Abstract
Objective
This review aims to systematically map and categorize the current state of wearable applications among oncology patients and to identify determinants impeding clinical implementation.
Methods
A Medline, Embase and clinicaltrials.gov search identified journal articles, conference abstracts, letters, reports, dissertations and registered studies on the use of wearables in patients with malignancies published up to 10 November 2021.
Results
Of 2509 records identified, 112 met the eligibility criteria. Of these, 9.8% (11/112) were RCTs and 47.3% (53/112) of publications were observational. Wearables were investigated pre-treatment (2.7%; 3/112), during treatment (34.8%; 39/112), post-treatment (17.9%; 20/112), in survivors (27.7%; 31/112) and in non-specified or multiple treatment phases (17.0%; 19/112). Medical-grade wearables were applied in 22.3% (25/112) of publications. Primary objectives ranged from technical feasibility (8.0%; 9/112), user feasibility (42.9%; 48/112) and correlational analysis (40.2%; 45/112) to outcome change analysis (8.9%; 10/112). Outcome change was mostly investigated regarding physical activity improvement (80.0%; 8/10). Most publications (42.9%; 48/112) and registered studies (39.3%; 24/61) featured multiple cancer types, with breast cancer as the most prevalent specific type (22.3% in publications, 16.4% in registered studies).
Conclusions
Most studies among oncology patients using wearables are focused on assessing the user feasibility of consumer-grade wearables, whereas rates of RCTs assessing clinical efficacy are low. Substantial improvements in clinically relevant endpoints by the use of wearables, such as morbidity and mortality are yet to be demonstrated.
Keywords: Wearables, oncology, remote patient monitoring, cancer, digital health
Introduction
An aging population with increasing cancer prevalence poses significant challenges to healthcare systems worldwide. 1 Personalized pre-treatment assessment becomes paramount due to diverse treatment options, including CAR-T cell therapy. During cancer treatment, patients may encounter complications like infections, frailty, cardiac disorders and treatment-specific side effects, for example, cytokine release syndrome.2–4 Decreased physical activity (PA), fatigue syndrome and mental health disorders are commonly observed during follow-up of cancer patients.5,6 Early detection of health deterioration could reduce rates of hospitalization or emergency visits, as shown in other medical fields.7–9 Moreover, delayed diagnosis and treatment of complications are associated with higher morbidity and mortality. 10 As outpatient treatment becomes more prevalent, disease monitoring largely depends on patient self-management and regular clinic visits, posing challenges to patients and healthcare professionals (HCP) and underscoring the need for effective monitoring techniques. 10
Wearable devices equipped with non-invasive sensors offer continuous health data monitoring under daily life conditions. These “wearables” utilize well-established sensor technologies, such as accelerometry, temperature probes and photoplethysmography, enabling subsequent or real-time data acquisition. Generating and processing vast amounts of “big data” is challenging yet offers vast opportunities in healthcare. It enables the creation of a “digital twin”—a virtual patient representation that helps anticipate clinical events and guide personalized treatment decisions by monitoring changes in real-time parameter. 11
Wearables have been successfully applied to early detect COVID-19, 12 heart failure, 13 Alzheimer’s disease 14 and seizures in patients with epilepsy, 15 as well as health deterioration or adverse events (AE) of certain therapies. 16 Additionally, wearables can objectify patient-reported outcomes (PRO), otherwise prone to subjectivity and highly depend on patient adherence, such as quality of life (QoL) 17 and pain. 18 Furthermore, wearables can serve as interventional tools to enhance PA,19–23 thereby improving overall well-being.20,23
Despite their potential across diverse applications, wearables are rarely implemented clinically in oncology. This scoping review aims to explore the current state of wearable application among oncology patients, focusing on determinants impeding clinical implementation by evaluating and extracting information from published clinical studies.
Methods
For this scoping review, the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA-ScR) were used. 24 The final protocol was registered with the Open Science Framework on 16 November 2021 (https://osf.io/bgmya). 25
We included publications focusing on wearable application in adult patients with a confirmed oncologic disease. The term “wearable” is defined as a mobile hardware device with integrated software that collects clinical data via non-invasive sensors by measuring different parameters. Medline, Embase and clinicaltrials.gov were searched up to 10 November 2021 for full-text journal articles, letters, reports, dissertations, conference abstracts and trial registrations in any language (with keywords in English) with no limitation in the date of publication. The search strategies employed are described in Supplemental Tables 1 and 2. Keywords for the search on clinicaltrials.gov were “wearable” and “oncology.”
Three authors (KC, MV and MJ) independently assessed the eligibility of records through title and abstract screening, followed by full-text screening. Disagreements were settled by a fourth author (JB) through consensus. Data extraction was performed by the first author (KC) and revised by a second author (MJ or MV), using a standardized form (Supplemental Table 3).
For quality assessment, full-text records’ adherence to research guidelines was evaluated using aspects of STROBE, CONSORT and TREND, which we merged into a shorter checklist (Supplemental Table 4). A three-point grading system was used to categorize guideline adherence as “full,” “partly” or “none.” Due to a heterogeneous nomenclature of study designs, publications were categorized according to the common nomenclature of study designs. 26 Study objectives were categorized by stage of maturity, modified after the World Health Organization “Monitoring and Evaluating Digital Health Interventions” manual to reflect the stages a wearable passes through from prototype to clinical implementation. 26 Collected parameters were categorized to warrant a coherent data analysis. A full list of categories with specifications is presented in Supplemental Table 5. In the case of study characteristics with more than one feature option (e.g. malignancy type, wearable type) reported shares might not add up to 100%. Registered planned studies from clinicaltrials.gov are presented as future perspectives.
Results
The literature search yielded a total of 2509 publications, with 112 publications meeting eligibility criteria after title, abstract and full-text screening (Figure 1). Main characteristics of included publications are listed in Table 1.
Figure 1.
PRISMA flow diagram of the screening and selection of publications.
Table 1.
Main characteristics of included publications; n = 112.
| Study ID | Number of participants (n) | Trial duration (days) | Study design (classified) | Treatment phase | Primary objective | Malignancy | Wearable detected parameters (classified) |
|---|---|---|---|---|---|---|---|
| Alberts et al. 57 | 65 | 30 | RCT | Survivor | User feasibility | Non-specific | Vital signs |
| Andraos et al. 30 | 38 | Not specified | Cross-sectional | During | Correlation of wearable data to clinical outcome | Non-specific | PA |
| Bade et al. 81 | 39 | 7 | Cross-sectional | Non-specific | User feasibility | Lung | PA |
| Blair et al. 82 | 54 | 91 | RCT | Survivor | User feasibility | Non-specific | PA; sedentary behavior |
| Carter et al. 83 | 29 | 90 | Secondary analysis | Survivor | Technical feasibility | Breast | Vital signs |
| Cereda et al. 84 | 10 | 27 | Prospective longitudinal | During | Technical feasibility | Myelogenous leukemia | Energy expenditure |
| Cos et al. 34 | 48 | 28 | Prospective longitudinal | Prior | Correlation of wearable data to clinical outcome | Pancreas | PA; vital signs; sleep |
| Courneya et al. 85 | 500 | Not specified | Prospective longitudinal | Survivor | User feasibility | Breast | PA; sedentary behavior; energy expenditure; posture |
| Dadhania et al. 86 | 42 | 82 | Prospective longitudinal | During | User feasibility | Glioma | PA |
| Dambrosio et al. 87 | 10 | Not specified | Prospective longitudinal | During | User feasibility | Leukemia | Vital Signs |
| Dambrosio et al. 31 | 17 | Not specified | Prospective longitudinal | During | Correlation of wearable data to clinical outcome | Leukemia | Vital signs |
| De Cocker et al. 88 | 9 | 7 | Cross-sectional | Survivor | User feasibility | Breast | PA |
| Dorion et al. 89 | 12 | 9 | Prospective longitudinal | During | Correlation of wearable data to clinical outcome | Non-specific | PA |
| Dreher et al. 90 | 65 | 270 | Prospective longitudinal | During | User feasibility | Breast | PA |
| Ferrante et al. 91 | 35 | 180 | RCT | Survivor | User feasibility | Breast | PA |
| Ferrante et al. 43 | 36 | 180 | RCT | Survivor | Outcome change | Breast | PA |
| Ferrante et al. 92 | 44 | 360 | Secondary analysis | Survivor | User feasibility | Breast | PA |
| Ferriolli et al. 33 | 182 | 7 | Prospective longitudinal | During | Correlation of wearable data to clinical outcome | Non-specific | PA; sedentary behavior; gait |
| Fink et al. 93 | not specified | Not specified | Prospective longitudinal | During | Correlation of wearable data to clinical outcome | Non-specific | ECG |
| Fischer et al. 94 | 20 | 7 | Secondary analysis | Non-specific | User feasibility | Non-specific | PA; sleep |
| Flora et al. 95 | 62 | 8.5 | Prospective longitudinal | During | Correlation of wearable data to clinical outcome | Non-specific | vital signs |
| Frensham et al. 96 | 8 | 42 | Quasi-experimental | Survivor | User feasibility | Non-specific | PA |
| Furtado et al. 97 | 34 | 7 | Cross-sectional | Post | User feasibility | Sarcoma | Gait |
| Gallicchio et al. 98 | 20 | 2.5 | Prospective longitudinal | Post | Technical feasibility | Thyroid | PA; vital signs; radiation |
| Gell et al. 99 | 19 | 28 | Quasi-experimental | Survivor | User feasibility | Non-specific | PA |
| Ghods et al. 100 | 40 | 21 | Prospective longitudinal | Non-specific | Correlation of wearable data to clinical outcome | Gastrointestinal | PA |
| Gresham et al. 101 | 37 | 14 | Prospective longitudinal | Non-specific | Correlation of wearable data to clinical outcome | Non-specific | PA |
| Groarke et al. 102 | 102 | 56 | Secondary analysis | Survivor | User feasibility | Non-specific | PA |
| Gupta et al. 103 | 24 | 84 | Cross-sectional | During | User feasibility | Non-specific | PA; sleep; sedentary behavior |
| Hardcastle et al. 104 | 68 | 84 | Secondary analysis | Survivor | User feasibility | Non-specific | PA |
