Next Article in Journal
Preclinical Evaluation of Novel Tyrosine-Kinase Inhibitors in Medullary Thyroid Cancer
Previous Article in Journal
Omega-3 Polyunsaturated Fatty Acids, Gut Microbiota, Microbial Metabolites, and Risk of Colorectal Adenomas
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Scoping Review to Assess Adherence to and Clinical Outcomes of Wearable Devices in the Cancer Population

1
Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110, Taiwan
2
Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
3
Graduate Institute of Biomedical Informatics, College of Medical Sciences and Technology, Taipei Medical University, Taipei 106, Taiwan
4
International Center for Health Information Technology, College of Medical Science and Technology, Taipei Medical University, Taipei 106, Taiwan
5
Division of Colorectal Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan
6
Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
7
School of Gerontology and Long-Term Care, College of Nursing, Taipei Medical University, Taipei 110, Taiwan
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(18), 4437; https://doi.org/10.3390/cancers14184437
Submission received: 8 August 2022 / Revised: 6 September 2022 / Accepted: 8 September 2022 / Published: 13 September 2022

Abstract

:

Simple Summary

The use of wearable devices in clinical care is gaining popularity among cancer patients. The COVID-19 pandemic highlighted the value of wearable devices for monitoring health. Wearable devices are used to record and monitor real-time data like physical activity, sleep metrics, and heart rate variables. The use of wearable devices can directly impact clinical decision-making. There are few pieces of evidence that prove that wearable could improve the quality of patient care while reducing the cost of care, such as remote health monitoring. The generated big data by the wearable device is both a challenge and an opportunity. Researchers can apply artificial intelligence and machine learning techniques to improve wearable devices and their usage among cancer patients. In this scoping review, we assessed the adherence to clinical outcomes of wrist-worn wearable devices in the cancer population.

Abstract

The use of wearable devices (WDs) in healthcare monitoring and management has attracted increasing attention. A major problem is patients’ adherence and acceptance of WDs given that they are already experiencing a disease burden and treatment side effects. This scoping review explored the use of wrist-worn devices in the cancer population, with a special focus on adherence and clinical outcomes. Relevant articles focusing on the use of WDs in cancer care management were retrieved from PubMed, Scopus, and Embase from 1 January 2017 to 3 March 2022. Studies were independently screened and relevant information was extracted. We identified 752 studies, of which 38 met our inclusion criteria. Studies focused on mixed, breast, colorectal, lung, gastric, urothelial, skin, liver, and blood cancers. Adherence to WDs varied from 60% to 100%. The highest adherence was reported in the 12-week studies. Most studies focused on physical activity, sleep analysis, and heart vital signs. Of the 10 studies that described patient-reported outcomes using questionnaires and personal interviews, 8 indicated a positive correlation between the patient-reported and wearable outcomes. The definitions of the outcome measures and adherence varied across the studies. A better understanding of the intervention standards in terms of the clinical outcomes could improve adherence to wearables.

1. Introduction

Considerable advancements have been made in the fields of biosensors and artificial intelligence, particularly in terms of their use for the detection and management of chronic illnesses [1]. Wearable devices (WDs) are electronic devices that can be easily worn on the human body to capture real-time healthcare data using receptors and transducers attached to the device [2]. According to Deloitte Global’s prediction analysis, 440 million individuals will be using WDs by the end of 2024, with more healthcare providers recommending their use and more consumers becoming more comfortable with using them in their daily lives [3]. WDs include any device that can be worn on the human body including wristwatches, glasses, chest straps, rings, and prosthetic sockets [4]. WDs are mainly used to track individuals’ daily activities, including sleep quality, physical activity, and heart rate [4,5]. The evolution of smart WDs is being accelerated by improved sensors, artificial intelligence, and advanced machine-learning algorithm–based technologies [6]. For example, few watches that are currently available have optical sensors that can collect real-time data on blood physiology and blood pressure through photoplethysmography [7].
Several studies have reported the use of wearables in the healthcare field with promising outcomes [8]. WDs have revolutionized the healthcare system and have reduced the load of hospitals by providing reliable information in a timely manner [8,9]. Various WDs with a wide range of types and employability are available for data collection [10]. The rich information collected by WDs can assist healthcare professionals in tracking a person’s health status (sleep quality, healthy posture, cognitive decline, and even early warning signs of infection and inflammation) [8,9,10]. Chronic health conditions result in a high financial and emotional burden on patients and their families [11]. In response to COVID-19, many healthcare professionals reengineered their pathways to promote “care in place,” which allows patients to track their health and participate in the self-care system [12].
Wearables in oncology may provide new, vital information on a patient’s health status (heart rate, blood pressure, activity level, sleep quality, and behavioral activity), which can improve cancer care management [10].
Cancer is the second leading cause of death, and treatment is costly; however, using wearable devices can be cost-effective as they require personalized and flexible patient treatment plans [13,14]. Patients can also track their own data while continuing their normal routines while their data are being transferred to the clinic; this approach in oncology is a work-smarter approach and, more importantly, provides a general improvement in the quality of life [15,16]. Few studies provided evidence regarding the effectiveness of WDs in improving the treatment outcomes of patients with cancer [17].
To understand the potential use of WDs in cancer management, the effect of an intervention on or the role of WDs in clinical outcomes should be investigated [10]. Evaluating patients’ outcomes and their adherence to WDs, as well as defining the criteria and valid data, can aid in introducing WDs into clinical practice [18]. Although a consensus and guidelines for designing and reporting trials that use wearables as a component of the intervention are lacking, this area of research is gaining attention [19] because WDs play a major role in healthcare management [8]. This scoping review examined the effectiveness of WDs in assisting patients in their care, particularly in cancer treatment. In addition, this review evaluated the effectiveness and feasibility of this type of intervention. The goal of this review is to explore the use of WDs in patients with different types of cancers given the increasing number of WD users.

2. Methods

2.1. Search Strategy

The search strategy was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analysis guidelines. A comprehensive literature search was conducted to identify scientific studies that analyzed WD-based interventions targeting patients with cancer and cancer survivors. We searched for studies published between 1 January 2017 and 7 March 2022, in the electronic databases PubMed, Scopus, and Embase. The search for studies was conducted on 9 March 2022. The search was conducted using MeSH (medical subject headings) such as wearables and cancer/devices and cancer/telemonitoring and cancer. The search was limited to articles published in English. The data collected were reviewed by the two authors of this study. If any reviewer considered an article to be potentially significant, the full text of the article was retrieved. In the case of a disagreement on a particular article, a third reviewer chose the article based on the exclusion and inclusion criteria.

