calls and visits being costly and having limited scalability. Future trials using multiphase optimization strategy (MOST) designs should be used to identify the most effective components to optimize the cost-effectiveness of the intervention.

    Finally, the study findings may not generalize to other populations or settings. The study population was primarily of Caribbean descent and l resided in a small geographic region of the United States. The parent study was carried out with Latinas in California who were predominantly of Mexican descent and reported being markedly less active at baseline but who showed similar increases in activity. Future research with other populations and settings would elucidate how generalizable things findings are.

    The study has several strengths. Data were taken from a randomized trial with rigorous methodology, including multiple validated measures of PA. The trial focused on an underserved population considered to be at high risk and included long-term follow-up. Costs were based on current market prices, including published pay scales and benefits for staff. We were also able to directly compare costs and cost-effectiveness to those in the previous parent randomized controlled trial.

    Conclusions

    Findings highlight that additional intervention components, particularly those necessitating more staff time, yield higher intervention costs; however, they may also lead to greater increases in PA. In this study, these differences were most apparent after long-term follow-up. This could suggest that greater investment may be most appropriate in individuals who would benefit the most from long-term adherence to activity guidelines, such as those at high risk for, or managing, chronic diseases that respond to lifestyle changes. As staff time was by far the most costly component, PA interventions may become more cost-effective and more widely disseminated, as they rely more on broad reach technology. Future research should investigate whether relying fully on automated technology without face-to-face intervention components yields similar long-term effectiveness.

    This study was funded by the National Institutes of Health and National Cancer Institute (R01CA159954).

    None declared.

    Edited by N Cahill; submitted 29.Apr.2025; peer-reviewed by V Surasani, O Dimgba; comments to author 16.Jun.2025; revised version received 07.Jul.2025; accepted 29.Jul.2025; published 25.Nov.2025.

    ©Britta Larsen, Dori Pekmezi, Sheri J Hartman, Shira Dunsiger, Todd Gilmer, Erik Groessl, Bess Marcus. Originally published in the Journal of Medical Internet Research (https://www.jmir.org), 25.Nov.2025.

    This is an open-access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work, first published in the Journal of Medical Internet Research (ISSN 1438-8871), is properly cited. The complete bibliographic information, a link to the original publication on https://www.jmir.org/, as well as this copyright and license information must be included.

    Continue Reading

  • Journal of Medical Internet Research

    Journal of Medical Internet Research

    The number of older adults is growing and expected to reach 2.1 billion worldwide by 2050 []. In Canada, approximately 18.5% of the population are ≥65 years old, representing more than 7 million Canadians [], and older Canadians constitute the fastest growing age group []. As a result, challenges have emerged with addressing the increasing needs of older adults who utilize health care services more frequently than other age groups []. eHealth, which refers to the use of information and communication technologies (ICTs) for health and encompasses a variety of mobile health (mHealth) apps [,] and internet use for health services and information delivery [,], presents an opportunity to enhance care quality, equity, efficiency, and management of health conditions among older adults [-].

    In recent years, there has been a growing interest worldwide in studying technology in the context of older adults’ care [-]. With the global challenges related to the shortage of providers and the shift of care delivery outside of medical settings, older adults are expected to play a more active role in the management of their health []. In addition, as baby boomers move into the “third age” of retirement characterized by searching for new experiences and learning new things, there is an increasing need to assess their knowledge and use of information technologies [], which can support their health and well-being. This is particularly important in the context of global health care systems estimating an increase in the proportion of older adults [] and the limited resources available to support them.

    The multidisciplinary life course theory (LCT) [,] emphasizes the integrated relation between a person’s choices and their socioeconomic context, alongside their capacity to make decisions within existing opportunities and constraints []. It is a multidisciplinary framework that integrates factors from various disciplines (eg, sociology, psychology) to understand human behavior. An individual’s family constitutes a “social group” that is embedded in a larger social context [], and questions related to this factor are at the core of the LCT []. The components characterizing the LCT include geographical location, social ties (eg, presence and attributes of family members and societal experiences), stages in life (eg, generational group differences), variability (eg, in gender, social class, education, wealth, family support), and personal control (eg, environmental opportunities and constraints), all of which can present circumstances that shape individuals’ perceptions and decisions []. Along the same line, the World Health Organization (WHO) emphasizes the importance of social determinants (ie, environment in which people live, work, age, and can access money and resources), as these can create avoidable differences in health across communities []. In addition, the Andersen model for health care utilization emphasizes the importance of enabling factors (eg, access to care) and need factors (eg, health problems) in shaping the utilization of health services []. Therefore, interactions with the health system, including factors related to access to resources, services utilization, health status, and perceived needs, represent contextual factors that can influence eHealth use.