| Hartman et al. 105 | 42 | 84 | Secondary analysis | Non-specific | User feasibility | Breast | PA; sedentary behavior |
| Hasan Shandhi et al. 29 | 12 | Not specified | Prospective longitudinal | Non-specific | User feasibility | Breast | ECG; left ventricular function |
| Helbrich et al. 106 | 99 | 28 | Prospective longitudinal | During | Correlation of wearable data to pro | Breast | PA |
| Hevroni et al. 38 | 40 | Not specified | Prospective longitudinal | During | Correlation of wearable data to pro | Multiple myeloma | Sleep |
| Holden et al. 107 | 80 | 8 | Prospective longitudinal | During | User feasibility | Colorectal | PA |
| Hoogland et al. 108 | 45 | 3 | Prospective longitudinal | Prior | Correlation of wearable data to pro | Hematologic | PA; sleep |
| Innominato et al. 32 | 77 | 3 | Secondary analysis | During | Correlation of wearable data to clinical outcome | Colorectal | PA; sleep |
| Innominato et al. 109 | 31 | 58 | Prospective longitudinal | During | Correlation of wearable data to clinical outcome | Non-specific | PA; sleep |
| Innominato et al. 17 | 268 | 3 | Secondary analysis | During | Correlation of wearable data to pro | Non-specific | PA; sleep |
| Ip et al. 110 | 10 | 22 | Quasi-experimental | During | User feasibility | NHL/ multiple myeloma | PA; vital signs |
| Izmailova et al. 111 | 188 | Not specified | Prospective longitudinal | During | Correlation of wearable data to clinical outcome | Non-specific | PA |
| Izmailova et al. 112 | 66 | Not specified | Prospective longitudinal | During | Correlation of wearable data to clinical outcome | Head/ neck | PA |
| Jakobsen et al. 39 | 40 | 1 | Cross-sectional | Non-specific | Correlation of wearable data to pro | Non-specific | sleep |
| Jeffery et al. 113 | 46 | 7 | Cross-sectional | Non-specific | User feasibility | Malignant pleural effusion | PA; sedentary behavior |
| Jonker et al. 114 | 24 | 90 | Prospective longitudinal | Post | User feasibility | Non-specific | PA |
| Jonker et al. 115 | 47 | 90 | Prospective longitudinal | Post | User feasibility | Non-specific | PA |
| Jovanovic et al. 116 | 38 | Not specified | Secondary analysis | Survivor | Correlation of wearable data to pro | Endometrial | PA |
| Jurdi et al. 41 | 63 | 13 | Prospective longitudinal | During | Correlation of wearable data to clinical outcome | Hematologic | PA; vital signs; sleep |
| Kerr et al. 60 | 30 | 7 | Cross-sectional | Survivor | Technical feasibility | Breast | PA |
| Khetrapal et al. 117 | 30 | 7 | Cross-sectional | During | User feasibility | Bladder | PA |
| Kim et al. 58 | 44 | 140 | Prospective longitudinal | During | Correlation of wearable data to clinical outcome | Thyroid | Vital signs |
| Kim et al. 118 | 40 | Not specified | Prospective longitudinal | During | Correlation of wearable data to pro | Gastrointestinal | PA; vital signs |
| Komarzynski et al. 119 | 31 | 30 | Prospective longitudinal | During | Correlation of wearable data to pro | Non-specific | Sleep |
| Kong et al. 47 | 152 | 7 | RCT | During | Outcome change | Breast | PA; energy expenditure |
| Koontz et al. 56 | 30 | 90 | Quasi-experimental | Survivor | User feasibility | Non-specific | PA |
| Korde et al. 37 | 40 | Not specified | Prospective longitudinal | During | Correlation of wearable data to pro | Multiple myeloma | PA; sleep |
| Laccetti et al. 120 | 15 | 182 | Prospective longitudinal | During | User feasibility | Prostate | PA; vital signs |
| LeBaron et al. 121 | not specified | Not specified | Cross-sectional | Non-specific | User feasibility | Non-specific | PA; vital signs |
| Lee et al. 48 | 100 | 84 | RCT | Post | Outcome change | Prostate | PA |
| Lemanska et al. 69 | 116 | 180 | Quasi-experimental | Survivor | Outcome change | Prostate | PA |
| Liu et al. 122 | 86 | 3 | Prospective longitudinal | During | Correlation of wearable data to pro | Breast | sleep |
| Low et al. 35 | 14 | 38 | Cross-sectional | During | Correlation of wearable data to pro | Gastrointestinal | PA; sleep; sedentary behavior |
| Low et al. 50 | 71 | 12 | Prospective longitudinal | Post | Correlation of wearable data to clinical outcome | Peritoneal | PA |
| Low et al. 123 | 24 | 28 | Cross-sectional | During | Correlation of wearable data to pro | Gastrointestinal | PA |
| Low et al. 124 | 44 | 81 | Prospective longitudinal | During | Correlation of wearable data to pro | Pancreas | PA; vital signs; sleep |
| Lozano-Lozano et al. 125 | 80 | 56 | Quasi-experimental | Survivor | User feasibility | Breast | PA; sedentary behavior |
| Lynch et al. 68 | 83 | 84 | RCT | Survivor | Outcome change | Breast | PA; sleep; sedentary behavior |
| Maddocks et al. 126 | 60 | 6 | Prospective longitudinal | Non-specific | User feasibility | Non-specific | PA; sedentary behavior; energy expenditure |
| Martin et al. 127 | 24 | 180 | Secondary analysis | Survivor | User feasibility | Breast | PA |
| Miropolsky et al. 128 | 13 | Not specified | Quasi-experimental | Survivor | User feasibility | Non-specific | PA |
| Nguyen et al. 129 | 14 | 14 | Cross-sectional | Survivor | User feasibility | Breast | PA; sedentary behavior |
| Nyrop et al. 44 | 100 | 84 | Prospective longitudinal | During | User feasibility | Breast | PA |
| O’Connor et al. 40 | 30 | 3 | Prospective longitudinal | Non-specific | Correlation of wearable data to pro | Non-specific | Sleep |
| Ohri et al. 130 | 38 | 70 | Prospective longitudinal | During | Correlation of wearable data to clinical outcome | Non-specific | PA |
| Otte et al. 131 | 55 | 7 | Prospective longitudinal | Survivor | Technical feasibility | Breast | Sleep |
| Palla et al. 52 | 54 | 5 | Quasi-experimental | Post | Correlation of wearable data to clinical outcome | Non-specific | Not specified |
| Pavic et al. 132 | 30 | 84 | Prospective longitudinal | Post | User feasibility | Non-specific | PA; vital signs |
| Pavic et al. 133 | 31 | 84 | Prospective longitudinal | Post | User feasibility | Non-specific | PA; vital signs; gait |
| Peterson et al. 134 | 137 | 10 | Cross-sectional | Non-specific | User feasibility | Non-specific | PA; vital signs |
| Peterson et al. 135 | 357 | Not specified | Prospective longitudinal | During | Correlation of wearable data to pro | Head/ neck | HR; vital signs |
| Pfister et al. 136 | 266 | 7 | Cross-sectional | Post | Technical feasibility | Breast | PA; sedentary behavior |
| Quintiliani et al. 137 | 10 | 70 | Prospective longitudinal | Survivor | User feasibility | Breast | PA |
| Rosenberg et al. 59 | 26 | 21 | Prospective longitudinal | Post | User feasibility | Prostate | PA; sedentary behavior |
| Rossi et al. 138 | 25 | 30 | Cross-sectional | Survivor | User feasibility | Endometrial | PA |
| Sada et al. 53 | 28 | 2 | Secondary analysis | Post | Correlation of wearable data to PRO | Non-Specific | PA; sleep; sedentary behavior; posture; balance |
| Schuler et al. 139 | 20 | 28 | Secondary analysis | During | User feasibility | Non-specific | PA |
| Skender et al. 27 | 198 | 10 | Cross-sectional | Non-specific | Technical feasibility | Colorectal | PA |
| Sugiyama et al. 140 | 21 | 39 | Prospective longitudinal | During | Correlation of wearable data to pro | Urothel | PA; vital signs; sleep; energy expenditure |
| Skipworth et al. 141 | 45 | 14 | Cross-sectional | Non-specific | Technical feasibility | Non-specific | PA |
| Slade et al. 142 | 20 | 30 | Prospective longitudinal | Post | User feasibility | Bladder | PA; vital signs; sleep |
| Strother et al. 45 | 33 | 36 | RCT | Post | Outcome change | Bladder | PA |
| Sun et al. 36 | 20 | 27 | Prospective longitudinal | Post | Correlation of wearable data to clinical outcome | Non-specific | PA |
| Timmerman et al. 54 | 17 | Not specified | Cross-sectional | Survivor | User feasibility | Lung | PA; vital signs |
| Tipps et al. 143 | not specified | 365 | Prospective longitudinal | Non-specific | Correlation of wearable data to pro | Glioblastoma | PA; sleep |
| Torrente et al. 144 | 112 | 7 | Cross-sectional | Non-specific | Correlation of wearable data to pro | Lung | PA; vital signs; radiation |
| Tramontano et al. 145 | not specified | Not specified | Prospective longitudinal | During | Correlation of wearable data to pro | Non-specific | PA; vital signs |
| Trinh et al. 146 | 46 | 84 | Quasi-experimental | Survivor | User feasibility | Prostate/ urothel | PA; sedentary behavior |
| Uhm et al. 49 | 356 | 84 | Quasi-experimental | Post | Outcome change | Breast | PA |
| Ungar et al. 147 | 67 | 28 | Quasi-experimental | Non-specific | Outcome change | Non-specific | PA |
| Vallance et al. 148 | 180 | 7 | Cross-sectional | Survivor | Correlation of wearable data to pro | Colon | PA; sedentary behavior |
| Valle et al. 55 | 35 | 180 | RCT | Survivor | Outcome change | Breast | PA |
| Van Blarigan et al. 149 | 41 | 84 | RCT | Survivor | User feasibility | Colorectal | PA; energy expenditure |
| Verma et al. 150 | 86 | 7 | Prospective longitudinal | Post | Correlation of wearable data to clinical outcome | Hematologic | Vital signs |
| Waliany et al. 151 | 25 | 10 | Prospective longitudinal | During | Technical feasibility | Breast | Energy expenditure |
| Waller et al. 46 | 22 | Not specified | RCT | Prior | Outcome change | Colorectal | PA |
| Ward et al. 152 | 80 | 4 | Prospective longitudinal | Post | User feasibility | Colorectal | PA |
| Wong et al. 153 | 26 | Not specified | Prospective longitudinal | Non-specific | Correlation of wearable data to clinical outcome | Non-specific | PA; energy expenditure |
| Wright et al. 154 | 10 | 30 | Quasi-experimental | Post | User feasibility | Non-specific | PA; vital signs |
| Wu et al. 51 | 43 | 28 | Prospective longitudinal | Post | Correlation of wearable data to clinical outcome | Gastric | PA |
| Wu et al. 155 | 10 | 362 | Secondary analysis | Non-specific | Correlation of wearable data to clinical outcome | Non-specific | PA |
| Xu et al. 42 | 91 | 88 | Quasi-experimental | Survivor | Outcome change | Breast | PA; vital signs |
| Zahiri et al. 156 | 139 | Not specified | Cross-sectional | Survivor | Correlation of wearable data to clinical outcome | Non-specific | Gait; posture |
An increasing number of published studies can be observed from the earliest eligible publication in 2007 until 2021 (26 publications) with a short-lasting decrease in 2020 (14 studies) (Figure 2). Most studies were published by first authors located in the USA (55.4%; 62/112), followed by Europe (29.5%; 33/112), Asia (6.3%; 7/112), Canada and Australia (both 4.5%; 5/112) (Supplemental Figure 4).
Figure 2.
Number of published studies (year 2007–2021) and registered studies on clinicaltrials.gov (year >2021) per treatment phase per year of publication.
The search of clinicaltrials.gov resulted in 117 registrations, of which 61 were eligible. 63.9% (39/61) of those were registered by researchers located in the USA (Supplemental Figure 5).
Study characteristics
Study characteristics are summarized in Table 2. Around 10% of publications were RCTs and nearly half of publications applied a prospective longitudinal design (47.3%; 53/112) followed by 19.6% (22/112) cross-sectional studies.
Table 2.