2.2. Criteria for the Inclusion of Studies

We included original research articles that were published in English and met the following criteria: (1) included cancer survivors and patients with cancer undergoing treatment; (2) focused on preventive care or health monitoring (physical activity, behavioral activity, and quality of life); (3) used a smartwatch or other types of wrist-worn WDs for assessing health; and (4) were designed as a randomized controlled trial (RCT), prospective clinical trial, quasi-experimental study, feasibility study, observational study, or pilot study.
We excluded studies that (1) focused on the prediction of cancer; (2) used telecommunication technologies, such as websites, telephones, and mobile applications alone; (3) did not involve the use of wrist-worn wearables or focused on some other health problems; (4) did not include the intervention as the primary focus; and (5) were review articles, trial protocols, trial registrations, conference papers, book chapters, notes, brief reports, letters, editorials, or case studies or were published in a language other than English.

2.3. Study Selection and Data Extraction

We initially screened the titles and abstracts of articles that met the inclusion criteria and could not be rejected with certainty. The references of the studies that met the inclusion criteria were manually searched to identify relevant research articles. Three researchers independently reviewed the full texts of the articles and extracted the following data from each included article: characteristics of the study (country of origin, sample size, authors, study design, study purpose, and publication year); patient characteristics (mean age, sex percentage, cancer type, and health status); intervention characteristics (total study duration including the follow-up period, intervention duration, and type of wearable and tools used); and study focus (behavioral health monitoring and preventive care). For this scoping review, data on adherence to wearables were extracted according to the different criteria mentioned in each article. Adherence was measured in terms of the percentage of valid wear time among patients or the percentage of total patients who completed the trial and the percentage of total evaluable days [20]. Furthermore, we categorized the studies based on the methods and outcomes (Figure 1).

2.4. Adherence Analysis

Adherence analysis results were extracted from the studies and analyzed according to the different criteria mentioned in the selected articles. The majority of included studies evaluated adherence through the completeness of the data collection, that is, the percentage of recruited patients who completed the study. The remaining studies had specific criteria for the adherence evaluation that are mentioned in results section. We used SPSS for visualization purposes in order to graphically represent adherence to wearable devices by the duration of the study intervention.

2.5. Outcomes and Analysis

This review examined the effectiveness of WDs in health monitoring (symptom analysis/recovery assessment/physical activity, behavioral activity, quality of life, and preventive care) among patients with cancer undergoing treatment (chemotherapy, radiotherapy, or pre- and post-surgery treatment) and cancer survivors. The primary outcome was adherence to wearables according to the different criteria of each study. The secondary outcomes included the wearable, patient-reported, and clinical outcomes of the intervention. All data are presented in a descriptive manner.

2.6. Ethical Consideration

This review did not require the approval of any national or institutional boards.

3. Results

3.1. Study Selection

A total of 752 articles were retrieved from Embase, Scopus, and PubMed. After the removal of the duplicates, 473 articles were evaluated. Finally, of the 473 articles, the full texts of 102 relevant articles were reviewed. At this stage, the articles were screened based on their study characteristics, including the type of wearables and their usage, and the primary focus of the study. The exclusion criteria are mentioned in the methodology and presented in Figure 1. Finally, 38 studies that met the inclusion criteria were included in this scoping review.

3.2. Study Characteristics

Among the 38 studies, 19 were conducted in North America (the United States and Canada), 6 in Europe (Ireland, Germany, the United Kingdom, the Netherlands, France, and Switzerland), 9 in Asia (Taiwan, Central China, India, Japan, and South Korea), and 4 in Oceania (Australia; Table 1).
Among the 38 studies, 12 were designed as RCTs, 7 as feasibility studies, 5 as observational studies, 4 as pilot studies, 3 as cohort studies, 2 as nonrandomized controlled trials, 2 as usability studies, 1 as a utility study, and 1 as a group and qualitative study (Table 1). The included studies had different time intervals for the intervention and follow-up durations. The minimum and maximum intervention durations were 1 and 52 weeks, respectively. All the studies had the same follow-up period of 12 weeks (Table 1).

3.3. Characteristics of Research Participants

Most of the studies included patients with different types of cancer, followed by those with breast, colorectal, lung, gastric, urothelial, skin, liver, and blood cancers. Of the 38 studies, 15 focused on cancer survivors and 23 on patients receiving chemotherapy or radiotherapy. The number of participants in the studies ranged from 8 to 555 and their mean age ranged from 17 to 73 years (Table 1).

3.4. Measurement Tools

In terms of measurement tools, the studies included in this scoping review used subjective self-reported questionnaires, WDs, and mobile applications to track the health status of the participants. Only four studies conducted personal interviews to analyze participants’ adherence to WDs and their study experiences [21,33,40,57]. Furthermore, 10 studies evaluated the correlation between the outcomes of WDs and questionnaires to validate the effectiveness of using WDs in patients with cancer [27,31,34,36,37,48,49,52,54,56]. Only two studies used an e-diary and a physical diary along with a WD to track the health status of the participants [31,36].

3.5. Major Study Focus

The studies included in this review mainly focused on physical activity, sleep quality, quality of life, unplanned healthcare encounters, moderate-to-vigorous physical activity (MVPA), sedentary behavior, and symptom burden. Only one study focused on preventive care management (Table 1).

3.6. Intervention Methods

The studies included in this review article employed different interventional approaches to improve the clinical outcomes of the participants. Most of the studies used a mobile application to provide the intervention [22,34,35,39,40,46,53,54,57]. Of the 38 studies, 3 provided the intervention by sending text messages [22,45,57], and 4 conducted a general group session or included a virtual support group [39,41,44]. Furthermore, 7 studies used in-app chat services; conducted behavioral counseling sessions, coaching programs, and group phone calls; and provided health education [21,22,23,24,30,38,42,46]. The remaining studies included a few other approaches (Table 1).