    Prior research on older adults’ use of technology for web-based socialization discussed the relevance of financial and knowledge barriers, as well as social factors related to family, social motivation, and appropriate environments [], and the need to investigate social and economic factors that persist as challenges to technology uptake []. Aside from a few studies that investigated social determinants in relation to telehealth use by athletic trainers [], eHealth engagement among people living with HIV [], eHealth literacy levels in older adults [], older adults’ perceptions and views on eHealth services [], and access to and preferences for patient portal use among older adults [], limited research exists in this area. In Canada, an earlier study with older adults focusing on health tracking behaviors showed that greater than 60% tracked their health manually, thus emphasizing the limited uptake of digital self-tracking in this group compared with the general population [] and the need to better understand their eHealth use patterns. Hong and Cho [], who reviewed instruments assessing eHealth behaviors, also called for national surveys adapted to technology development that may be leveraged to analyze eHealth behaviors for informing evidence-based policies.

    Despite increasing scholarly interest in older adults’ use of technology, few studies have provided a comprehensive, nationally representative, and theory-driven portrait of eHealth engagement in this population. Most prior work has been limited to localized samples, focused on specific technologies (eg, patient portals, telemonitoring pilots), or relied on instruments not adapted to current technological realities. This lack of evidence impairs the ability of policymakers and health system leaders to design equitable digital health strategies that address the needs of the fastest-growing group of health care users.

    Grounded in the LCT and Andersen health care utilization model, this study makes 3 important contributions. First, it provided the first national baseline of eHealth use among older Canadian adults across a broad range of applications, enabling pre- and postpandemic comparisons and international benchmarking. Second, it revealed how social determinants and health care system interactions shape digital health behaviors, offering conceptual insights into the factors that enable or constrain digital engagement in later life. Third, it highlighted equity-relevant gaps, identifying groups at heightened risk of digital exclusion and pointing to where targeted interventions are most urgently needed.

    Study Design

    A cross-sectional survey of Canadian older adults assessing their use of specific technologies or ICTs, eHealth, and health IT solutions was conducted online using a self-administered computer-assisted web interface (web surveys: 1500/2000, 75%) and by phone using computer-assisted telephone interviews (phone surveys: 500/2000, 25%).

    Settings and Participants

    A random national sample of 2000 Canadian residents from all provinces, aged 65 years and older, and who spoke English or French was selected from a proprietary online panel of more than 400,000 households owned by Léger, Canada’s largest and leading research and analytics firm. To ensure a random sample, no quotas were set initially, and the data were weighted after sampling according to gender, age, and region to maximize the representativeness of the Canadian population of older adults.

    Assessments and Data Sources

    In line with the conceptual framework shown in , the survey instrument included 3 main sections: (1) sociodemographic characteristics, living environment, and health system interaction factors; (2) eHealth and ICT use; and (3) fall detection technology (FDT) and telemonitoring technology (TM) use.

    Figure 1. Conceptual framework of social and system-level determinants shaping older adults’ use of eHealth applications.

    Grounded in the principles of the LCT, sociodemographic variables were measured with standardized indicators including gender, age, marital status, income, education, language, and employment. Questions assessing the living environment included the region (representing provinces), community (rural/suburb/urban/metropolitan), place of residence (home/retirement home/long-term care), and living arrangement (alone/with family/with spouse or partner) to give a comprehensive overview of the older adult social context.

    To measure interactions with the health care system, we used the following indicators: private insurance (yes/no), family physician (yes/no), home care (yes/no), willingness to pay for quicker access to services (yes/no), hospitalizations and emergency room visits in the past 6 months (yes/no), and perception of own health (categorical). Satisfaction with the health care system, with access to health care services, and with the health care services received were also included in the survey (categorical).

    The outcomes of interest in this study were divided into 3 categories that represent eHealth use: (1) digital health engagement, including the extent of internet use for actively searching and accessing health-related information and services online (eg, searching for online information about a health condition, accessing laboratory test results); (2) digital health communication, including the extent of willingness or interest in using digital means to communicate about health services (eg, use of email to discuss a health condition with a physician, obtain information about trusted websites); and (3) mobile health technology use, including the extent of use of mobile apps, FDTs, and TMs. The questions assessing FDT and TM use were binary (yes/no). The frequencies of use of mobile apps for health and internet for accessing information, resources, and communicating with providers were measured on a 5-point scale (1=Never to 5=Always). Interest or willingness to exchange online information was also assessed on a 5-point scale (1=Not at all to 5=Totally).

    The survey instrument was pretested with 47 respondents by phone and on the web. A change was subsequently made to the skip pattern for one of the questions. Data collection was completed over a 3-week period in 2018. Although the data are not very recent, they provide a robust baseline for pre- and post–COVID-19 pandemic comparisons, especially in the absence of data on eHealth use among older adults.

    Data Analysis

    Descriptive data analysis was conducted to gain an understanding of the profile of older adults and their technology-related behaviors. Bivariate nonparametric tests were used for analyses of associations between continuous dependent variables (eg, search online for information) and categorical independent variables (eg, age, education). Specifically, nonparametric 2-independent samples analyses (Mann-Whitney) were conducted for independent variables with only 2 categories, and nonparametric K-independent samples analyses (Kruskal-Wallis) were conducted for independent variables with more than 2 categories. χ2 and Fischer’s exact tests for categorical variables were used to examine the associations between the binary dependent variables related to mobile technology use and the determinants (eg, sociodemographic characteristics, living environment) and interactions with the health system variables. Multivariate regression analysis for eHealth scale questions and binary logistic regression analysis for binary mHealth technology questions (TM and FDT) were performed to examine the significant relationships between eHealth use and the social and health system interaction variables that showed significant associations at the bivariate analysis level. To assess potential multicollinearity among the independent variables for the multivariate analysis, we calculated the variance inflation factor (VIF) for each predictor. All independent variables, with the exception of marital status (ie, “married”; VIF~10), had a VIF lower than the general threshold of 10 []. Therefore, we retained all theoretically relevant variables to maintain the breadth of the models, which was the main aim with our modeling approach.