Main characteristics in publications and registered studies.
| Published studies | Registered studies (clinicaltrials.gov) | |||
|---|---|---|---|---|
| Study characteristics | ||||
| Number | 112 | 61 | ||
| Study design | RCT | 9.8% (n = 11) | 44.3% (n = 27) | |
| Quasi-experimental | 11.6% (n = 13) | 11.5% (n = 7) | ||
| Prospective longitudinal | 47.3% (n = 53) | 32.8% (n = 20) | ||
| Cross-sectional | 19.6% (n = 22) | 11.5% (n = 7) | ||
| Secondary analyses | 11.6% (n = 13) | 0% (n = 0) | ||
| Mean no. of participants (min − max) | 64.4 (8–500) | 97.9 (10–528) | ||
| Weighted mean study duration (days) (95% CI) | 36.6 (25.1–47.5) | - | ||
| Planned mean study duration (days) (min − max) | - | 124.3 (1 h – 365 days) | ||
| Device characteristics | ||||
| Device type | Medical-grade | 22.3% (n = 25) | 11.5% (n = 7) | |
| Consumer-grade | 50.0% (n = 56) | 50.8% (n = 31) | ||
| Not specified | 34.8% (n = 39) | 37.7% (n = 23) | ||
| Parameters | PA | 80.4% (n = 90) | 70.5% (n = 43) | |
| Vital signs | 23.2% (n = 26) | 31.1% (n = 19) | ||
| Sleep | 15.2% (n = 17) | 11.5% (n = 7) | ||
| Device company | Fitbit | 31.3% (n = 35) | 37.7% (n = 23) | |
| ActiGraph | 15.2% (n = 17) | 3.3% (n = 2) | ||
| PAL technologies | 5.4% (n = 6) | 0% (n = 0) | ||
| GARMIN | 5.4% (n = 6) | 8.2% (n = 5) | ||
| Participants’ characteristics | ||||
| Demographics | Weighted mean age (years) (95% CI) | 50.1 (48.2–51.9) | - | |
| Female ratio (%) (95% CI) | 51.5 (45.3–57.7) | - | ||
| Malignancy type | Multiple | 42.9% (48/112) | 39.3% (24/112) | |
| Breast | 22.3% (n = 25) | 16.4% (n = 10) | ||
| Hematologic/lymphatic | 8.0% (n = 9) | 3.3% (n = 2) | ||
| Prostate/Urothel | 8.0% (n = 9) | 8.2% (n = 5) | ||
| Colorectal | 6.3% (n = 7) | 3.3% (n = 2) | ||
| Gastrointestinal | 4.5% (n = 5) | 6.6 (n = 4) | ||
| Lung | 2.7% (n = 3) | 11.5% (n = 7) | ||
| Treatment phase | Pre-treatment | 2.7% (n = 3) | 6.6% (n = 4) | |
| During treatment | 34.8% (n = 39) | 23.0% (n = 14) | ||
| Post-treatment | 17.9% (n = 20) | 26.2% (n = 16) | ||
| Survivor | 27.7% (n = 31) | 11.5% (n = 7) | ||
| Not specified/multiple | 17.0% (n = 19) | 32.8% (n = 20) | ||
Only 8.9% (10/112) of publications conducted outcome change analyses (Figure 3), applying a weighted mean duration of 80.2 days (47.8–112.7).
Figure 3.
Primary study objectives of published studies according to the stage of maturity, modified after “WHO monitoring and evaluating digital health interventions.” The sizes of the boxes do not correspond to the number of studies of the respective category.
Interestingly, compared to this, the share of registered RCTs is four times higher. The share of registered outcome change analysis is three times higher compared to publications, applying a higher planned average duration of 129.1 days.
Device characteristics
Device characteristics are shown in Table 2. 22.3% (25/112) of publications applied medical-grade devices. Within the subset of EU-based publications, 27.8% (5/18) incorporated medical-grade devices, while in USA-based publications, medical-grade devices were used in 17.7% (11/62) of publications.
The most commonly used sensors were accelerometers and actigraphs (85.7%; 96/112), followed by HR sensors (16.1%; 18/112), temperature sensors (3.6%; 4/112) and ECG (1.8%; 2/112). In 76.8% (86/112) of publications, one sensor was applied, 21.4% (24/112) utilized two to four sensors and 1.8% (2/112) did not specify applied sensors.
Registered studies do not show a clear trend to applying multiple sensors: 65.6% (40/61) of registered studies are planning to employ one sensor, followed by two sensors (18.0%; 11/61), five sensors (1.6%; 1/61) or not specified sensor count (9.9%; 9/91).
Participants’ characteristics
Participants’ characteristics are reported in Table 2. Included populations had a weighted mean age of 50.1 (48.2–51.9) years and a mean female participant ratio of 51.5%, or 45.2%, excluding studies assessing a sex-specific malignancy. Most publications (42.9%) and registered studies (39.3%) featured multiple cancer types, with breast cancer as the prevalent specific type (22.3% in publications, 16.4% in registered studies).
Primary objectives
Figure 3 depicts the primary study objectives of publications according to the maturity stage of the assessed wearable.
Technical feasibility
8.0% (9/112) of publications assessed the accuracy, reliability, or validation of wearables, for example, minimum number of nights needed to obtain accurate results of objective sleep 29 or association of wearable and pedometer measurements. 27
User feasibility
Most publications (42.9%; 48/112) examined user feasibility, for example, by assessing wearable usage, trial completion, data transmission or adverse events. This heterogeneity of feasibility definition is described in detail by other reviewers. 28 A single study described wearable applications as “potentially feasible,” 29 and all remaining studies concluded a high level of feasibility.
The share of feasibility assessment as the primary objective will be exceeded by registered studies (50.1%; 31/61).
Correlational analysis
Correlational analyses of wearable data to clinical outcomes and to PRO were conducted by 40.2% (45/112) of publications. Wearable data were demonstrated to correlate to hospitalization rate, 30 AE, 31 overall survival, 32 performance, 33 risk, 34 symptoms35,36 and QoL. 37 However, consistent correlations of wearable data to subjectively reported PA or sleep were not established due to inaccurate sleep tracking and misinterpretation of inactivity as sleep by wearables.38–40 Similarly, according to the authors, a correlation between activity and hospitalization duration could not be demonstrated consistently due to variations in the study population. 41
Fewer registered studies plan to conduct correlational analyses (19.7%; 12/61), resembling clinical endpoints of published studies. One novel approach is a correlational analysis of wearable data and pain.
Outcome change analysis
A small share of publications analyzed outcome change (8.9%; 10/112), predominantly aiming to improve PA as the primary endpoint (80%; 8/10). Two publications measured weight loss as a primary endpoint (20%; 2/10).42,43 A beneficial effect of wearables was mainly apparent among patients characterized as posttreatment and cancer survivors. Two publications report a lack of positive outcomes after wearable-supported interventions, due to a lack of motivation 44 and underpowered study conditions, 45 according to the authors.
Registered trials plan to conduct more outcome change analyses (27.9%; 17/61) with a broader variability of clinical endpoints, which will be reported in the respective treatment phases of the following paragraph.
Treatment phases
The distribution of treatment phases in which the studies were conducted is presented in Table 2. Study endpoints within respective treatment phases and respective stages of maturity are presented in Figure 4.
Figure 4.
Study endpoints within the continuum of oncology care, categorized according to study objectives’ stage of maturity (n = 112). The number in brackets represents the number of studies in the respective category. Technical feasibility is not displayed.
Pretreatment phase
Of the three publications that assessed patients pre-treatment, two studies were conducted as prospective longitudinal studies and one study was an RCT. 46 Wearables were reported to be successfully applied for surgical risk assessment 34 and as a prehabilitation tool before surgery. 46
Registered studies show a relative increase in studies utilizing wearables in the pre-treatment phase. Of the three studies that plan to apply wearables for prehabilitation, two will conduct outcome change analysis (NCT03902834, NCT03689634) and one will assess feasibility (NCT03162718).
During treatment phase
Of the 39 publications that assessed patients during treatment, one was designed as RCT, aiming to increase PA 47 and 26 publications (66.7%) conducted correlational analyses.
Of the 20 registered studies, during treatment, wearables will mainly be applied for feasibility assessment. An exercise intervention for balance improvement by wearable application represents a novel investigational approach in oncology (NCT02773329).
Posttreatment phase
Of the 20 publications which assessed patients post-treatment, one was designed as RCT. 45 Wearables were reported to be successfully applied to increase PA,42,48,49 predict hospitalization50,51 and toxicities 52 and objectify symptoms, 53 among others. One study implementing an intervention to increase PA did not show a significant benefit due to underpowered study conditions, according to the authors. 45
Of the 16 registered studies, wearables post-treatment are planned to be applied to conduct exercise interventions to increase PA mostly, either the latter being the primary endpoint (NCT05046756, NCT05031052, NCT04907591, NCT04783168) or investigating subsequent effects of PA increase, for example, reduced readmissions (NCT04314778), decreased insulin resistance (NCT04649671), improved bone mineral density (NCT04144127) and changes in mental health (NCT03874754). Further registered studies aim to decrease ultraviolet (UV) exposure (NCT03344796, NCT03518229).
Survivor phase
Of the 31 publications including cancer survivors, 21 conducted feasibility assessments. Seven studies were designed as RCTs, assessing feasibility and outcome change of exercise interventions. Wearables were reported to be successfully applied to increase PA 54 and to reduce body weight. 43
Of the nine registered studies, wearable-based interventions aim to increase PA (NCT03612596) and reduce UV exposure (NCT03927742). Feasibility assessment of a breathing intervention represents a novel approach to wearable application in oncology (NCT03773380).
No specific treatment phase
Of the 19 published studies that did not specify a treatment phase or analyzed cohorts from multiple treatment phases, most studies included cohorts with multiple malignancy types (57.9%; 11/19).
One study of 12 registered studies plans to decrease cardiotoxicity risk by implementing a preventive, artificial intelligence-based intervention in a clinical setting (NCT04885088).
Quality assessment
The full evaluation is given in Supplemental Table 4. The majority of publications eligible for quality assessment did not specify their study design in the title or abstract by using commonly recognized terms (56.3%; 36/64). Eligibility criteria, sources and methods of selection of participants and methods of follow-up were not defined by 31.3% (20/64) of studies.
Study size determination was described in only 12.5% (8/64) of studies. Most studies did not address missing data handling 81.3% (52/64). Number of participants at each stage of the study was not reported by 31.3% (20/64) of studies. Generalizability was not discussed by 70.3% (45/64) of studies. Reasons for limited generalizability were selection bias to patient selection, for example, due to required technical 55 /smartphone access56,57 or required smartphone literacy 58 and small sample size.59,60
Discussion
For this scoping review, 112 research papers, published between 2007 and 2021, were identified that examined wearable applications in oncology, regarding their technical and user feasibility, correlation of wearable data to PRO and to clinical outcomes and their capability to change participants’ lifestyles. This study indicates a rising use of wearables addressing variable endpoints in oncology research across the continuum of oncology care.
The surge in studies on wearables in oncology is driven by technological advancements, a focus on patient empowerment and personalized medicine, the potential for cost-effectiveness and early detection and intervention, interdisciplinary collaboration, demand for increasing outpatient treatment and surveillance and growing regulatory interest. This exponential growth of publications on wearable applications has been documented not only in oncology28,61–63 but also in other medical fields,64,65 for example, chronic diseases. 66 In oncology, prior reviews assessed wearable devices, but limited their research to specific data items, for example, activity data, 62 analyzing wearable usage in a certain oncologic treatment phase, 28 or investigating particular objectives, for example, adherence and clinical outcomes.28,67
The average age of study participants was ∼50 years, with this younger demographic particularly receptive to new technologies. A restricted access to wearable usage due to lack of familiarity with smart devices and technical requirements such as reliable internet connection or the necessity of owning a compatible private smartphone limit generalizability of included studies.55–57 Further selection biases discussed by authors of included studies arise due to higher motivation 68 of study participants, higher educational level 69 and a healthier lifestyle 70 compared to the general cancer populations. In order to antagonize digital health disparities, these aspects should be considered while interpreting the results of the respective trials. For real-world implementation, these aspects should be addressed by wearable manufacturers, HCPs and research centers.