3.7. Interventional Outcomes

3.7.1. Adherence

The adherence rate was calculated for both the follow-up and intervention periods by determining the average time of the intervention across the 38 studies. Of the 38 studies in this review, 26 examined adherence in terms of the completeness of the data collection, and the remaining 12 studies followed different criteria to examine adherence (Table 2). Of the 38 studies, 6 determined the acceptability of WDs by conducting qualitative personal interviews and evaluating the usage of the device on different days during the study [24,29,45,47,52,57]. Seven studies determined feasibility and four studies evaluated the retention of WDs according to the same criteria used for determining acceptability [4,20,21,23,24,39,41,42,45,47,52,55]. Only one study focused on adherence based on the completion of an exercise program [55].
The adherence during the intervention periods was visualized graphically using SPSS. Figure 2 shows the adherence rate to the exhibited intervention duration. All of the studies were divided into 11 segments based on the duration of each study’s intervention. The average adherence percentage was calculated for each segment. The intervention-based segments included studies with weeks 1 [48,49], 2 [27,55], 3 [50,52,57], 4 [36,37,53], 7 [28], 8 [32,33,34], 10 [24,30,41], 12 [20,21,23,25,26,38,39,40,42,43,44,45,46,47,54,56], 17 [29], 24 [22,31] and 43 [35]. In the graph, (1.84) indicates the studies grouped in this segment that had a one-week intervention with an average adherence of 84%. According to our analysis, 16 out of 38 studies used a 12-week intervention period with an average adherence of 86% (Figure 2) [20,21,23,25,26,38,39,40,42,43,44,45,46,47,54,56]. The follow-up periods in the reported studies were calculated by subtracting the intervention duration from the total study duration. There are only five studies with a 12-week follow-up period, accounting for 24 weeks of total study duration. These studies showed an average adherence rate of 91% [20,21,25,38,43]. Studies with an intervention period of 4 or 7 weeks also observed high adherence to WDs (i.e., 92% and 93%, respectively) [28,36,37,53] (Figure 2). There is one unique study that enrolled patients across four seasons of the year. This study showed a 100% adherence rate throughout the 52-week study duration [52]. The adherence outcomes are summarized in Table 2.

3.7.2. Clinical Outcomes

Wearable technology can directly affect clinical decision making and improve the quality of patient care while reducing the cost of care including that of patient rehabilitation outside of hospitals. The studies in this review focused on physical activity, sleep quality, quality of life, unplanned healthcare encounters, MVPA, sedentary behavior, and symptom burden.
Most of the studies in this review evaluated the feasibility of and adherence to WDs. Of the 38 studies, only 10 determined the correlation between patient-reported and objective outcomes; these studies reported that the use of WDs significantly improved clinical outcomes [27,31,34,36,37,48,49,52,54,56]. In addition, other studies demonstrated significant improvements in clinical outcomes; however, these studies did not observe a correlation between patient-reported and clinical outcomes. Of the 38 studies, 12 included patients with different types of cancer, 6 included cancer survivors, and 11 included patients with breast cancer. Of the 11 studies, 5 focused on patients receiving chemotherapy or radiotherapy. Moreover, of the 38 studies, 5 included patients with colorectal cancer, 4 included patients with lung cancer, and 2 included patients with gastric cancer. Furthermore, 4 of the 38 studies included patients with urothelial carcinoma, skin cancer, blood cancer, and liver cancer. The clinical outcomes are summarized in Table 3.

4. Discussion

4.1. Summary and Findings

The use of WDs as gadgets for tracking daily activities, particularly physical activity, has become widespread [59]. WDs are also used for patient and disease management. To the best of our knowledge, this is the first scoping review to examine adherence to WDs in the cancer population. We performed a scoping review of articles evaluating the heterogenous use of wrist-worn devices in patients with cancer. By using our search strategy, we initially retrieved 752 studies, of which 38 were finally included in this scoping review. Most of the studies in the review included patients with mixed cancer types, followed by those with breast cancer. In addition, other studies included cancer survivors and patients with cancer receiving chemotherapy or radiotherapy. The included studies were designed as RCTs and nonrandomized controlled, observational, feasibility/pilot, cohort, and group studies. The sample sizes ranged from 8 to 555. All the studies included used either subjective questionnaires or mobile applications along with a WD. The intervention duration varied from 1 to 52 weeks. Furthermore, 89% (34/38) of the studies evaluated physical activity as the clinical outcome. The outcomes of using WDs varied among the studies based on the intervention program and the usage rate of WDs. Of the 38 studies, 10 compared WD outcomes and patient-reported outcomes (determined using subjective questionnaires) and examined whether the use of WDs improved clinical outcomes. Of the 10 studies, 8 reported that using WDs considerably improved clinical outcomes. The study designs and outcomes varied among the included studies.

4.2. eHealth Tools for Cancer Care

Other studies explored the use of eHealth tools involving patient self-reporting of medication and healthcare management and their effects on the health of users [60]. More high-quality studies are warranted before the standard implementation of eHealth tools. In oncology, patients are increasingly required to manage their own illnesses; thus, WDs can be a valuable tool in the management of cancer during therapy [61]. However, technical and clinical adherence to such devices are essential aspects that should be explored because they determine the usage rate of devices among patients [62]. In this scoping review, we evaluated the adherence to WDs that was reported in the included studies. The adherence rate can be calculated using various factors including wear time, the number of patients using the device, data collection while wearing the device, and the number of evaluable days [20]. A wide range of data were collected during the days on which the device was worn and no data were collected on the days on which patients missed wearing the device. Because of the variations in the data collection and adherence, the effectiveness of WDs for the health management of patients with cancer remains unclear. Whether patients wear the device when they feel comfortable based on the provided intervention should be evaluated. Thus, before designing WD-related studies or large interventional studies, we need to define the criteria and set a fixed wearable time to understand the adherence to devices.