    Ethical Considerations

    Ethics approval was granted by the University of Ottawa Ethics Research Board, and all data were anonymized. Eligible respondents were provided with an information letter that explained the scope and purpose of the project. It specified that their participation was voluntary and completing the questionnaire indicated their consent for participating in this study. Léger panelists are rewarded for their participation over time using a series of financial incentives that can be accumulated and cashed out or donated for a charitable cause. Participants in this survey who completed this survey were rewarded 1200 Léger Opinion points (CAD $20=20,000 points; a currency exchange rate of CAD $1=US $0.71 is applicable) that can be redeemed through their preferred reward method.

    Sample Characteristics

    reveals the variations in social determinants and living conditions. The majority of respondents were 65 years to 69 years old (663/2000, 33.2%), were women (1092/2000, 54.6%), were married (1201/2000, 60.1%), had a college or university degree (1243/2000, 62.2%), spoke English at home (1496/2000, 74.8%), earned less than CAD $75,000 (1184/2000, 59.2%), and lived in Ontario (760/2000, 38%) or Quebec (505/2000, 25%), which are the most populous and largest health care jurisdictions in the country. In addition, 68.5% (1369/2000) lived in metropolitan cities or suburbs, and 96.1% (1922/2000) lived in their own homes or apartments; one-third (665/2000, 33.3%) lived alone.

    Table 1. Study sample sociodemographic characteristics and interaction patterns with the health care system (n=2000).
    Characteristics and interaction patterns Results
    Gender, n (%)
    Male 908 (45.4)
    Female 1092 (54.6)
    Age (years), n (%)
    65-69 663 (33.2)
    70-74 479 (23.9)
    75-79 360 (18)
    80-84 323 (16.2)
    85 175 (8.7)
    Highest education level, n (%)
    Elementary school 42 (2.1)
    Intermediate school 56 (2.8)
    High school 630 (31.5)
    College degree 523 (26.2)
    University, undergraduate 476 (23.8)
    University, graduate degree 244 (12.2)
    Other 28 (1.4)
    Marital status, n (%)
    Single 141 (7)
    Married 1201 (60.1)
    Widowed 401 (20.1)
    Separated/divorced 248 (12.4)
    Other 9 (0.4)
    Income (CAD $), n (%)
    <25,000 202 (10.1)
    25,000-49,999 589 (29.4)
    50,000-74,999 393 (19.6)
    75,000-99,999 270 (13.5)
    100,000-124,999 122 (6.1)
    ≥125,000 61 (3.1)
    I prefer not to answer 363 (18.2)
    Employment, n (%)
    Employed, full-time 77 (3.8)
    Employed, part-time 94 (4.7)
    Retired 1790 (89.5)
    Other 39 (1.9)
    Home language, n (%)
    French 459 (23)
    English 1496 (74.8)
    Other 45 (2.2)
    Region, n (%)
    Prairies (Alberta, Saskatchewan, Manitoba) 293 (14.7)
    British Columbia 287 (14.3)
    Maritimes (New Brunswick, Nova Scotia, Prince Edward Island) 126 (6.3)
    Newfoundland and Labrador 30 (1.5)
    Ontario 760 (38)
    Quebec 505 (25.2)
    Residential community, n (%)
    Rural (<2500 persons) 306 (15.3)
    Small town (2500‐10,000 persons) 306 (15.3)
    Suburb (10,000‐50,000 persons) 479 (23.9)
    Metropolitan (>50,000 persons) 890 (44.5)
    I don’t know 19 (1)
    Residence type, n (%)
    My home or apartment 1922 (96.1)
    A retirement home 76 (3.8)
    Other 2 (0.1)
    Living situation, n (%)
    Alone 665 (33.3)
    With your wife/husband/partner 1205 (60.3)
    With family or friends 119 (5.9)
    Other 11 (0.5)
    Private insurance, n (%)
    Yes 1098 (54.9)
    No 902 (45.1)
    Family physician, n (%)
    Yes 1896 (94.8)
    No 104 (5.2)
    Home care, n (%)
    Yes 83 (4.1)
    No 1917 (95.9)
    Willingness to pay for quicker access, n (%)
    Yes 647 (32.4)
    No 1353 (67.6)
    Perception of own health, n (%)
    Excellent 242 (12.1)
    Very good 694 (34.7)
    Good 725 (36.2)
    Fair 281 (14)
    Poor 59 (2.9)
    Hospitalization (past 6 months), n (%)
    Yes 164 (8.2)
    No 1836 (91.8)
    Number of hospitalizations (past 6 months), median (range; IQR) 1 (1-6; 1-1)
    Emergency room visits (past 6 months), n (%)
    Yes 278 (13.9)
    No 1717 (85.9)
    I don’t know 5 (0.2)
    Number of emergency visits (past 6 months), median (range; IQR) 1 (1-8; 1-2)

    aA currency exchange rate of CAD $1=US $0.71 is applicable.

    bOf 164 older adults who reported being hospitalized.

    cOf 278 older adults who reported emergency room visits.