Further, the evaluation of the efficacy of wearables on clinical outcomes, a crucial step before clinical implementation, was only conducted sparsely, primarily showing success in survivor cohorts and in posttreatment cohorts. Despite a slight trend toward assessment during and posttreatment, critical treatment phases require further investigation, addressing clinical endpoints beyond change in PA.
More homogenous participant cohorts, especially concerning a specific cancer type, could add value to research results since physical conditions of patients with different cancer types and treatment modalities often vary. Evidence of wearable application in specific cancer cohorts might facilitate clinical implementation of wearables, since patients are often bound to cancer-specific centers or wards and thus could be elected and approached easily.
The clinical impact on oncology outcomes currently mainly seems favorable to PA improvement, in line with Jo et al., who investigated clinical outcomes of wearable application in patients with chronic diseases. 66 Successful long-term PA improvement and subsequent clinical changes remain to be shown. Only two studies confirmed a significant PA increase after 3 months compared to a control group with no intervention and no wearable.46,68 Of those, one was a single-center study with 22 participants, which achieved a significant improvement in the 6 Minutes-Walk-Test. However, a significant change in daily active minutes could not be achieved. 46 Lynch et al. describe a significant PA increase and sedentary time reduction in breast cancer survivors after three months. 68 Further studies indicated PA increase or weight loss up to 3 months, but without the comparison to a control group, 42 or with no significant difference to included control groups.42,43,48,49,69 Only two studies present outcome change results after 6 months: Lemanska et al. report a PA decrease after initial PA increase after 3 months (with no control group) and Kong et al. found increased PA levels with no significant difference to a control group without wearable.46,69
In line with an increase in PA, two of the above-mentioned publications observed significant weight loss after 3 months,42,43 whereas QoL remained unaltered. 42 Future studies should address additional clinical endpoints induced by altered PA. Limited change can be observed considering the number of applied sensors in published studies and planned studies. Wearable research in oncology populations is predominantly based on one sensor—both in published and registered studies. Despite these trends, longer study duration and inclusion of well-defined patient populations with respective control groups are desirable. The high share of feasibility analyses of publications and planned studies should proceed to more advanced study objectives. Innovative technology with multi-sensor approaches could enhance wearables’ potential in predicting diseases and optimizing treatment selection through the concept of a “digital twin.”
A high share of studies published by US-based authors was observed in line with other reviews.62,70 Strict regulatory laws concerning medical certification in the EU were presumed as a cause. 70 This assumption can only be agreed with regarding prospective medical certification since this review found a high share of studies that applied consumer-grade devices without medical certification—both in the EU and the USA. More strict data privacy and security regulations in the EU complicate research in this sensitive field, both from researchers’ and from participants’ points of view, who might be skeptical concerning the adaption of new technologies and especially concerning the processing of sensitive personal health data. 71 One-third of the adult US population is estimated to use wearables 72 and more than 80% of US wearable users report willingness to share wearable information with HCPs. 73 In contrast, only 19% of the EU population between 16 and 74 years reported to use wearables in 2020. 74 Although this information cannot be directly compared, wearable usage is likely more common in the USA than in Europe.
“No evidence, no implementation—no implementation, no evidence”—this paradox stated by Guo et al. encapsulates the challenges of digital health, relating to a lack of real-world evidence of digital health solutions. 75 Evidence-based evaluation of wearables is crucial for patients, HCPs, investors, regulatory bodies, wearable developers and benefactors. Observational study designs with small sample sizes were prevalent in this scoping review, mainly focusing on feasibility and correlational analyses during cancer treatment and in cancer survivors. Despite low flexibility and long periods from planning to publication and difficulties of blinding, RCTs remain the gold standard for efficacy assessment. Only a small proportion of publications were RCTs, while a higher share of registered studies plans to conduct RCTs, focusing on during and post-treatment evaluations.
Innovative study designs are needed to address the challenges encountered in digital health research, for example, dynamics of digitization, software updates and software bugs. “Pragmatic RCT,” adaptive designs and virtual trials are possible approaches to complement conventional RCT. 76 Alternative study designs such as Continuous Evaluation of Evolving Behavioral Intervention Technologies (CEEBIT), Multiphase Optimization Strategy (MOST), Sequential Multiple Assignment Randomized Trials (SMART), Factorial/Fractional RCT and Micro-Randomized Trials (MRT) are seen as helpful tools for the development of digital health interventions.76–78 Various frameworks and guidelines for the evaluation of digital health interventions are reviewed by Kernebeck et al., including, for example, a CONSORT-/STROBE expansion for eHealth interventions. 79
Moreover, our review indicates a need for further efforts to use medical-grade wearables for accurate data collection. The results call for investigations of wearables’ short-term and long-term effectiveness in oncology populations, including survival rates, malignancy recurrence rates and hospitalization rates. Wearables’ capability to potentially predict symptoms and identify patients’ deterioration early may facilitate patient care with malignant diseases and result in improved clinical outcomes. Besides health outcome changes, positive effects of wearable application on healthcare processes should be demonstrated, for example, in terms of economic or financial evaluation and workload reduction of HCPs.
The strengths and limitations of the present study merit consideration. First, the application of a wearable and measurement of parameters, which can be consciously controlled by the patient (e.g. PA), could be seen as an intervention per se, according to the Hawthorne effect. 80 In this review, studies intending to change collected parameters (e.g. through a step goal) were considered interventional studies and studies which applied wearables to, for example, foresee clinical events, were considered observational studies. Second, the categorization of parameters was conducted carefully, with an attempt to achieve a meaningful and objective evaluation. However, any categorization can lead to a certain bias in the results—especially when analyzing and classifying heterogeneous studies. Furthermore, the categories are partly overlapping and are not as clear in practice as they are in theory, which might lead to restricted reproducibility. Third, technical devices new to the market, such as wearables, are prone to fast-moving development of hardware and software. Since trials often present momentary assessment of devices, technical changes, especially in early published studies, cannot warrant continued validity. This also applies to this scoping review. Fourth, the interpretation of clinicaltrials.gov is limited by the fact that details of the study, such as trial performance, completion and publication, are not reliable. Aimed study designs and methods could be adapted due to altered circumstances and downward adjustment of planned study quality seems more likely than vice versa.
Conclusions
Although less than 10% of clinical evaluations of wearables in oncology patients were done by RCTs so far, wearables appear to show promise for improving clinical care of patients with malignant diseases. Challenges in evidence creation, study design and regulatory aspects need to be addressed for their effective implementation. Long-term outcome change of wearables should be analyzed to demonstrate their effectiveness. Wearable application should be assessed in critical treatment phases, analyzing endpoints beyond PA change. Application of medical-grade wearables is desirable, optimally able to process big data sets or even enabling the adaption of algorithms based on the “digital twin” concept.
Supplemental Material
Supplemental material, sj-docx-1-dhj-10.1177_20552076241233998 for Application of wearables for remote monitoring of oncology patients: A scoping review by Katharina Cloß, Marlo Verket, Dirk Müller-Wieland, Nikolaus Marx, Katharina Schuett, Edgar Jost, Martina Crysandt, Fabian Beier, Tim H Brümmendorf, Guido Kobbe, Julia Brandts and Malte Jacobsen in DIGITAL HEALTH
Supplemental material, sj-xlsx-2-dhj-10.1177_20552076241233998 for Application of wearables for remote monitoring of oncology patients: A scoping review by Katharina Cloß, Marlo Verket, Dirk Müller-Wieland, Nikolaus Marx, Katharina Schuett, Edgar Jost, Martina Crysandt, Fabian Beier, Tim H Brümmendorf, Guido Kobbe, Julia Brandts and Malte Jacobsen in DIGITAL HEALTH
Supplemental material, sj-xlsx-3-dhj-10.1177_20552076241233998 for Application of wearables for remote monitoring of oncology patients: A scoping review by Katharina Cloß, Marlo Verket, Dirk Müller-Wieland, Nikolaus Marx, Katharina Schuett, Edgar Jost, Martina Crysandt, Fabian Beier, Tim H Brümmendorf, Guido Kobbe, Julia Brandts and Malte Jacobsen in DIGITAL HEALTH
Supplemental material, sj-xlsx-4-dhj-10.1177_20552076241233998 for Application of wearables for remote monitoring of oncology patients: A scoping review by Katharina Cloß, Marlo Verket, Dirk Müller-Wieland, Nikolaus Marx, Katharina Schuett, Edgar Jost, Martina Crysandt, Fabian Beier, Tim H Brümmendorf, Guido Kobbe, Julia Brandts and Malte Jacobsen in DIGITAL HEALTH
Supplemental material, sj-xlsx-5-dhj-10.1177_20552076241233998 for Application of wearables for remote monitoring of oncology patients: A scoping review by Katharina Cloß, Marlo Verket, Dirk Müller-Wieland, Nikolaus Marx, Katharina Schuett, Edgar Jost, Martina Crysandt, Fabian Beier, Tim H Brümmendorf, Guido Kobbe, Julia Brandts and Malte Jacobsen in DIGITAL HEALTH
Supplemental material, sj-xlsx-6-dhj-10.1177_20552076241233998 for Application of wearables for remote monitoring of oncology patients: A scoping review by Katharina Cloß, Marlo Verket, Dirk Müller-Wieland, Nikolaus Marx, Katharina Schuett, Edgar Jost, Martina Crysandt, Fabian Beier, Tim H Brümmendorf, Guido Kobbe, Julia Brandts and Malte Jacobsen in DIGITAL HEALTH
Supplemental material, sj-xlsx-7-dhj-10.1177_20552076241233998 for Application of wearables for remote monitoring of oncology patients: A scoping review by Katharina Cloß, Marlo Verket, Dirk Müller-Wieland, Nikolaus Marx, Katharina Schuett, Edgar Jost, Martina Crysandt, Fabian Beier, Tim H Brümmendorf, Guido Kobbe, Julia Brandts and Malte Jacobsen in DIGITAL HEALTH
Supplemental material, sj-pptx-8-dhj-10.1177_20552076241233998 for Application of wearables for remote monitoring of oncology patients: A scoping review by Katharina Cloß, Marlo Verket, Dirk Müller-Wieland, Nikolaus Marx, Katharina Schuett, Edgar Jost, Martina Crysandt, Fabian Beier, Tim H Brümmendorf, Guido Kobbe, Julia Brandts and Malte Jacobsen in DIGITAL HEALTH
Acknowledgments
None.
Footnotes
Contributorship: KC contributed to conceptualization, methodology, data curation, formal analysis and writing–original draft. MV contributed to conceptualization, methodology, data curation and review and editing. DM-W contributed to supervision, conceptualization and review and editing. NM contributed to review and editing. KS contributed to review and editing. EJ contributed to review and editing. MC contributed to review and editing. FB contributed to review and editing. TB contributed to review and editing. GK contributed to review and editing. JB contributed to project administration, conceptualization, methodology, data curation, visualization and review and editing. MJ contributed to project administration, conceptualization, methodology, data curation, visualization and review and editing.
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Ethical approval: Not applicable.
Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This research received funding from the internal grant program of the University Hospital Aachen, RWTH Aachen University, Germany. Dr Marx is supported by the Deutsche Forschungsgemeinschaft (German Research Foundation; TRR 219; Project-ID 322900939 [M03, M05]).