4.3. Strength and Limitations

To the best of our knowledge, this scoping review is the first to determine adherence to specific wrist-worn devices. However, the choice of a WD and its outcomes are crucial. The selection of appropriate clinical variables for measurement is crucial based on the purpose of WD use when incorporated into patients’ daily routines. The outcomes must be based on the type of disease. We agree with other researchers that monitoring specific outcomes is crucial because unnecessary outcomes would not substantially affect patients’ treatments. Most of the studies investigated the effectiveness of WDs for improving physical activity and quality of life; however, this may be attributed to the increasing knowledge of fitness and increased physical training. Furthermore, the secondary objective of most of the studies was to examine the clinical outcomes of WDs in terms of healthcare management. Only 10 studies reported a positive correlation between the use of WDs and patient-reported outcomes.
Since each article used its own definition, it was not possible for us to compare the adherence data of all studies, which is one of the significant limitations of our review. Another limitation is that the included studies mainly involved patients with mixed cancer types, followed by those with breast cancer. Moreover, most of the studies were conducted in North America and Europe. Thus, the findings of this study might not be applicable to populations from other geographical areas. We were unable to conduct a meta-analysis or systematic review due to the numerous variations in the study design, clinical outcomes, and adherence definitions. As a result, we conducted a scoping review and presented a tabulated analysis of our studies.

5. Conclusions

This study reports that the definitions of the outcome measures and adherence varied across the studies. There was a limited consensus among the studies for the measured variables during treatment. Adherence to wearable devices was affected by the changes in the intervention or study design. A better understanding of the interventional period of wearable devices in terms of clinical outcomes is urgently needed. Studies using WDs and subjective questionnaires encouraged patient engagement for better cancer care management. Adherence to WDs varied from 60% to 100% depending on the intervention period. The highest adherence was reported in the 12-week studies. Most studies focused on physical activity, sleep analysis, and heart vital signs. Of the 10 studies that described patient-reported outcomes using questionnaires and personal interviews, 8 indicated a positive correlation between patient-reported and wearable outcomes. Furthermore, for a better understanding of adherence behavior, we need large intervention studies. This can provide us with a clear picture of the clinical outcomes of using wearable devices.

Author Contributions

S.S.-A., L.-J.K. and Y.H. designed the study conception and design. Y.H., U.U. and E.D. carried out the database collection and primary analysis of the database. Y.H., U.U. and E.D. wrote the first manuscript draft of the article. S.S.-A. and L.-J.K. completed the proofreading and editing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by Taipei Medical University and Taipei Medical University Hospital under a joint research grant (109TMU-TMUH-11). This Study is also partially funded by H2020, iHelp project under the research grant (GA: 101017441 and MOST, Taiwan: 110-2923-E038-001-MY3).

Conflicts of Interest

All the authors declared that there are no conflicts of interest.