    Health Care System Interaction

    Of the repondents, who normally have access to a national health insurance, 54.9% (1098/2000) reported having private insurance (). The majority had a regular family physician (1896/2000, 94.8%), did not receive home care services (1917/2000, 95.9%), and reported good to excellent health (1661/2000, 83.1%). In addition, 8.2% (164/2000) and 13.9% (278/2000) had hospitalizations and emergency visits, respectively, in the past 6 months, and one-third (647/2000, 32.4%) were willing to pay out of pocket for quicker access to services. Overall, 61.8% (1237/2000) reported being diagnosed with one or more chronic conditions, and 13.7% (275/2000) indicated having fallen in the past 6 months (falls: median 1, IQR).

    Overall, the satisfaction of respondents with the health care system was high (very satisfied: 366/2000, 18.3%; satisfied: 1086/2000, 54.3%). In addition, 79.1% (1583/2000) indicated good to high satisfaction with access to health care services, and 84.4% (1689/2000) were satisfied to very satisfied with the health care services received over the past 2 years.

    ICT and eHealth

    The vast majority of surveyed older adults owned a computer (1703/2000, 85.2%), 57.7% (1153/2000) reported having a tablet or iPad, and 53.9% (1077/2000) reported having a smartphone (). Fewer owned wearables or mobile devices (238/2000, 11.9%). In addition, 90.3% (1654/2000) reported using email, 50.1% (917/2000) used phone text messaging (eg, WhatsApp, Messenger), and 53.6% (983/2000) used Facebook. Except for a small percent of respondents (238/2000, 11.9%), participants in our study confirmed having used the internet over the past 6 months, mostly daily (1472/2000, 83.6%) or a few times a week (180/2000, 10.2%).

    Despite frequent internet and email use and high prevalence of ICTs, the use of internet to connect with a health care professional, access test results or patient portal, or book a medical appointment was limited, although moderate use was reported for using the internet to search for online information about health conditions (median 3, IQR 2-3 on the 5-point Likert scale; ). The respondents expressed low willingness to use email to exchange information about their health with their health care professionals (median 2, IQR 1-3 on the 5-point Likert scale) and a moderate interest in obtaining information about trusted websites relevant to their health condition and accessing their medical records (median 3, IQR 1-5 on the 5-point Likert scale).

    The prevalence of use of TMs and FDTs was low (189/2000, 9.4% and 84/2000, 4.2%, respectively), despite familiarity with these technologies. Among the respondents who had previously or were currently using TMs and FDTs, 81% (153/189) and 86% (72/84), respectively, indicated their willingness to use these technologies again in the future, which indicates satisfaction with these technologies.

    Social Determinants, Health Care System Interaction, and eHealth Use

    Bivariate analyses showed significant associations (P<.05) between sociodemographic and living environment characteristics and the majority of questions assessing eHealth use (). For example, being a man, in the younger age group (ie, 65‐69 years), and holding a graduate university degree were significantly associated with a higher willingness or interest in using the internet to access medical records, exchange medical information with family physicians or health care providers, and obtain information on trusted websites to consult about one’s condition (all P<.001). On the other hand, being a woman, being separated or divorced, and having a lower income were significantly associated with higher frequency of internet use to search for online information about health problems (all P<.001) and the tendency to self-diagnose (P<.001, P=.02, and P=.03, respectively). Living in metropolitan areas and in one’s own home were significantly associated with higher frequency of using mobile apps for health (P=.03) and TM use (P=.04). Being married or having a partner was also significantly associated with using mobile apps for health (P=.01) and FDT use (P=.01).