Guarantor: MJ, JB.
Informed consent: This manuscript presents a literature review of publicly available information. Since no individual data were used for the analysis presented in this manuscript, the requirement for consent is not applicable.
Registration: The review was registered in the Open Science Framework register by first author KC (née Hagenkamp) (https://osf.io/bgmya).
ORCID iD: Katharina Cloß https://orcid.org/0009-0007-4430-1242
Supplementary material: Supplemental material for this article is available online.
References
- 1.Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021; 71: 209–249. DOI: 10.3322/caac.21660 [DOI] [PubMed] [Google Scholar]
- 2.Varricchi G, Galdiero MR, Marone G, et al. Cardiotoxicity of immune checkpoint inhibitors. ESMO Open 2017; 2: e000247. DOI: 10.1136/esmoopen-2017-000247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Guan M, Zhou YP, Sun JL, et al. Adverse events of monoclonal antibodies used for cancer therapy. Biomed Res Int 2015; 2015: 428169. DOI: 10.1155/2015/428169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Handforth C, Clegg A, Young C, et al. The prevalence and outcomes of frailty in older cancer patients: a systematic review. Ann Oncol 2015; 26: 1091–1101. DOI: 10.1093/annonc/mdu540. [DOI] [PubMed] [Google Scholar]
- 5.Romero SAD, Jones L, Bauml JM, et al. The association between fatigue and pain symptoms and decreased physical activity after cancer. Support Care Cancer 2018; 26: 3423–3430. DOI: 10.1007/s00520-018-4203-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Niedzwiedz CL, Knifton L, Robb KA, et al. Depression and anxiety among people living with and beyond cancer: a growing clinical and research priority. BMC Cancer 2019; 19: 20191011. DOI: 10.1186/s12885-019-6181-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Katsaras T, Milsis A, Rizikari M, et al. The use of the “Healthwear” wearable system in chronic patients’ early hospital discharge, control randomized clinical trial. Int Symp Med Inf Commun Technol 2011; 5: 143–146. DOI: 10.1109/ISMICT.2011.5759815. [DOI] [Google Scholar]
- 8.Kwasnicki RM, Ali R, Jordan SJ, et al. A wearable mobility assessment device for total knee replacement: a longitudinal feasibility study. Int J Surg 2015; 18: 14–20. DOI: 10.1016/j.ijsu.2015.04.032. [DOI] [PubMed] [Google Scholar]
- 9.Mohamadipanah H, Wise B, Witt A, et al. Performance assessment using sensor technology. J Surg Oncol 2021; 124: 200–215. DOI: 10.1002/jso.26519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Hanna TP, King WD, Thibodeau S, et al. Mortality due to cancer treatment delay: systematic review and meta-analysis. Br Med J 2020; 371: m4087. DOI: 10.1136/bmj.m4087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Coorey G, Figtree GA, Fletcher DF, et al. The health digital twin to tackle cardiovascular disease-a review of an emerging interdisciplinary field. NPJ Digit Med 2022; 5: 126. DOI: 10.1038/s41746-022-00640-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Cheong SHR, Ng YJX, Lau Y, et al. Wearable technology for early detection of COVID-19: a systematic scoping review. Prev Med 2022; 162: 107170. DOI: 10.1016/j.ypmed.2022.107170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Singhal A, Cowie MR. The role of wearables in heart failure. Curr Heart Fail Rep 2020; 17: 125–132. DOI: 10.1007/s11897-020-00467-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Varatharajan R, Manogaran G, Priyan MK, et al. Wearable sensor devices for early detection of Alzheimer disease using dynamic time warping algorithm. Cluster Comput 2017; 21: 681–690. DOI: 10.1007/s10586-017-0977-2. [DOI] [Google Scholar]
- 15.Verdru J, Van Paesschen W. Wearable seizure detection devices in refractory epilepsy. Acta Neurol Belg 2020; 120: 1271–1281. DOI: 10.1007/s13760-020-01417-z. [DOI] [PubMed] [Google Scholar]
- 16.Breteler MJM, KleinJan E, Numan L, et al. Are current wireless monitoring systems capable of detecting adverse events in high-risk surgical patients? A descriptive study. Injury 2020; 51: S97–S105. DOI: 10.1016/j.injury.2019.11.018. [DOI] [PubMed] [Google Scholar]
- 17.Innominato PF, Komarzynski S, Palesh OG, et al. Circadian rest-activity rhythm as an objective biomarker of patient-reported outcomes in patients with advanced cancer. Cancer Med 2018; 7: 4396–4405. 10.1002/cam4.1711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Johnson A, Yang F, Gollarahalli S, et al. Use of mobile health apps and wearable technology to assess changes and predict pain during treatment of acute pain in sickle cell disease: feasibility study. JMIR Mhealth Uhealth 2019; 7: e13671. DOI: 10.2196/13671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ferguson T, Olds T, Curtis R, et al. Effectiveness of wearable activity trackers to increase physical activity and improve health: a systematic review of systematic reviews and meta-analyses. Lancet Digit Health 2022; 4: e615–e626. DOI: 10.1016/S2589-7500(22)00111-X. [DOI] [PubMed] [Google Scholar]
- 20.Singh B, Zopf EM, Howden EJ. Effect and feasibility of wearable physical activity trackers and pedometers for increasing physical activity and improving health outcomes in cancer survivors: a systematic review and meta-analysis. J Sport Health Sci 2022; 11: 184–193. DOI: 10.1016/j.jshs.2021.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Brickwood KJ, Watson G, O’Brien J, et al. Consumer-based wearable activity trackers increase physical activity participation: systematic review and meta-analysis. JMIR mHealth uHealth 2019; 7: e11819. DOI: 10.2196/11819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Franssen WMA, Franssen G, Spaas J, et al. Can consumer wearable activity tracker-based interventions improve physical activity and cardiometabolic health in patients with chronic diseases? A systematic review and meta-analysis of randomised controlled trials. Int J Behav Nutr Phys Act 2020; 17: 57. DOI: 10.1186/s12966-020-00955-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Zahrt OH, Evans K, Murnane E, et al. Effects of wearable fitness trackers and activity adequacy mindsets on affect, behavior, and health: longitudinal randomized controlled trial. J Med Internet Res 2023; 25: e40529. DOI: 10.2196/40529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Tricco AC, Lillie E, Zarin W, et al. PRISMA extension for scoping reviews (PRISMA-ScR): checklist and explanation. Ann Intern Med 2018; 169: 467–473. DOI: 10.7326/M18-0850. [DOI] [PubMed] [Google Scholar]
- 25.Hagenkamp K, Jacobsen M. Application of wearable sensors for remote monitoring of oncology patients: a scoping review, 10.17605/OSF.IO/BGMYA (2021). [DOI]
- 26.World Health Organization. Monitoring and evaluating digital health interventions: a practical guide to conducting research and assessment. Geneva: WHO, 2016. [Google Scholar]
- 27.Skender S, Schrotz-King P, Bohm J, et al. Repeat physical activity measurement by accelerometry among colorectal cancer patients–feasibility and minimal number of days of monitoring. BMC Res Notes 2015; 8: 222. DOI: 10.1186/s13104-015-1168-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Beauchamp UL, Pappot H, Hollander-Mieritz C. The use of wearables in clinical trials during cancer treatment: systematic review. JMIR mHealth uHealth 2020; 8: e22006. 10.2196/22006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Hasan Shandhi MM, Aras M, Wynn S, et al. Cardiac function monitoring for patients undergoing cancer treatments using wearable seismocardiography: a proof-of-concept study. Conf Proc IEEE Eng Med Biol Soc 2020; 2020: 4075–4078. DOI: 10.1109/EMBC44109.2020.9176074. [DOI] [PubMed] [Google Scholar]
- 30.Andraos TY, Asaro AM, Garg MK, et al. Real-time activity monitoring to prevent admissions during radiotherapy (RAMPART). Int J Radiat Oncol Biol Phys 2019; 105: E463. DOI: 10.1016/j.ijrobp.2019.06.1424. [DOI] [Google Scholar]
- 31.Dambrosio N, Porter M, Bauer E, et al. Identifying neutropenic fever earlier: an application of a skin patch for continuous temperature monitoring. Blood 2018; 132: 4718. DOI: 10.1182/blood-2018-99-114181. [DOI] [Google Scholar]
- 32.Innominato PF, Giacchetti S, Bjarnason GA, et al. Prediction of overall survival through circadian rest-activity monitoring during chemotherapy for metastatic colorectal cancer. Int J Cancer 2012; 131: 2684–2692. 10.1002/ijc.27574. [DOI] [PubMed] [Google Scholar]
- 33.Ferriolli E, Skipworth RJE, Hendry P, et al. Physical activity monitoring: a responsive and meaningful patient-centered outcome for surgery, chemotherapy, or radiotherapy? J Pain Symptom Manage 2012; 43: 1025–1035. 10.1016/j.jpainsymman.2011.06.013. [DOI] [PubMed] [Google Scholar]
- 34.Cos H, Li D, Williams G, et al. Predicting outcomes in patients undergoing pancreatectomy using wearable technology and machine learning: prospective cohort study. JMIR 2021; 23: e23595. 10.2196/23595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Low CA, Dey AK, Ferreira D, et al. Estimation of symptom severity during chemotherapy from passively sensed data: exploratory study. JMIR 2017; 19: e420. 10.2196/jmir.9046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Sun V, Dumitra S, Ruel N, et al. Wireless monitoring program of patient-centered outcomes and recovery before and after major abdominal cancer surgery. JAMA surg 2017; 152: 852–859. 10.1001/jamasurg.2017.1519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Korde N, Tavitian E, Derkach A, et al. Association of patient activity bioprofiles with Hrqol and clinical responses: a prospective novel trial using mobile wearables in newly diagnosed multiple myeloma patients. Blood 2020; 136: 26–28. DOI: 10.1182/blood-2020-142082. [DOI] [Google Scholar]