References

  1. Chung, A.E.; Jensen, R.E.; Basch, E.M. Leveraging Emerging Technologies and the “Internet of Things” to Improve the Quality of Cancer Care. J. Oncol. Pract. 2016, 12, 863–866. [Google Scholar] [CrossRef] [PubMed]
  2. Ravizza, A.; De Maria, C.; Di Pietro, L.; Sternini, F.; Audenino, A.L.; Bignardi, C. Comprehensive Review on Current and Future Regulatory Requirements on Wearable Sensors in Preclinical and Clinical Testing. Front. Bioeng. Biotechnol. 2019, 7, 313. [Google Scholar] [CrossRef] [PubMed]
  3. Loucks, J. Wearable Technology in Health Care: Getting Better All the Time. TMT Predictions 2022; Deloitte: New York, NY, USA, 2021. [Google Scholar]
  4. Sabry, F.; Eltaras, T.; Labda, W.; Alzoubi, K.; Malluhi, Q. Machine Learning for Healthcare Wearable Devices: The Big Picture. J. Healthc. Eng. 2022, 2022, 4653923. [Google Scholar] [CrossRef] [PubMed]
  5. Wu, M. Wearable Technology Applications in Healthcare: A Literature Review. Online J. Nurs. Inform. 2019, 26, 23. [Google Scholar]
  6. Dunn, J.; Runge, R.; Snyder, M. Wearables and the medical revolution. Per. Med. 2018, 15, 429–448. [Google Scholar] [CrossRef]
  7. Lazazzera, R.; Belhaj, Y.; Carrault, G. A New Wearable Device for Blood Pressure Estimation Using Photoplethysmogram. Sensors 2019, 19, 2557. [Google Scholar] [CrossRef]
  8. Iqbal, S.M.A.; Mahgoub, I.; Du, E.; Leavitt, M.A.; Asghar, W. Advances in healthcare wearable devices. NPJ Flex. Electron. 2021, 5, 9. [Google Scholar] [CrossRef]
  9. Aceto, G.; Persico, V.; Pescapé, A. Industry 4.0 and Health: Internet of Things, Big Data, and Cloud Computing for Healthcare 4.0. J. Ind. Inf. Integr. 2020, 18, 100129. [Google Scholar] [CrossRef]
  10. Dias, D.; Paulo Silva Cunha, J. Wearable Health Devices-Vital Sign Monitoring, Systems and Technologies. Sensors 2018, 18, 2414. [Google Scholar] [CrossRef]
  11. Park, J.; Look, K.A. Relationship Between Objective Financial Burden and the Health-Related Quality of Life and Mental Health of Patients With Cancer. J. Oncol. Pract. 2018, 14, e113–e121. [Google Scholar] [CrossRef]
  12. Chowthi-Williams, A.; Davis, G. Successful Change Management in Health Care: Being Emotionally and Cognitively Ready; Routledge: London, UK, 2022. [Google Scholar]
  13. Farahani, B.; Firouzi, F.; Chang, V.; Badaroglu, M.; Constant, N.; Mankodiya, K. Towards fog-driven IoT eHealth: Promises and challenges of IoT in medicine and healthcare. Future Gener. Comput. Syst. 2018, 78, 659–676. [Google Scholar] [CrossRef] [Green Version]
  14. Wang, H.; Naghavi, M.; Allen, C.; Barber, R.M.; Bhutta, Z.A.; Carter, A.; Casey, D.C.; Charlson, F.J.; Chen, A.Z.; Coates, M.M.; et al. Global, regional, and national life expectancy, all-cause mortality, and cause-specific mortality for 249 causes of death, 1980-2015: A systematic analysis for the Global Burden of Disease Study 2015. Lancet 2016, 388, 1459–1544. [Google Scholar] [CrossRef]
  15. Reilly, C.M.; Bruner, D.W.; Mitchell, S.A.; Minasian, L.M.; Basch, E.; Dueck, A.C.; Cella, D.; Reeve, B.B. A literature synthesis of symptom prevalence and severity in persons receiving active cancer treatment. Support Care Cancer 2013, 21, 1525–1550. [Google Scholar] [CrossRef]
  16. A Low, C.; Dey, A.K.; Ferreira, D.; Kamarck, T.; Sun, W.; Bae, S.; Doryab, A. Estimation of Symptom Severity During Chemotherapy From Passively Sensed Data: Exploratory Study. J. Med. Internet Res. 2017, 19, e420. [Google Scholar] [CrossRef] [PubMed]
  17. Coughlin, S.S.; Caplan, L.S.; Stone, R. Use of consumer wearable devices to promote physical activity among breast, prostate, and colorectal cancer survivors: A review of health intervention studies. J. Cancer Surviv. 2020, 14, 386–392. [Google Scholar] [CrossRef]
  18. Herrington, W.G.; Goldsack, J.C.; Landray, M.J. Increasing the use of mobile technology-derived endpoints in clinical trials. Clin. Trials 2018, 15, 313–315. [Google Scholar] [CrossRef]
  19. Byrom, B.; Watson, C.; Doll, H.; Coons, S.J.; Eremenco, S.; Ballinger, R.; McCarthy, M.; Crescioni, M.; O’Donohoe, P.; Howry, C.; et al. Selection of and Evidentiary Considerations for Wearable Devices and Their Measurements for Use in Regulatory Decision Making: Recommendations from the ePRO Consortium. Value Health 2018, 21, 631–639. [Google Scholar] [CrossRef]
  20. Beauchamp, U.L.; Pappot, H.; Holländer-Mieritz, C. The Use of Wearables in Clinical Trials During Cancer Treatment: Systematic Review. JMIR mHealth and uHealth 2020, 8, e22006. [Google Scholar] [CrossRef]
  21. Ballinger, T.J.; Althouse, S.K.; Olsen, T.P.; Miller, K.D.; Sledge, J.S. A Personalized, Dynamic Physical Activity Intervention Is Feasible and Improves Energetic Capacity, Energy Expenditure, and Quality of Life in Breast Cancer Survivors. Front. Oncol. 2021, 11, 626180. [Google Scholar] [CrossRef]
  22. Nguyen, N.H.; Vallance, J.K.; Buman, M.P.; Moore, M.M.; Reeves, M.M.; Rosenberg, D.E.; Boyle, T.; Milton, S.; Friedenreich, C.M.; English, D.R.; et al. Effects of a wearable technology-based physical activity intervention on sleep quality in breast cancer survivors: The ACTIVATE Trial. J. Cancer Surviv. 2021, 15, 273–280. [Google Scholar] [CrossRef]
  23. Ferrante, J.M.; Lulla, A.; Williamson, J.D.; Devine, K.A.; Ohman-Strickland, P.; Bandera, E.V. 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. [Google Scholar] [CrossRef] [PubMed]
  24. Lynch, B.M.; Nguyen, N.H.; Moore, M.M.; Reeves, M.M.; Rosenberg, D.E.; Boyle, T.; Vallance, J.K.; Milton, S.; Friedenreich, C.M.; English, D.R. 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. [Google Scholar] [CrossRef] [PubMed]
  25. Pope, Z.C.; Zeng, N.; Zhang, R.; Lee, H.Y.; Gao, Z. Effectiveness of Combined Smartwatch and Social Media Intervention on Breast Cancer Survivor Health Outcomes: A 10-Week Pilot Randomized Trial. J. Clin. Med. 2018, 7, 140. [Google Scholar] [CrossRef] [PubMed]