    Table 2. Bivariate associations between social determinants and eHealth use among older adults in the study sample (n=2000), with P<.05 considered statistically significant.
    eHealth use Sex Age Education Marital status Employment Income Region Language Community Live in… Live with…
    Internet use to…, on a scale from 1 to 5 (1=Never; 5=Always), P value
     Search online for information <.001 .13 <.001 <.001 .66 <.001 .03 .14 .001 .46 .19
     Self-diagnose <.001 .001 .03 .02 .049 .03 .07 .001 .009 .83 .49
     Access lab results .40 .20 .11 <.001 .045 <.001 <.001 .003 <.001 .20 <.001
    .24 .051 .68 .009 .20 .34 <.001 .16 .03 .83 .005
    .002 .15 <.001 .02 .69 .004 .001 .20 <.001 .20 .004
     Participate in discussion forums .44 .01 .66 .84 .20 .39 .36 .005 .67 .31 .70
    Willingness/interest in…, on a scale from 1 to 5 (1=Not at all; 5=Totally), P value
     Use email to discuss health <.001 <.001 <.001 <.001 .04 <.001 <.001 .04 .19 <.001 <.001
     Obtaining information on trusted websites <.001 <.001 <.001 <.001 <.001 <.001 <.001 .39 <.001 <.001 <.001
     Accessing online medical records <.001 <.001 <.001 <.001 .04 <.001 <.001 .033 <.001 <.001 <.001
    Use of…, on a scale from 1 to 5 (1=Not at all; 5=Totally), P value
     Mobile apps ≥.99 .007 .11 .01 .74 .002 .007 .16 <.001 .21 .03
    Use of… (yes/no), P value
     Wearables ≥.99 .95 .87 .35 .89 ≥.99 .73 .66 .64 .82 .10
     TM .25 .19 .46 .76 .91 .59 .63 .03 .03 .04 .63
     FDT .11 .01 .55 .01 .95 .52 .99 .54 .67 .004 .03

    aEMR: electronic medical record.

    bTM: telemonitoring technologies.

    cFDT: fall detection technologies.

    When examining the relationship between interactions with the health care system and eHealth use (), having private insurance and a willingness to pay out of pocket for quicker access to health care services were significantly associated with most outcome variables (eg, most P≤.001). Respondents who indicated not receiving home care services and no hospitalizations nor emergency visits reported more FDT use (P<.001, P=.002, and P=.002, respectively). Those having an excellent perception of health reported more use of mobile apps for health, higher frequency of internet use to participate in discussion forums about their health, and more willingness or interest in using email to exchange medical information with their physician about their condition.

    Table 3. Bivariate associations between health care system–related variables and eHealth use among older adults in the study sample (n=2000), with P<.05 considered statistically significant.
    eHealth use Family physician Private insurance Home care services Pay for quicker access Perception of health Hospitalizations Emergency visits
    Internet use to…, on a scale from 1 to 5 (1=Never; 5=Always), P value
     Search online for information .17 .001 .01 <.001 .43 .56 .004
     Self-diagnose .09 .20 .02 <.001 .29 .60 .80
     Ask health care professional .18 .04 .19 <.001 .35 .67 .58
    .04 .008 .77 <.001 .005 .002 .20
     Access patient portal/EMR .02 .02 .54 <.001 .27 .002 .04
     Book appointment .005 .004 .63 .001 .92 .12 .07
    .90 .25 .66 .02 .03 .78 .76
    Willingness/interest in…, on a scale from 1 to 5 (1=Not at all; 5=Totally), P value
    .67 .001 .002 <.001 .04 .16 .87
    .26 <.001 .001 <.001 .94 .046 .46
    .09 <.001 .002 <.001 .48 .12 .98
    Use of…, on a scale from 1 to 5 (1=Never; 5=Always), P value
     Mobile apps for health .03 <.001 .85 <.001 .03 .40 .44
    Use of… (yes/no), P value
     Wearables .54 .48 .09 .81 .52 ≥.99 ≥.99
     TM ≥.99 .06 .13 .27 .33 ≥.99 .78
     FDT .58 .82 <.001 .81 07 .002 .002

    aEMR: electronic medical record.

    bTM: telemonitoring technologies.

    cFDT: fall detection technologies.

    Multivariate Analysis

    Multivariate analyses examined the relationship between eHealth use and the sociodemographic characteristics and interactions with the health care system variables that were significant at the bivariate analysis level while controlling for other variables. presents which relationships were significant (P<.05) in the multivariate analyses; the detailed results (standardized coefficients, odds ratios [ORs], confidence intervals, and P values) are presented in .

    Table 4. Multivariate analysis of significant bivariate associations between social determinants and eHealth use among older adults in the study sample.
    eHealth use Sex Age Education Marital status Employment Income Region Language Community Live in… Live with…
    Internet use to…, on a scale from 1 to 5 (1=Never; 5=Always)
     Search online for information
     Self-diagnose
     Ask health care professional
     Access lab results
     Access patient portal/EMR
     Book appointment
     Participate in discussion forums
    Willingness/interest in…, on a scale from 1 to 5 (1=Not at all; 5=Totally)
     Use email to discuss health
     Obtaining information on trusted websites
     Accessing online medical records
    Use of…, on a scale from 1 to 5 (1=Never; 5=Always)
     Mobile apps for health
    Use of… (yes/no)
     TM
     FDT

    aLinear regression analyses.

    bSignificant association (P<.05).

    cEMR: electronic medical record.

    dLogistic regression analyses.

    eTM: telemonitoring technologies.

    fFDT: fall detection technologies.

    eHealth use (ie, digital health engagement and digital health communication) was significantly associated with several sociodemographic variables (). Women reported more internet use to search for online information about health problems (β=0.148, P<.001) and self-diagnosis (β=0.079, P=.001), whereas older age (≥85 years) was consistently associated with lower frequency of eHealth use (internet use for self-diagnosing, interest in obtaining information about trusted websites for their health condition, accessing online medical records, and use of mobile apps for health). Interestingly, respondents with a lower income indicated a higher frequency of searching for online information about health conditions or problems (β=–0.122 for those earning between CAD $50,000 and CAD $75,000 compared with those earning less than CAD $25,000; P=.007), and English-speaking respondents (compared with their French-speaking counterparts) had a higher frequency of self-diagnosing (β=0.098, P<.001) and participating in online forums to discuss aspects related to their health (β=0.051, P=.04).