- 38.Hevroni G, Mastey D, Tavitian E, et al. Using mobile wearables to establish sleep bioprofiles in newly diagnosed multiple myeloma (MM) patients. JCO 2021; 39: 15. DOI: 10.1200/JCO.2021.39.15_suppl.8040. [DOI] [Google Scholar]
- 39.Jakobsen G, Engstrom M, Thronaes M, et al. Sleep quality in hospitalized patients with advanced cancer: an observational study using self-reports of sleep and actigraphy. Supportive Care Cancer 2020; 28: 2015–2023. 10.1007/s00520-019-04998-5. [DOI] [PubMed] [Google Scholar]
- 40.O’Connor B, Uí Dhuibhir P, Higgins S, et al. Cancer-related insomnia: wireless monitoring of sleep metrics in advanced disease. BMJ Support Palliat Care 2017; 7: A1. DOI: 10.1136/bmjspcare-2017-00133.2. [DOI] [Google Scholar]
- 41.Jurdi NE, Nock N, Fu P, et al. Physical activity and sleep measures using a fitness tracking device during hematopoietic cell transplantation: a pilot study. Transplant Cell Ther 2021; 27: 502.e501–502.e506. 10.1016/j.jtct.2021.02.015. [DOI] [PubMed] [Google Scholar]
- 42.Xu L, Zhou C, Ling Y, et al. Effects of short-term unsupervised exercise, based on smart bracelet monitoring, on body composition in patients recovering from breast cancer. Integr Cancer Ther 2021; 20: 15347354211040780. 10.1177/15347354211040780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Ferrante JM, Doose M, Bator A, et al. Virtual weight loss program for African-American breast cancer survivors: preliminary results. JCO 2017; 35: 163. DOI: 10.1200/JCO.2017.35.5_suppl.163. [DOI] [Google Scholar]
- 44.Nyrop KA, Deal AM, Choi SK, et al. Measuring and understanding adherence in a home-based exercise intervention during chemotherapy for early breast cancer. Breast Cancer Res Treat 2018; 168: 43–55. 10.1007/s10549-017-4565-1. [DOI] [PubMed] [Google Scholar]
- 45.Strother M, Koepsell K, Song L, et al. Financial incentives and wearable activity monitors to increase ambulation after cystectomy: a randomized controlled trial. Urol Oncol 2021; 39: 434.e431–434.e438. 10.1016/j.urolonc.2020.11.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Waller E, Rahman S, Sutton P, et al. Randomised controlled trial of patients undergoing prehabilitation with wearables versus standard of care before major abdominal cancer surgery. Colorectal Dis 2020; 22: 7. DOI: 10.1111/codi.15168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kong S, Lee JK, Kang D, et al. Comparing the effectiveness of a wearable activity tracker in addition to counseling and counseling only to reinforce leisure-time physical activity among breast cancer patients: a randomized controlled trial. Cancers (Basel) 2021; 13: 2692. DOI: 10.3390/cancers13112692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Lee BJ, Park YH, Lee JY, et al. Smartphone application versus pedometer to promote physical activity in prostate cancer patients. Telemed J E Health 2019; 25: 1231–1236. 10.1089/tmj.2018.0233. [DOI] [PubMed] [Google Scholar]
- 49.Uhm KE, Yoo JS, Chung SH, et al. Effects of exercise intervention in breast cancer patients: is mobile health (mHealth) with pedometer more effective than conventional program using brochure? Breast Cancer Res Treat 2017; 161: 443–452. 10.1007/s10549-016-4065-8. [DOI] [PubMed] [Google Scholar]
- 50.Low CA, Bovbjerg DH, Ahrendt S, et al. Fitbit step counts during inpatient recovery from cancer surgery as a predictor of readmission. Ann Behav Med 2018; 52: 88–92. 10.1093/abm/kax022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Wu J-M, Ho T-W, Chang Y-T, et al. Wearable-based mobile health app in gastric cancer patients for postoperative physical activity monitoring: focus group study. JMIR mHealth UHealth 2019; 7: e11989. 10.2196/11989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Palla I, Lorenzoni V, D’Isidoro C, et al. A new organizational integrated home-care model: an Italian pilot study. Value Health 2016; 19: A747–A748. [Google Scholar]
- 53.Sada YH, Poursina O, Zhou H, et al. Harnessing digital health to objectively assess cancer-related fatigue: the impact of fatigue on mobility performance. PloS one 2021; 16: e0246101. 10.1371/journal.pone.0246101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Timmerman JG, Tonis TM, Dekker-van Weering MGH, et al. Co-creation of an ICT-supported cancer rehabilitation application for resected lung cancer survivors: design and evaluation. BMC Health Serv Res 2016; 16: 155. 10.1186/s12913-016-1385-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Valle CG, Deal AM, Tate DF. Preventing weight gain in African American breast cancer survivors using smart scales and activity trackers: a randomized controlled pilot study. J Cancer Surviv 2017; 11: 133–148. 10.1007/s11764-016-0571-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Koontz BF, Levine E, McSherry F, et al. Increasing physical activity in cancer survivors through a text-messaging exercise motivation program (ICanSTEP). Supportive Care Cancer 2021; 29: 7339–7349. 10.1007/s00520-021-06281-y. [DOI] [PubMed] [Google Scholar]
- 57.Alberts NM, Leisenring WM, Flynn JS, et al. Wearable respiratory monitoring and feedback for chronic pain in adult survivors of childhood cancer: a feasibility randomized controlled trial from the childhood cancer survivor study. JCO Clin Cancer Inform 2020; 4: 1014–1026. 10.1200/CCI.20.00070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kim K-H, Lee J, Ahn CH, et al. Association between thyroid function and heart rate monitored by wearable devices in patients with hypothyroidism. Endocrinol Metab 2021; 36: 1121–1130. DOI: 10.3803/EnM.2021.1216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Rosenberg D, Kadokura EA, Bouldin ED, et al. Acceptability of Fitbit for physical activity tracking within clinical care among men with prostate cancer. AMIA Annual Symposium proceedings AMIA Symposium 2016; 2016: 1050-1059. [PMC free article] [PubMed]
- 60.Kerr J, Carlson J, Godbole S, et al. Improving hip-worn accelerometer estimates of sitting using machine learning methods. Med Sci Sports Exerc 2018; 50: 1518–1524. DOI: 10.1249/MSS.0000000000001578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Magalhaes B, Fernandes C, Santos C, et al. The use of mobile applications for managing care processes during chemotherapy treatments: a systematic review. Cancer Nurs 2021; 44: E339–E360. 10.1097/NCC.0000000000000823. [DOI] [PubMed] [Google Scholar]
- 62.Gresham G, Schrack J, Gresham LM, et al. Wearable activity monitors in oncology trials: current use of an emerging technology. Contemp Clin Trials 2018; 64: 13–21. DOI: 10.1016/j.cct.2017.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Huang Y-W, Wu C-S, Chuang C-K, et al. Real-time and label-free detection of the prostate-specific antigen in human serum by a polycrystalline silicon nanowire field-effect transistor biosensor. Anal Chem 2013; 85: 7912–7918. https://doi.org/10.1021/ac401610 s. [DOI] [PubMed] [Google Scholar]
- 64.Cudejko T, Button K, Willott J, et al. Applications of wearable technology in a real-life setting in people with knee osteoarthritis: a systematic scoping review. J Clin Med 2021; 10: 20211130. DOI: 10.3390/jcm10235645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Wells CI, Xu W, Penfold JA, et al. Wearable devices to monitor recovery after abdominal surgery: scoping review. BJS Open 2022; 6: 31. DOI: 10.1093/bjsopen/zrac031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Jo A, Coronel BD, Coakes CE, et al. Is there a benefit to patients using wearable devices such as fitbit or health apps on mobiles? A systematic review. Am J Med 2019; 132: 1394–1400 e1391. DOI: 10.1016/j.amjmed.2019.06.018. [DOI] [PubMed] [Google Scholar]
- 67.Huang Y, Upadhyay U, Dhar E, et al. A scoping review to assess adherence to and clinical outcomes of wearable devices in the cancer population. Cancers (Basel) 2022; 14: 20220913. DOI: 10.3390/cancers14184437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Lynch BM, Nguyen NH, Moore MM, et al. A randomized controlled trial of a wearable technology-based intervention for increasing moderate to vigorous physical activity and reducing sedentary behavior in breast cancer survivors: the ACTIVATE trial. Cancer 2019; 125: 2846–2855. 10.1002/cncr.32143. [DOI] [PubMed] [Google Scholar]
- 69.Lemanska A, Poole K, Griffin B, et al. Community pharmacy: an innovative way to deliver cancer survivorship interverntions. Support Care Cancer 2019; 27: S94–S95. DOI: 10.1007/s00520-019-04813-1. [DOI] [Google Scholar]
- 70.Huhn S, Axt M, Gunga HC, et al. The impact of wearable technologies in health research: scoping review. JMIR Mhealth Uhealth 2022; 10: e34384. DOI: 10.2196/34384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.What is GDPR, the EU’s new data protection law?, https://gdpr.eu/what-is-gdpr/ (accessed 26 July 2023).
- 72.Dhingra LS, Aminorroaya A, Oikonomou EK, et al. Use of wearable devices in individuals with or at risk for cardiovascular disease in the US, 2019 to 2020. JAMA Netw Open 2023; 6: e2316634. DOI: 10.1001/jamanetworkopen.2023.16634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Rising CJ, Gaysynsky A, Blake KD, et al. Willingness to share data from wearable health and activity trackers: analysis of the 2019 health information national trends survey data. JMIR Mhealth Uhealth 2021; 9: e29190. DOI: 10.2196/29190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Statistisches Bundesamt. Pressemitteilung Nr. 078 vom 22 Februar 2021, https://www.destatis.de/DE/Presse/Pressemitteilungen/2021/02/PD21_078_639.html (2021, accessed 28 August 2023).