  26. Mcneil, J.; Brenner, D.R.; Stone, C.R.; O’Reilly, R.; Ruan, Y.; Vallance, J.K.; Courneya, K.S.; Thorpe, K.; Klein, D.J.; Friedenreich, C.M. Activity Tracker to Prescribe Various Exercise Intensities in Breast Cancer Survivors. Med. Sci. Sports Exerc. 2019, 51, 930–940. [Google Scholar] [CrossRef]
  27. Wu, H.S.; Gal, R.; Van Sleeuwen, N.C.; Brombacher, A.C.; Ijsselsteijn, W.A.; May, A.M.; Monninkhof, E.M.; Jerome, G.; Arnoldussen, B.; Lynch, B. Breast Cancer Survivors’ Experiences With an Activity Tracker Integrated Into a Supervised Exercise Program: Qualitative Study. JMIR mHealth and uHealth 2019, 7, e10820. [Google Scholar] [CrossRef] [PubMed]
  28. Che Bakri, N.A.; Kwasnicki, R.M.; Dhillon, K.; Khan, N.; Ghandour, O.; Cairns, A.; Darzi, A.; Leff, D.R. Objective Assessment of Postoperative Morbidity After Breast Cancer Treatments with Wearable Activity Monitors: The “BRACELET” Study. Ann. Surg. Oncol. 2021, 28, 5597–5609. [Google Scholar] [CrossRef]
  29. Gandhi, A.; Samuel, S.R.; Kumar, K.V.; Saxena, P.P.; Mithra, P. Effect of a Pedometer-based Exercise Program on Cancer Related Fatigue and Quality of Life amongst Patients with Breast Cancer Receiving Chemotherapy. Asian Pac. J. Cancer Prev. 2020, 21, 1813–1818. [Google Scholar] [CrossRef]
  30. Nelson, S.H.; Weiner, L.S.; Natarajan, L.; Parker, B.A.; Patterson, R.E.; Hartman, S.J. Continuous, objective measurement of physical activity during chemotherapy for breast cancer: The Activity in Treatment pilot study. Transl. Behav. Med. 2020, 10, 1031–1038. [Google Scholar] [CrossRef]
  31. Champ, C.E.; Ohri, N.; Klement, R.J.; Cantor, M.; Beriwal, S.; Glaser, S.M.; Smith, R.P. Assessing Changes in the Activity Levels of Breast Cancer Patients During Radiation Therapy. Clin. Breast Cancer 2018, 18, e1–e6. [Google Scholar] [CrossRef]
  32. Helbrich, H.; Braun, M.; Hanusch, C.; Mueller, G.; Falk, H.; Flondor, R.; Harbeck, N.; Hermelink, K.; Wuerstlein, R.; Keim, S.; 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. [Google Scholar] [CrossRef]
  33. Li, L.; Wang, L.; Sun, Q.; Xiao, P.; Duan, Y.; Liu, X.; Zhou, J.; Xie, J.; Cheng, A.S. Effect of Two Interventions on Sleep Quality for Adolescent and Young Adult Cancer Survivors: A Pilot Randomized Controlled Trial. Cancer Nurs. 2022, 45, E560–E572. [Google Scholar] [CrossRef] [PubMed]
  34. Hasnain, Z.; Nilanon, T.; Li, M.; Mejia, A.; Kolatkar, A.; Nocera, L.; Shahabi, C.; Philips, F.A.C.; Lee, J.S.; Hanlon, S.E.; et al. Quantified Kinematics to Evaluate Patient Chemotherapy Risks in Clinic. JCO Clin. Cancer Inform. 2020, 4, 583–601. [Google Scholar] [CrossRef] [PubMed]
  35. Nilanon, T.; Nocera, L.P.; Martin, A.S.; Kolatkar, A.; May, M.; Hasnain, Z.; Ueno, N.T.; Yennu, S.; Alexander, A.; Mejia, A.E.; et al. Use of Wearable Activity Tracker in Patients With Cancer Undergoing Chemotherapy: Toward Evaluating Risk of Unplanned Health Care Encounters. JCO Clin. Cancer Inform. 2020, 4, 839–853. [Google Scholar] [CrossRef] [PubMed]
  36. Yurkiewicz, I.R.; Simon, P.; Liedtke, M.; Dahl, G.; Dunn, T. Effect of Fitbit and iPad Wearable Technology in Health-Related Quality of Life in Adolescent and Young Adult Cancer Patients. J. Adolesc. Young Adult Oncol. 2018, 7, 579–583. [Google Scholar] [CrossRef] [PubMed]
  37. Miyaji, T.; Kawaguchi, T.; Azuma, K.; Suzuki, S.; Sano, Y.; Akatsu, M.; Torii, A.; Kamimura, T.; Ozawa, Y.; Tsuchida, A.; et al. Patient-generated health data collection using a wearable activity tracker in cancer patients-a feasibility study. Support. Care Cancer 2020, 28, 5953–5961. [Google Scholar] [CrossRef]
  38. Komarzynski, S.; Huang, Q.; Lévi, F.A.; Palesh, O.G.; Ulusakarya, A.; Bouchahda, M.; Haydar, M.; Wreglesworth, N.I.; Morère, J.-F.; Adam, R.; et al. The day after: Correlates of patient-reported outcomes with actigraphy-assessed sleep in cancer patients at home (inCASA project). Sleep 2019, 42, zsz146. [Google Scholar] [CrossRef]
  39. Hardcastle, S.J.; Maxwell-Smith, C.; Hince, D.; Bulsara, M.K.; Boyle, T.; Tan, P.; Levitt, M.; Salama, P.; Mohan, G.R.K.A.; Salfinger, S.; et al. The wearable activity technology and action-planning trial in cancer survivors: Physical activity maintenance post-intervention. J. Sci. Med. Sport 2021, 24, 902–907. [Google Scholar] [CrossRef]
  40. Devine, K.A.; Viola, A.; Levonyan-Radloff, K.; Mackowski, N.; Bozzini, B.; Chandler, A.; Xu, B.; Ohman-Strickland, P.; Mayans, S.; Farrar-Anton, A.; et al. Feasibility of FitSurvivor: A technology-enhanced group-based fitness intervention for adolescent and young adult survivors of childhood cancer. Pediatr. Blood Cancer 2020, 67, e28530. [Google Scholar] [CrossRef]
  41. Pavic, M.; Klaas, V.; Theile, G.; Kraft, J.; Tröster, G.; Guckenberger, M. Feasibility and Usability Aspects of Continuous Remote Monitoring of Health Status in Palliative Cancer Patients Using Wearables. Oncology 2020, 98, 386–395. [Google Scholar] [CrossRef]
  42. Mendoza, J.A.; Baker, K.S.; Moreno, M.A.; Whitlock, K.; Abbey-Lambertz, M.; Waite, A.; Colburn, T.; Chow, E.J. A Fitbit and Facebook mHealth intervention for promoting physical activity among adolescent and young adult childhood cancer survivors: A pilot study. Pediatr. Blood Cancer 2017, 64, e26660. [Google Scholar] [CrossRef]
  43. Cadmus-Bertram, L.; Tevaarwerk, A.J.; Sesto, M.E.; Gangnon, R.; Van Remortel, B.; Date, P. Building a physical activity intervention into clinical care for breast and colorectal cancer survivors in Wisconsin: A randomized controlled pilot trial. J. Cancer Surviv. 2019, 13, 593–602. [Google Scholar] [CrossRef] [PubMed]
  44. Hardcastle, S.J.; Jiménez-Castuera, R.; Maxwell-Smith, C.; Bulsara, M.K.; Hince, D. 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. [Google Scholar] [CrossRef] [PubMed]
  45. Maxwell-Smith, C.; Hince, D.; Cohen, P.; Bulsara, M.K.; Boyle, T.; Platell, C.; Tan, P.; Levitt, M.; Salama, P.; Tan, J.; et al. A randomized controlled trial of WATAAP to promote physical activity in colorectal and endometrial cancer survivors. Psychooncology 2019, 28, 1420–1429. [Google Scholar] [CrossRef] [PubMed]