    Variation in eHealth use was significantly associated with the region and community of residence. Compared with rural areas, living in the suburbs or metropolitan areas was consistently associated with a higher frequency of using mobile apps for health (β=0.135, P<.001 for metropolitan areas) and internet use for looking for information about health conditions (β=0.130, P<.001 for suburban areas), self-diagnosing (β=0.114, P<.001 for suburban areas), accessing laboratory results (suburban areas: β=0.092, P=.006; metropolitan areas: β=0.077, P=.03), accessing patient portals (suburban areas: β=0.088, P=.01; metropolitan areas: β=0.077, P=.35), and booking appointments online (β=0.105, P=.005 for metropolitan areas). Residing in retirement homes as opposed to one’s own home was significantly associated with more FDT use (OR=0.366, 95% CI 0.145‐0.923).

    There were considerable differences across provinces in Canada, which may be attributed to systemic variation in availability of digital health services leading to variable access and eHealth use. When compared with persons from central Canada (ie, Prairies), Canadians residing in British Columbia reported a higher frequency of using the internet to ask health care professionals about their health (β=0.062, P=.04) and mobile apps for health (β=0.064, P=.04) and more willingness or interest in using email to exchange information about their health condition with a physician (β=0.087, P=.002) and access their online medical records (β=0.083, P=.002). Residents of Ontario also reported a higher frequency of using the internet to search for health information about their conditions (β=0.075, P=.03) and access laboratory results (β=0.297, P<.001) and patient portals (β=0.132, P<.001), whereas respondents from the Maritimes provinces were more interested or willing to use the internet to email their physicians about their health condition (β=0.093, P<.001), obtain information on trusted websites to consult about their conditions (β=0.062, P=.01), and access their online medical records (β=0.089, P<.001). Current use of the internet to access patient portals (β=0.082, P=.01) and interest in accessing online medical records (β=0.120, P=.005) were also higher for older adults in Quebec compared with residents of central Canada.

    The enabling and need factors related to the interactions with the health care system investigated in this study revealed a pattern of association with eHealth use. Among the enabling factors, willingness to pay out of pocket for quicker access to health care services and having private insurance were consistently and significantly related to a higher frequency of eHealth use. Specifically, willingness to pay out of pocket was significantly associated with higher frequency of eHealth use for all the measures, with the exception of mobile app and FDT use (). Respondents who did not have private insurance reported lower frequency for searching for information about their health problem or condition (β=–0.054, P=.03) and online access to laboratory results (β=–0.055, P=.02) and patient portals (β=–0.063, P=.009). With regard to the need variables, older adults who did not have emergency visits in the past 6 months (ie, less needs) reported a significantly lower frequency of searching for information about their health problem or condition (β=–0.086, P<.001) and accessing patient portals (β=–0.058, P=.02) but more FDT use (OR=2.16, 95% CI 1.228‐3.800). However, those who did not receive home care services indicated a higher frequency of searching for information about their health problem or condition (β=0.052, P=.03) and more FDT use (OR=3.427, 95% CI 1.550‐7.596) compared with those receiving home services. Last, a high perceived health status (as excellent) was significantly associated with more frequent mobile app use as opposed to a fair (β=–0.073, P=.02) or good (β=–0.074, P=.049) perception of health.

    Table 5. Multivariate analysis of significant bivariate associations between health care system interaction variables and eHealth use among older adults in the study sample.
    eHealth use Family physician Private insurance Home care services Pay for quicker access Perception of health Emergency visits Hospitalizations
    Internet use to…, on a scale from 1 to 5 (1=Never; 5=Always)
     Search online for information
     Self-diagnose
     Ask health care professional
     Access lab results
     Access patient portal/EMR
     Book appointment
     Participate in discussion forums
    Willingness/interest in…, on a scale from 1 to 5 (1=Not at all; 5=Totally)
     Use email to discuss health
     Obtaining information on trusted websites
     Accessing online medical records
    Use of…, on a scale from 1 to 5 (1=Never; 5=Always)
     Mobile apps for health
    Use of… (yes/no)
     TM
     FDT

    aLinear regression analyses.

    bSignificant association (P<.05).

    cEMR: electronic medical record.

    dLogistic regression analyses.

    eTM: telemonitoring technologies.

    fFDT: fall detection technologies.

    Principal Findings

    This national survey provides the first comprehensive portrait of eHealth use among older Canadians, offering a valuable baseline for ongoing monitoring and international comparisons. Although the analyses incorporated a broad range of social and health system determinants, our goal was to capture the multidimensional nature of digital engagement and application use in later life, which sets the stage for future specific and targeted investigations. To enhance clarity, the Discussion section highlights the most important findings and their implications.