- 75.Guo C, Ashrafian H, Ghafur S, et al. Challenges for the evaluation of digital health solutions-A call for innovative evidence generation approaches. NPJ Digit Med 2020; 3: 110. DOI: 10.1038/s41746-020-00314-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Hemkens LG. [Benefit assessment of digital health applications-challenges and opportunities]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2021; 64: 1269–1277. DOI: 10.1007/s00103-021-03413-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Gensorowsky D, Lampe D, Hasemann L, et al. “Alternative study designs” for the evaluation of digital health applications - a real alternative? Z Evid Fortbild Qual Gesundhwes 2021; 161: 33–41. DOI: 10.1016/j.zefq.2021.01.006. [DOI] [PubMed] [Google Scholar]
- 78.Hrynyschyn R, Prediger C, Stock C, et al. Evaluation methods applied to digital health interventions: what is being used beyond randomised controlled trials?-A scoping review. Int J Environ Res Public Health 2022; 19: 20220425. DOI: 10.3390/ijerph19095221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Kernebeck S, Kramer U, Redaelli Met al. et al. Bewerten, aber wie? – Kriterien für die evaluation und das reporting von Studien zu Gesundheits- und Medizin-apps. Monitor Versorgungsforschung 2021; 14: 69–75. DOI: 10.24945/mvf.02.21.1866-0533.2300. [DOI] [Google Scholar]
- 80.Schwartz D, Fischhoff B, Krishnamurti T, et al. The Hawthorne effect and energy awareness. Proc Natl Acad Sci U S A 2013; 110: 15242–15246. DOI: 10.1073/pnas.1301687110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Bade BC, Brooks MC, Nietert SB, et al. Assessing the correlation between physical activity and quality of life in advanced lung cancer. Integr Cancer Ther 2018; 17: 73–79. 10.1177/1534735416684016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Blair CK, Harding E, Wiggins C, et al. A home-based mobile health intervention to replace sedentary time with light physical activity in older cancer survivors: randomized controlled pilot trial. JMIR Cancer 2021; 7: e18819. 10.2196/18819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Carter SJ, Rogers LQ, Bowles HR, et al. Inverse association between changes in energetic cost of walking and vertical accelerations in non-metastatic breast cancer survivors. Eur J Appl Physiol 2019; 119: 2457–2464. DOI: 10.1007/s00421-019-04227-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Cereda E, Turrini M, Ciapanna D, et al. Assessing energy expenditure in cancer patients: a pilot validation of a new wearable device. JPEN 2007; 31: 502–507. [DOI] [PubMed] [Google Scholar]
- 85.Courneya KS, McNeely ML, Culos-Reed SN, et al. The Alberta moving beyond breast cancer (AMBER) cohort study: recruitment, baseline assessment, and description of the first 500 participants. BMC Cancer 2016; 16: 481. 10.1186/s12885-016-2534-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Dadhania S, Pakzad-Shahabi L, Mistry S, et al. Brainwear: longitudinal, objective assessment of physical activity in 42 HGG patients. Neuro Oncol 2021; 23: ii13. DOI: 10.1093/neuonc/noab180.043. [DOI] [Google Scholar]
- 87.Dambrosio N, Gannon J, Eager J, et al. Feasibility of monitoring of body temperature for patients undergoing stem cell transplant or intensive chemotherapy. JCO 2017; 35: 15. DOI: 10.1200/JCO.2017.35.15_suppl.e21630. [DOI] [Google Scholar]
- 88.De Cocker K, Charlier C, Van Hoof E, et al. Development and usability of a computer-tailored pedometer-based physical activity advice for breast cancer survivors. Eur J Cancer Care 2015; 24: 673–682. 10.1111/ecc.12225. [DOI] [PubMed] [Google Scholar]
- 89.Dorion V, Lambert L, Frazzi A, et al. A pilot study in the use of activity trackers for assessing response to palliative radiotherapy. Cureus 2017; 9: e1871. 10.7759/cureus.1871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Dreher N, Hadeler EK, Hartman SJ, et al. Fitbit usage in patients with breast cancer undergoing chemotherapy. Clin Breast Cancer 2019; 19: 443–449.e441. 10.1016/j.clbc.2019.05.005. [DOI] [PubMed] [Google Scholar]
- 91.Ferrante JM, Devine KA, Bator A, et al. Feasibility and potential efficacy of commercial mHealth/eHealth tools for weight loss in African American breast cancer survivors: pilot randomized controlled trial. Transl Behav Med 2018; 10: 938–948. DOI: 10.1093/tbm/iby124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Ferrante JM, Lulla A, Williamson JD, et al. Patterns of fitbit use and activity levels among African American breast cancer survivors during an eHealth weight loss randomized controlled trial. Am J Health Promot 2022; 36: 94–105. DOI: 10.1177/08901171211036700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Fink LS, Schinkoethe T, Wuerstlein R, et al. Substantial ninety-nine percent reduction of turnaround time for QTc diagnostics using smartphone-based telecardiology. JCO 2020; 38: 15. DOI: 10.1200/JCO.2020.38.15_suppl.e14155. [DOI] [Google Scholar]
- 94.Fischer I, Horneber M, Falge C, et al. Actigraphy and ecological momentary assessment of fatigue in cancer outpatients. Onkologie 2010; 33: 179–180. DOI: 10.1159/000321410. [DOI] [Google Scholar]
- 95.Flora C, Tyler J, Mayer C, et al. High-frequency temperature monitoring for early detection of febrile adverse events in patients with cancer. Cancer Cell 2021; 39: 1167–1168. DOI: 10.1016/j.ccell.2021.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Frensham LJ, Zarnowiecki DM, Parfitt G, et al. The experiences of participants in an innovative online resource designed to increase regular walking among rural cancer survivors: a qualitative pilot feasibility study. Supportive Care Cancer 2014; 22: 1923–1929. 10.1007/s00520-014-2177-4. [DOI] [PubMed] [Google Scholar]
- 97.Furtado S, Godfrey A, Del Din S, et al. Are accelerometer-based functional outcome assessments feasible and valid after treatment for lower extremity sarcomas? Clin Orthop Relat Res 2019; 478: 482–503. 10.1097/CORR.0000000000000883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Gallicchio R, Scapicchio D, Nardelli A, et al. Assessment of residual radioactivity by a comprehensive wireless, wearable device in thyroid cancer patients undergoing radionuclide therapy and comparison with the results of a home device: a feasibility study. IEEE J Transl Eng Health Med 2021; 9: 2700306. 10.1109/JTEHM.2020.3042118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Gell NM, Tursi A, Grover KW, et al. Female cancer survivor perspectives on remote intervention components to support physical activity maintenance. Supportive Care Cancer 2019; 28: 2185–2194. 10.1007/s00520-019-05038-y. [DOI] [PubMed] [Google Scholar]
- 100.Ghods A, Shahrokni A, Ghasemzadeh H, et al. Remote monitoring of the performance status and burden of symptoms of patients with gastrointestinal cancer via a consumer-based activity tracker: quantitative cohort study. JMIR Cancer 2021; 7: e22931. DOI: 10.2196/22931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Gresham G, Hendifar AE, Spiegel B, et al. Wearable activity monitors to assess performance status and predict clinical outcomes in advanced cancer patients. NPJ Digit Med 2018; 1: 27. 10.1038/s41746-018-0032-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Groarke JM, Richmond J, Mc Sharry J, et al. Acceptability of a mobile health behavior change intervention for cancer survivors with obesity or overweight: nested mixed methods study within a randomized controlled trial. JMIR mHealth uHealth 2021; 9: e18288. 10.2196/18288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Gupta A, Stewart T, Bhulani N, et al. Feasibility of wearable physical activity monitors in patients with cancer. JCO Clin Cancer Inform 2018; 2: 1–10. 10.1200/CCI.17.00152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Hardcastle SJ, Jimenez-Castuera R, Maxwell-Smith C, et al. Fitbit wear-time and patterns of activity in cancer survivors throughout a physical activity intervention and follow-up: exploratory analysis from a randomised controlled trial. PloS one 2020; 15: e0240967. 10.1371/journal.pone.0240967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Hartman SJ, Nelson SH, Weiner LS. Patterns of fitbit use and activity levels throughout a physical activity intervention: exploratory analysis from a randomized controlled trial. JMIR mHealth uHealth 2018; 6: e29. 10.2196/mhealth.8503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Helbrich H, Braun M, Hanusch C, et al. Congruence and trajectories of device-measured and self-reported physical activity during therapy for early breast cancer. Breast Cancer Res Treat 2021; 188: 351–359. 10.1007/s10549-021-06195-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Holden T, Hill M, Ward WH, et al. Feasibility of using fitness tracker to assess activity level and toxicities in colorectal cancer (CRC) patients (Pts). JCO 2020; 38: 4. DOI: 10.1200/JCO.2020.38.4_suppl.91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Hoogland AI, Bulls HW, Gonzalez BD, et al. Circadian rhythmicity as a predictor of quality of life in allogeneic hematopoietic cell transplant patients. J Pain Symptom Manage 2019; 57: 952–960.e951. 10.1016/j.jpainsymman.2019.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Innominato PF, Komarzynski S, Mohammad-Djafari A, et al. Clinical relevance of the first domomedicine platform securing multidrug chronotherapy delivery in metastatic cancer patients at Home: the inCASA European project. JMIR 2016; 18: e305. DOI: 10.2196/jmir.6303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Ip A, Churnetski MC, Calzada O, et al. The impact of a physical activity intervention can be accurately assessed by smart watches in patients completing autologous stem cell transplantation for lymphoma or multiple myeloma: results of a feasibility study. Blood 2018; 132. DOI: 10.1182/blood-2018-99-119757. [DOI] [Google Scholar]
- 111.Izmailova E, Huang C, Cantor M, et al. Daily step counts to predict hospitalizations during concurrent chemoradiotherapy for solid tumors. JCO 2019; 37. DOI: 10.1200/JCO.2019.37.27-suppl.293. [DOI] [Google Scholar]
- 112.Izmailova ES, Cohen A, Kabarriti R, et al. Daily step counts to predict hospitalizations during chemoradiotherapy for head and neck cancer. JCO 2021; 39. DOI: 10.1200/JCO.2021.39.15_suppl.1571. [DOI] [Google Scholar]
- 113.Jeffery E, Lee YG, McVeigh J, et al. Feasibility of objectively measured physical activity and sedentary behavior in patients with malignant pleural effusion. Supportive Care Cancer 2017; 25: 3133–3141. 10.1007/s00520-017-3721-9. [DOI] [PubMed] [Google Scholar]
- 114.Jonker L, Plas M, de Bock GH, et al. Implementing eHealth for postoperative home monitoring of elderly cancer patients: a feasibility study. Eur J Surg Oncol 2020; 46: e159. DOI: 10.1016/j.ejso.2019.11.420. [DOI] [Google Scholar]
- 115.Jonker LT, Plas M, de Bock GH, et al. Remote home monitoring of older surgical cancer patients: perspective on study implementation and feasibility. Ann Surg Oncol 2021; 28: 67–78. 10.1245/s10434-020-08705-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Jovanovic JL, Hughes DC, Baum GP, et al. Accelerometry and self-report in sedentary populations. Am J Health Behav 2011; 35: 71–80. DOI: 10.5993/AJHB.35.1.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Khetrapal P, Williams N, Tan M, et al. The use of fitness tracker in monitoring functional activity of patients undergoing radical cystectomy for bladder cancer: a feasibility report and analysis as part of the multi-centre randomised iROC trial. J Urol 2018; 199: e950. DOI: 10.1016/j.juro.2018.02.2282. [DOI] [Google Scholar]
- 118.Kim KN, Shah N, Grewal AS, et al. Effect of activity monitoring on quality of life in patients with gastrointestinal cancer undergoing chemoradiation. JCO 2021; 39. DOI: 10.1200/JCO.2021.39.15_suppl.e18671. [DOI] [Google Scholar]
- 119.Komarzynski S, Huang Q, Levi FA, et al. The day after: correlates of patient-reported outcomes with actigraphy-assessed sleep in cancer patients at home (inCASA project). Sleep 2019; 42: 146. DOI: 10.1093/sleep/zsz146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Laccetti AL, Tidwell RS, Sheth NP, et al. Remote patient monitoring using smart phone derived patient reported outcomes and Fitbit data to enable longitudinal predictive modeling in prostate cancer: feasibility results and lessons on platform development. JCO 2019; 37. DOI: 10.1200/JCO.2019.37.15_suppl.e18068. [DOI] [Google Scholar]