  46. Van Blarigan, E.L.; Chan, H.; Van Loon, K.; Kenfield, S.A.; Chan, J.M.; Mitchell, E.; Zhang, L.; Paciorek, A.; Joseph, G.; Laffan, A.; 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. [Google Scholar] [CrossRef]
  47. Cheong, I.Y.; An, S.Y.; Cha, W.C.; Rha, M.Y.; Kim, S.T.; Chang, D.K.; Hwang, J.H. Efficacy of Mobile Health Care Application and Wearable Device in Improvement of Physical Performance in Colorectal Cancer Patients Undergoing Chemotherapy. Clin. Colorectal Cancer 2018, 17, e353–e362. [Google Scholar] [CrossRef]
  48. Van Blarigan, E.L.; Dhruva, A.; Atreya, C.E.; Kenfield, S.A.; Chan, J.M.; Milloy, A.; Kim, I.; Steiding, P.; Laffan, A.; Zhang, L.; et al. Feasibility and Acceptability of a Physical Activity Tracker and Text Messages to Promote Physical Activity During Chemotherapy for Colorectal Cancer: Pilot Randomized Controlled Trial (Smart Pace II). JMIR Cancer 2022, 8, e31576. [Google Scholar] [CrossRef]
  49. Shih, C.A.-O.; Chou, P.A.-O.; Chou, T.A.-O.; Huang, T.A.-O. Measurement of Cancer-Related Fatigue Based on Heart Rate Variability: Observational Study. J. Med. Internet Res. 2021, 23, e25791. [Google Scholar] [CrossRef]
  50. Bade, B.C.; Brooks, M.C.; Nietert, S.B.; Ulmer, A.; Thomas, D.D.; Nietert, P.J.; Scott, J.B.; Silvestri, G.A. Assessing the Correlation Between Physical Activity and Quality of Life in Advanced Lung Cancer. Integr. Cancer Ther. 2018, 17, 73–79. [Google Scholar] [CrossRef]
  51. Finley, D.J.; Stevens, C.J.; Emond, J.A.; Batsis, J.A.; Fay, K.A.; Darabos, C.; Sacks, O.A.; Cook, S.B.; Lyons, K.D. Potential effectiveness of a surgeon-delivered exercise prescription and an activity tracker on pre-operative exercise adherence and aerobic capacity of lung cancer patients. Surg. Oncol. 2021, 37, 101525. [Google Scholar] [CrossRef]
  52. Kong, S.; Park, H.Y.; Kang, D.; Lee, J.K.; Lee, G.; Kwon, O.J.; Shim, Y.M.; Zo, J.I.; Cho, J. Seasonal Variation in Physical Activity among Preoperative Patients with Lung Cancer Determined Using a Wearable Device. J. Clin. Med. 2020, 9, 349. [Google Scholar] [CrossRef]
  53. Ghods, A.; Shahrokni, A.; Ghasemzadeh, H.; Cook, D. 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. [Google Scholar] [CrossRef] [PubMed]
  54. Wu, J.-M.; Ho, T.-W.; Chang, Y.-T.; Hsu, C.; Tsai, C.J.; Lai, F.; Lin, M.-T. Wearable-Based Mobile Health App in Gastric Cancer Patients for Postoperative Physical Activity Monitoring: Focus Group Study. JMIR mHealth and uHealth 2019, 7, e11989. [Google Scholar] [CrossRef] [PubMed]
  55. Kim, Y.; Seo, J.; An, S.Y.; Sinn, D.H.; Hwang, J.H. Efficacy and Safety of an mHealth App and Wearable Device in Physical Performance for Patients With Hepatocellular Carcinoma: Development and Usability Study. JMIR mHealth and uHealth 2020, 8, e14435. [Google Scholar] [CrossRef] [PubMed]
  56. Yang, C.-F.J.; Aibel, K.; Meyerhoff, R.; Wang, F.; Harpole, D.; Abernethy, A.P.; Leblanc, T.W. Actigraphy assessment of sleep quality among patients with acute myeloid leukaemia during induction chemotherapy. BMJ Supportive Palliat. Care 2018, 8, 274–277. [Google Scholar] [CrossRef]
  57. Sugiyama, Y.; Naiki, T.; Tasaki, Y.; Kataoka, T.; Mimura, Y.; Kondo, Y.; Etani, T.; Iida, K.; Nozaki, S.; Ando, R.; et al. Effectiveness of continuous monitoring by activity tracker of patients undergoing chemotherapy for urothelial carcinoma. Cancer Treat. Res. Commun. 2020, 25, 100245. [Google Scholar] [CrossRef] [PubMed]
  58. Robinson, J.K.; Durst, D.A.; Gray, E.; Kwasny, M.; Heo, S.Y.; Banks, A.; Rogers, J.A. Sun exposure reduction by melanoma survivors with wearable sensor providing real-time UV exposure and daily text messages with structured goal setting. Arch. Dermatol. Res. 2021, 313, 685–694. [Google Scholar] [CrossRef] [PubMed]
  59. Lancaster, K.; Abuzour, A.; Khaira, M.; Mathers, A.; Chan, A.; Bui, V.; Lok, A.; Thabane, L.; Dolovich, L. The Use and Effects of Electronic Health Tools for Patient Self-Monitoring and Reporting of Outcomes Following Medication Use: Systematic Review. J. Med. Internet Res. 2018, 20, e294. [Google Scholar] [CrossRef]
  60. Bakker, J.P.; Goldsack, J.C.; Clarke, M.; Coravos, A.; Geoghegan, C.; Godfrey, A.; Heasley, M.G.; Karlin, D.; Manta, C.; Peterson, B.; et al. A systematic review of feasibility studies promoting the use of mobile technologies in clinical research. NPJ Digit. Med. 2019, 2, 47. [Google Scholar] [CrossRef]
  61. Gresham, G.; Schrack, J.; Gresham, L.M.; Shinde, A.M.; Hendifar, A.E.; Tuli, R.; Rimel, B.; Figlin, R.; Meinert, C.L.; Piantadosi, S. Wearable activity monitors in oncology trials: Current use of an emerging technology. Contemp. Clin. Trials 2018, 64, 13–21. [Google Scholar] [CrossRef]
  62. Kadhim, K.T.; Alsahlany, A.M.; Wadi, S.M.; Kadhum, H.T. An Overview of Patient’s Health Status Monitoring System Based on Internet of Things (IoT). Wirel. Pers. Commun. 2020, 114, 2235–2262. [Google Scholar] [CrossRef]
Figure 1. PRISMA flow diagram of the screening and selection of studies.
Figure 1. PRISMA flow diagram of the screening and selection of studies.
Cancers 14 04437 g001
Figure 2. Graphical representation of adherence to wearable devices by duration of intervention.
Figure 2. Graphical representation of adherence to wearable devices by duration of intervention.
Cancers 14 04437 g002
Table 1. Summary of Included studies.
Table 1. Summary of Included studies.
Country of StudyTopic (Type of Cancer and Status)Study Design Tools Used Participant (%)
Gender
The United States of America [21]Breast Cancer (n = 57)Survivors Feasibility Study Wearable Device and Questionnaire100% Women
Australia [22]Breast Cancer (n = 80)Survivors RCT Wearable Device + Text Messages and Personal Interviews + Mobile Application 100% Women
The United States of America [23]Breast Cancer (n = 34)Survivors RCT Wearable Device and Questionnaire (Correlation)100% Women
Australia [24]Breast Cancer (n = 80)Survivors RCT Wearable Device and Questionnaire (Correlation)100% Women
The United States of America [25]Breast Cancer (n = 20)Survivors RCT Wearable Device + Group Sessions and Phone Calls 100% Women