    First, although the vast majority of older adults owned digital devices and reported frequent internet use, their actual engagement with eHealth tools remained limited. Use of TMs, FDTs, patient portals, and online appointment systems was low across the sample. This disconnect suggests that access alone is insufficient; awareness, perceived usefulness, and provider support are critical enablers. Importantly, those who had used TMs and FDTs expressed high willingness to use them again, underscoring the importance of initial exposure and positive experiences.

    Second, consistent with prior literature, eHealth use was stratified by sociodemographic advantage. Younger age, higher income, and residence in metropolitan areas were associated with greater engagement, while older age, lower income, rural residence, and institutional living environments were linked to reduced use. These patterns reveal persistent inequities within the older adult population, even in a context of high technological readiness. Targeted efforts are needed to ensure that those with the greatest health needs and fewest resources are not left behind.

    Third, interactions with the health care system emerged as powerful predictors of eHealth use. Having private insurance, willingness to pay for quicker access, and better perceived health were all associated with higher digital engagement. Conversely, those who had received home care services or experienced recent emergency visits were less likely to use digital tools. This pattern indicates that eHealth may currently serve those with fewer immediate health needs and greater resources, rather than those most in need of coordinated care. Integrating digital health into routine pathways, rather than leaving it as an “optional extra,” is essential for equity.

    Fourth, women reported greater use of the internet for health information and self-diagnosis. Although our data do not allow us to assess outcomes such as health anxiety, prior research has suggested that extensive online searching can sometimes heighten distress (ie, “cyberchondria”) [,]. This represents a possible area for future investigation.

    Several additional associations were observed, including the roles of education, language, and provincial differences, that further illuminate the diversity of older adults’ digital health engagement, which was quite variable across the country. For example, residents of Ontario and British Columbia reported higher use of patient portals and access to laboratory results, while those in the Maritimes expressed greater willingness to communicate with providers online. These contextual nuances reinforce the importance of tailoring digital health strategies to local and cultural environments.

    Comparison With Studies in Other Jurisdictions

    Our findings contribute to a growing body of international literature on digital health use among older adults and offer several contrasts with studies conducted in the United States and Europe.

    First, older Canadian adults in our sample reported higher levels of interest and perceived usefulness of digital health tools across age groups, including those older than 75 years. This contrasts with US and European studies that consistently report lower adoption rates among the oldest and most socioeconomically disadvantaged groups [-]. Second, although previous studies often focus on access and skill gaps, the so-called first- and second-level digital divides, our findings highlight a third dimension: motivational readiness. Many respondents expressed a willingness to use digital tools despite limited experience, particularly when they felt supported by the health care system. This dimension was less frequently emphasized in prior large-scale survey studies []. Third, the Canadian context appears to moderate some of the demographic divides found elsewhere. For example, racial and ethnic disparities in digital health use reported in the United States [] were not observed in our sample, potentially reflecting Canada’s publicly funded and more equity-oriented health care system. Finally, our data indicate relatively high willingness to reuse certain digital health technologies, particularly TMs and various mobile apps. This is an encouraging finding given the infrastructural and digital literacy barriers reported in many European contexts [,] and suggests that, when appropriately introduced and supported, older adults can develop positive experiences with digital health tools, even in rural areas.

    Taken together, our findings suggest that older Canadian adults are not uniformly resistant to digital health but rather face a complex set of motivational, attitudinal, and structural barriers. Understanding these factors in light of international comparisons can inform more targeted, inclusive, and equity-driven eHealth policies.

    Study Implications and Avenues for Future Research

    The findings of this national survey of older Canadians have several implications for policy, practice, and future research on digital health equity. First, the results underscore the need to move beyond binary notions of access (eg, having internet or not) to understand the multidimensional nature of digital engagement. Although structural factors such as age, education, and income remain important, our findings reveal that attitudinal variables such as perceived usefulness, trust in digital tools, and self-efficacy are equally, if not more, influential in predicting eHealth adoption. This suggests that interventions should not only address material barriers but also focus on building digital confidence and relevance in health contexts.

    Second, the relatively high levels of expressed interest in eHealth technologies among Canadian older adults, even those with limited prior experience, challenge persistent narratives of older adults as digitally disengaged or resistant. This observation contrasts with trends reported in European contexts [,], where digital disengagement remains widespread, particularly among the oldest-old, women, and rural residents. Similarly, in the United States, studies by James et al [] and Schuster et al [] identified persistent digital divides by race and mental health status. In contrast, our findings point to a more complex and optimistic outlook in Canada, where universal health care, relatively equitable access to health services, and targeted digital health initiatives may be shaping more inclusive digital health trajectories.

    Third, the Canadian context provides a unique lens to understand how publicly funded health care systems may buffer against some of the exclusionary forces observed in more market-based systems. For instance, unlike in the United States, where the use of digital health tools is often mediated by private insurance coverage or provider-specific platforms, Canadian respondents interact with a publicly funded system that, in principle, offers more universal access to tools such as e-prescriptions and teleconsultation portals. However, our findings indicate that actual use of digital services like these remains limited, suggesting that accessibility alone does not ensure engagement and that awareness, support, and perceived usefulness remain critical enablers.