- 121.LeBaron V, Alam R, Blackhall L, et al. Using smart health to monitor cancer pain. Support Care Cancer 2021; 29: S45. DOI: 10.1007/s00520-021-06285-8. [DOI] [Google Scholar]
- 122.Liu L, Natarajan L, He F, et al. Actigraphy-based pre-treatment nap time predicts quality of life during chemotherapy in breast cancer patients. Sleep 2009; 32: A323. [Google Scholar]
- 123.Low CA, Dey AK, Ferreira D, et al. Using passive smartphone and wearable sensors to estimation patient-reported stress and symptoms during chemotherapy. Psychosom Med 2018; 80: A138. DOI: 10.1097/PSY.0000000000000578. [DOI] [Google Scholar]
- 124.Low CA, Li M, Vega J, et al. Digital biomarkers of symptom burden self-reported by perioperative patients undergoing pancreatic surgery: prospective longitudinal study. JMIR Cancer 2021; 7: e27975. 10.2196/27975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Lozano-Lozano M, Cantarero-Villanueva I, Martin-Martin L, et al. A mobile system to improve quality of life via energy balance in breast cancer survivors (BENECA mHealth): prospective test-retest quasiexperimental feasibility study. JMIR mHealth uHealth 2019; 7: e14136. 10.2196/14136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Maddocks M, Byrne A, Johnson CD, et al. Physical activity level as an outcome measure for use in cancer cachexia trials: a feasibility study. Support Care Cancer 2010; 18: 1539–1544. DOI: 10.1007/s00520-009-0776-2. [DOI] [PubMed] [Google Scholar]
- 127.Martin CL, Tate DF, Valle CG. Nonadherence to daily self-weighing and activity tracking is associated with weight fluctuations among African American breast cancer survivors. PLoS one 2018; 13: e0199751. 10.1371/journal.pone.0199751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Miropolsky EM, Scott Baker K, Abbey-Lambertz M, et al. Participant perceptions on a fitbit and Facebook intervention for young adult cancer survivors: a qualitative study. J Adolesc Young Adult Oncol 2020; 9: 410–417. 10.1089/jayao.2019.0072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Nguyen NH, Hadgraft NT, Moore MM, et al. A qualitative evaluation of breast cancer survivors’ acceptance of and preferences for consumer wearable technology activity trackers. Supportive Care Cancer 2017; 25: 3375–3384. 10.1007/s00520-017-3756-y. [DOI] [PubMed] [Google Scholar]
- 130.Ohri N, Kabarriti R, Bodner WR, et al. Continuous activity monitoring during concurrent chemoradiation therapy. Int J Radiat Oncol 2016; 96: E522. DOI: 10.1016/j.ijrobp.2016.06.1937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Otte JL, Payne JK, Carpenter JS. Nighttime variability in wrist actigraphy. J Nurs Meas 2011; 19: 105–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Pavic M, Klaas V, Theile G, et al. Feasibility and usability aspects of continuous remote monitoring of health status in palliative cancer patients using wearables. Oncology 2019; 98: 386–395. 10.1159/000501433. [DOI] [PubMed] [Google Scholar]
- 133.Pavic M, Klaas V, Theile G, et al. Mobile health technologies for continuous monitoring of cancer patients in palliative care aiming to predict health status deterioration: a feasibility study. J Palliat Med 2020; 23: 678–685. 10.1089/jpm.2019.0342. [DOI] [PubMed] [Google Scholar]
- 134.Peterson SK, Basen-Engquist K, Demark-Wahnefried W, et al. Feasibility of using home-based mobile sensors for remote patient monitoring in cancer care and prevention. JCO 2014; 32. DOI: 10.1200/jco.2014.32.15_suppl.9585. [DOI] [Google Scholar]
- 135.Peterson SK, Garden AS, Shinn EH, et al. Using mobile and sensor technology to identify early dehydration risk in head and neck cancer patients undergoing radiation treatment: impact on symptoms. JCO 2018; 36. DOI: 10.1200/JCO.2018.36.15-suppl.6063. [DOI] [Google Scholar]
- 136.Pfister T, Matthews CE, Wang Q, et al. Comparison of two accelerometers for measuring physical activity and sedentary behaviour. BMJ Open Sport Exerc Med 2017; 3: e000227. 10.1136/bmjsem-2017-000227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Quintiliani LM, Mann DM, Puputti M, et al. Pilot and feasibility test of a mobile health-supported behavioral counseling intervention for weight management among breast cancer survivors. JMIR Cancer 2016; 2: e4. DOI: 10.2196/cancer.5305. [DOI] [PubMed] [Google Scholar]
- 138.Rossi A, Frechette L, Miller D, et al. Acceptability and feasibility of a fitbit physical activity monitor for endometrial cancer survivors. Gynecol Oncol 2018; 149: 470–475. 10.1016/j.ygyno.2018.04.560. [DOI] [PubMed] [Google Scholar]
- 139.Schuler T, Hruby G, Kwong C, et al. Quantitative results from the tracker study: feasibility of real-time activity and weight monitoring during routine care of oncology patients. J Med Imaging Radiat Oncol 2019; 63: 100. [Google Scholar]
- 140.Sugiyama Y, Naiki T, Tasaki Y, et al. Effectiveness of continuous monitoring by activity tracker of patients undergoing chemotherapy for urothelial carcinoma. Cancer Treat Res Commun 2020; 25: 100245. 10.1016/j.ctarc.2020.100245. [DOI] [PubMed] [Google Scholar]
- 141.Skipworth RJ, Stene GB, Dahele M, et al. Patient-focused endpoints in advanced cancer: criterion-based validation of accelerometer-based activity monitoring. Clin Nutr 2011; 30: 812–821. DOI: 10.1016/j.clnu.2011.05.010. [DOI] [PubMed] [Google Scholar]
- 142.Slade AD, Cardinal JR, Martin CR, et al. Feasibility of wearable activity trackers in cystectomy patients to monitor for postoperative complications. Curr Urol 2021; 15: 209–213. DOI: 10.1097/CU9.0000000000000030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Tipps M, Hultman M, Banerji N, et al. Validation of readiband™ actigraph and associated sleep/wake classification algorithms in predicting cancer-related fatigue in high grade glioblastoma. Neuro-Oncology 2019; 21: vi132. DOI: 10.1093/neuonc/noz175.551. [DOI] [Google Scholar]
- 144.Torrente M, Franco F, Collazo Lorduy A, et al. Detecting elevated risk of diminished quality of life among lung cancer patients by using wearable devices. J Thorac Oncol 2021; 16: S1130–S1131. DOI: 10.1016/j.jtho.2021.08.561. [DOI] [Google Scholar]
- 145.Tramontano A, Scippacercola F, Magliulo M. Estimating quality of life variation using BLE wearable devices on android based architecture. Int J Radiat Oncol Biol Phys 2018; 102: e286. DOI: 10.1016/j.ijrobp.2018.07.913. [DOI] [Google Scholar]
- 146.Trinh L, Arbour-Nicitopoulos KP, Sabiston CM, et al. RiseTx: testing the feasibility of a web application for reducing sedentary behavior among prostate cancer survivors receiving androgen deprivation therapy. Int J Behav Nutr Phys Act 2018; 15: 49. 10.1186/s12966-018-0686-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Ungar N, Sieverding M, Weidner G, et al. A self-regulation-based intervention to increase physical activity in cancer patients. Psychol Health Med 2016; 21: 163–175. 10.1080/13548506.2015.1081255. [DOI] [PubMed] [Google Scholar]
- 148.Vallance JK, Boyle T, Courneya KS, et al. Accelerometer-assessed physical activity and sedentary time among colon cancer survivors: associations with psychological health outcomes. J Cancer Surviv 2015; 9: 404–411. 10.1007/s11764-014-0409-8. [DOI] [PubMed] [Google Scholar]
- 149.Van Blarigan EL, Chan H, Van Loon K, et al. Self-monitoring and reminder text messages to increase physical activity in colorectal cancer survivors (Smart Pace): a pilot randomized controlled trial. BMC Cancer 2019; 19: 218. 10.1186/s12885-019-5427-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Verma N, Haji-Abolhassani I, Ganesh S, et al. A novel wearable device for continuous temperature monitoring fever detection. IEEE J Transl Eng Health Med 2021; 9: 9490248. DOI: 10.1109/JTEHM.2021.3098127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Waliany S, Dieli-Conwright CM, Frankel PH, et al. Validation and feasibility of a caloric expenditure measuring device in women with early-stage breast cancer. Supportive Care Cancer 2014; 22: 2329–2336. 10.1007/s00520-014-2212-5. [DOI] [PubMed] [Google Scholar]
- 152.Ward WH, Meeker C, Hill M, et al. Feasibility of using a fitness tracker to assess activity level and toxicity in colorectal cancer patients. Ann Surg Oncol 2019; 26: S109. DOI: 10.1245/s10434-019-07174-5. [DOI] [Google Scholar]
- 153.Wong K, Shen J, Ramezani R, et al. A pilot study of a wearable monitoring system as an adjunct to geriatric assessment in older adults with cancer. JCO 2020; 38: 15. DOI: 10.1200/JCO.2020.38.15_suppl.2062. [DOI] [Google Scholar]
- 154.Wright AA, Raman N, Staples P, et al. The HOPE pilot study: harnessing patient-reported outcomes and biometric data to enhance cancer care. JCO Clin Cancer Inform 2018; 2: 1–12. 10.1200/CCI.17.00149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Wu CY, Tibbitts D, Beattie Z, et al. Continuous unobtrusive assessment of meaningful change in older adults with cancer: the pacific aging & cancer study collaborative-PACS collaborative. JCO 2021; 39: 1569–1569. DOI: 10.1200/JCO.2021.39.15_suppl.1569. [DOI] [Google Scholar]
- 156.Zahiri M, Chen KM, Zhou H, et al. Using wearables to screen motor performance deterioration because of cancer and chemotherapy-induced peripheral neuropathy (CIPN) in adults - Toward an early diagnosis of CIPN. J Geriatr Oncol 2019; 10: 960–967. DOI: 10.1016/j.jgo.2019.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supplemental material, sj-docx-1-dhj-10.1177_20552076241233998 for Application of wearables for remote monitoring of oncology patients: A scoping review by Katharina Cloß, Marlo Verket, Dirk Müller-Wieland, Nikolaus Marx, Katharina Schuett, Edgar Jost, Martina Crysandt, Fabian Beier, Tim H Brümmendorf, Guido Kobbe, Julia Brandts and Malte Jacobsen in DIGITAL HEALTH
Supplemental material, sj-xlsx-2-dhj-10.1177_20552076241233998 for Application of wearables for remote monitoring of oncology patients: A scoping review by Katharina Cloß, Marlo Verket, Dirk Müller-Wieland, Nikolaus Marx, Katharina Schuett, Edgar Jost, Martina Crysandt, Fabian Beier, Tim H Brümmendorf, Guido Kobbe, Julia Brandts and Malte Jacobsen in DIGITAL HEALTH
Supplemental material, sj-xlsx-3-dhj-10.1177_20552076241233998 for Application of wearables for remote monitoring of oncology patients: A scoping review by Katharina Cloß, Marlo Verket, Dirk Müller-Wieland, Nikolaus Marx, Katharina Schuett, Edgar Jost, Martina Crysandt, Fabian Beier, Tim H Brümmendorf, Guido Kobbe, Julia Brandts and Malte Jacobsen in DIGITAL HEALTH
Supplemental material, sj-xlsx-4-dhj-10.1177_20552076241233998 for Application of wearables for remote monitoring of oncology patients: A scoping review by Katharina Cloß, Marlo Verket, Dirk Müller-Wieland, Nikolaus Marx, Katharina Schuett, Edgar Jost, Martina Crysandt, Fabian Beier, Tim H Brümmendorf, Guido Kobbe, Julia Brandts and Malte Jacobsen in DIGITAL HEALTH
Supplemental material, sj-xlsx-5-dhj-10.1177_20552076241233998 for Application of wearables for remote monitoring of oncology patients: A scoping review by Katharina Cloß, Marlo Verket, Dirk Müller-Wieland, Nikolaus Marx, Katharina Schuett, Edgar Jost, Martina Crysandt, Fabian Beier, Tim H Brümmendorf, Guido Kobbe, Julia Brandts and Malte Jacobsen in DIGITAL HEALTH
Supplemental material, sj-xlsx-6-dhj-10.1177_20552076241233998 for Application of wearables for remote monitoring of oncology patients: A scoping review by Katharina Cloß, Marlo Verket, Dirk Müller-Wieland, Nikolaus Marx, Katharina Schuett, Edgar Jost, Martina Crysandt, Fabian Beier, Tim H Brümmendorf, Guido Kobbe, Julia Brandts and Malte Jacobsen in DIGITAL HEALTH
Supplemental material, sj-xlsx-7-dhj-10.1177_20552076241233998 for Application of wearables for remote monitoring of oncology patients: A scoping review by Katharina Cloß, Marlo Verket, Dirk Müller-Wieland, Nikolaus Marx, Katharina Schuett, Edgar Jost, Martina Crysandt, Fabian Beier, Tim H Brümmendorf, Guido Kobbe, Julia Brandts and Malte Jacobsen in DIGITAL HEALTH
Supplemental material, sj-pptx-8-dhj-10.1177_20552076241233998 for Application of wearables for remote monitoring of oncology patients: A scoping review by Katharina Cloß, Marlo Verket, Dirk Müller-Wieland, Nikolaus Marx, Katharina Schuett, Edgar Jost, Martina Crysandt, Fabian Beier, Tim H Brümmendorf, Guido Kobbe, Julia Brandts and Malte Jacobsen in DIGITAL HEALTH