    Based on these findings, several avenues for future research emerge. Longitudinal studies are needed to assess how digital engagement evolves over time among older adults, especially as younger cohorts with greater baseline digital skills age into retirement. Moreover, future research should examine how relational dimensions, such as the support of health care professionals, caregivers, and peers, mediate eHealth use among older adults. Comparative studies across health systems and sociopolitical contexts would further elucidate the interplay of systemic and individual-level factors in shaping digital engagement. Building on these findings, targeted investigation of specific eHealth applications using mixed methods and qualitative approaches would provide a more in-depth understanding of the interaction of factors influencing their use. Finally, there is a need to develop and evaluate intervention strategies that go beyond digital literacy training. These should include motivational and psychosocial components, co-designed with older adults, to enhance perceived value and usability of digital tools in real-life care scenarios. Integrating such approaches into existing health care pathways can ensure that digital transformation in health systems is truly inclusive, responsive, and sustainable for an aging society.

    Study Limitations

    It is important to note some limitations associated with this research. The cross-sectional nature of this study precludes a thorough assessment of causal relationships between the social determinants and variables related to the interactions with the health care system in relation to eHealth use. For example, the odds of using FDTs were considerably higher among older adults who did not receive home care services nor were hospitalized in the past 6 months. However, it was not possible to determine whether FDT use precluded the need for home care and prevented hospitalizations or whether FDT was used due to the absence of home services and better health.

    The respondents’ profile points to a relatively high level of education (62% had a college degree or higher) with the majority residing in their own homes and not alone, living in suburbs or metropolitan areas, having a regular family physician, and having private health insurance (ie, generally good access to a broad range of health care services despite limited use of home care services). Thus, we may expect a lower level of eHealth penetration among the broader older adult population, which further underscores the current suboptimal benefits that older adults are gaining from these technologies.

    Since the data were collected from a single country, the generalizability of the results is limited unless the survey is replicated in other contexts. The online nature of the survey, although complemented with phone surveys, may still have excluded potential respondents who did not have access to the internet or phone calls. In addition, the closed-ended questions included in the survey did not allow us to fully uncover the reasons behind some of the association patterns that were observed.

    When assessing multicollinearity, marital status (ie, “married”) had a VIF close to 10, indicating potential multicollinearity with other predictors in the models. This is expected, as marital status is closely associated with other sociodemographic variables like cohabitation. Although this multicollinearity may affect the precision of the estimated effect of marital status (ie, compresses the ß coefficient and makes it more likely to find false negatives or more conservative results), it does not bias the model overall.

    Last, we must stress that there are persistent challenges with collecting comprehensive data from older adult populations [,,]. Although the data from this study are not very recent, they provide a baseline for future studies assessing changes in eHealth use among older adults post-COVID-19 pandemic. In addition, since the eHealth construct’s evolution is gradual and eHealth use among Canadian citizens in general is reported to be changing slowly [], the findings continue to be relevant and present a foundation for longitudinal comparisons.

    Conclusion

    This study offers the first comprehensive national assessment of eHealth use among older Canadian adults and provides a valuable baseline for ongoing monitoring and international benchmarking, including pre- and postpandemic comparisons. Our findings highlight the importance of accounting for social determinants and interactions with the health care system when investigating eHealth use in this population.

    Our findings are also relevant beyond Canada. They demonstrate how universal health care systems mitigate, but do not eliminate, digital divides, offering lessons for other jurisdictions seeking to advance inclusive digital health strategies. The study reveals a new dimension of the digital divide, namely motivational readiness, which complements traditional access- and skill-based divides. Recognizing this attitudinal and relational component shifts the focus of interventions from infrastructure alone to trust-building, perceived usefulness, and provider support, thereby broadening the policy tool kit for promoting digital equity in aging societies.

    We would like to thank Hamidreza Kavandi and Danielle Cruise for their support in this project.

    This research project was supported by a Social Sciences and Humanities Research Council of Canada grant (#435-2017-1399). The funder was not involved in the study design, data collection, analysis, interpretation, or writing of the manuscript.

    The data gathered during this study are not publicly available and cannot be shared due to confidentiality and original ethics approval restrictions.

    All authors contributed to the conception, design/methodology, analysis/results interpretation, development, revision, and final approval of the paper.

    None declared.

    Edited by Amaryllis Mavragani, Taiane de Azevedo Cardoso; submitted 06.Feb.2025; peer-reviewed by Enno van der Velde, Ranganathan Chandrasekaran; final revised version received 26.Sep.2025; accepted 20.Oct.2025; published 25.Nov.2025.

    © Mirou Jaana, Haitham Tamim, Guy Paré. Originally published in the Journal of Medical Internet Research (https://www.jmir.org), 25.Nov.2025.

    This is an open-access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work, first published in the Journal of Medical Internet Research (ISSN 1438-8871), is properly cited. The complete bibliographic information, a link to the original publication on https://www.jmir.org/, as well as this copyright and license information must be included.

    Continue Reading