Canada [26]Breast Cancer (n = 41)Survivors RCT Wearable Device and Questionnaires (Correlation)100% Women
The Netherlands [27]Breast Cancer (n = 8)Survivors Qualitative StudyWearable Device and Questionnaires100% Women
United Kingdom [28]Breast Cancer (n = 39)Under Treatment Non-RCTWearable Device, Questionnaire, and Behavioral Counseling Session 100% Women
India [29]Breast Cancer (n = 44)Under Treatment Non-RCTWearable Device + General group session + Questionnaire + Mobile Application95.4% Women
The United States of America [30]Breast Cancer (n = 32)Under Treatment Pilot StudyWearable Device + Mobile application + Text Messages 100% Women
The United States of America [31]Breast Cancer (n = 10)Under Treatment RCT Wearable Device and Questionnaire100% Women
Germany [32] Breast Cancer (n = 99)Under Treatment Feasibility Study Wearable Device and Questionnaire100% Women
Central China [33]Mixed Cancer (n = 112)Under Treatment RCT Wearable Device76.2% Women
The United States of America [34]Mixed Cancer (n = 38)Under Treatment Utility Study/Predictive Study Wearable Device + Mobile application and Interview 52% Women
The United States of America [35]Mixed Cancer (n = 41)Under Treatment Observational Study Wearable Device + Mobile application + Questionnaire 56% Women
The United States of America [36]Mixed Cancer (n = 33)Under Treatment Prospective cohort Study Wearable Devices and Spirometer57.5% Women
Japan [37]Mixed Cancer (n = 30)Under Treatment Feasibility Study Wearable Device70% Men
France [38]Mixed Cancer (n = 31)Under Treatment Pilot StudyWearable Device + Mobile Application + Questionnaire55% Men
Ireland [39] Mixed Cancer (n = 61)Survivors RCT Wearable Devices + Goal-setting session + Telephone-delivered health-coaching sessions50% Men
The United States of America [40]Mixed Cancer (n = 32)Survivors Feasibility Study Wearable Device + Two group sessions + support phone call51% Men
Switzerland [41]Mixed Cancer (n = 30)Survivors Feasibility Study Fitbit + iPad (preloaded apps) + Questionnaires70% Men
The United States of America [42]Mixed Cancer (n = 59)Survivors Pilot StudyWearable Device and Questionnaire59.3% Women
The United States of America [43] Mixed Cancer (n = 47)Survivors RCT Wearable Device + Questionnaire + Social Media Intervention (Health Education)96% Women
Australia [44] Colorectal and Endometrial cancer (n = 29)Survivors RCT Mobile application (in-app chat service) + Wearable device + Questionnaires58% Women
Western Australia [45] Colorectal Cancer (n = 61)Survivors RCT Wearable Device + mHealth app + Peer-based virtual support group + Qualitative Interviews 50% Women
The United States of America [46]Colorectal Cancer (n = 39)Survivors RCT Wearable Device and Questionnaire-based study58% Women
South Korea [47] Colorectal Cancer (n = 75)Under Treatment Feasibility Study Wearable device + Questionnaires + e-Patient Diary 58.7% Men
The United States of America [48] Colorectal Cancer (n = 40)Under Treatment Pilot StudyWearable Device and Questionnaire-based study56.8% Women
Taiwan [49] Lung Cancer (n = 12)Under Treatment Observational Study Wearable Device and Questionnaire-based study58.33% Men
The United States of America [50] Lung Cancer (n = 30)Under Treatment Observational Study Wearable Device + Questionnaire + Educational handbook + Social support + Email-based coaching67% Men
The United States of America [51]Lung Cancer (n = 18)Under Treatment Observational Study Wearable Device and Questionnaire (Correlation)44% Women
South Korea [52] Lung Cancer (n = 555)Under Treatment Usability StudyWearable Devices + Questionnaire+ Educational handbook+ Social support + Email-based coaching61% Men
The United States of America [53] Gastric cancer (n = 27)Under Treatment Cohort Study Wearable Device + Mobile Application62.96% Men
Taiwan [54] Gastric Cancer (n = 43)Under Treatment Group Study Wearable Devices + Questionnaires51% Men
South Korea [55] Liver Cancer (n = 31)Under Treatment Usability StudyWearable Device + Daily text messages+ Questionnaire 84% Men
The United States of America [56] Blood Cancer (n = 11)Under Treatment Feasibility Study Diary + Accelerometer66.6% Men
Japan [57] Urothelial Carcinoma (n = 21)Under Treatment Cohort Study Wearable Device84% Men
The United States of America [58]Skin Cancer (n = 60)Survivor Observational Study Wearable Devices + Questionnaire + Interviews 60% Women
Table 2. Adherence to wearable devices in the cancer population.
Table 2. Adherence to wearable devices in the cancer population.
Country of StudyTotal Study Duration (in Weeks) Intervention
Duration (in Weeks)
Patients
Recruited
Criteria for EvaluationAdherence
(in
Percentage)
The United States of America [21]241260Percentage of enrolled patients who completed all assessments (10 h per day for 4 days in a week)95
Australia [22]241283Based on the given assessment completion94
The United States of America [23]522444Collection of data (days with less than 1000 steps considered as non-adherent)65
Australia [24]121283Completeness of data collection96
The United States of America [25]101030Completeness of data collection67
Canada [26]241245Completeness of data collection 88
The Netherlands [27]121210Based on data collection and
total wearing days
80
United Kingdom [28]2256Collected data on the different days (39 patients*14 days)89
India [29]7744Users’ tolerance ability to the intensity of the program that was set using the rate of perceived exertion (RPE) 93
The United States of America [30]171732Days were considered “valid” if there was any wear time
recorded (5 min threshold)
100
The United States of America [31]101010Completeness of data collection 100
Germany [32] 2424112Completeness of data collection95
Central China [33]88143Completeness of data collection78
The United States of America [34]8845Completeness of data collection84
The United States of America [35]20834Completeness of data collection68
The United States of America [36]434344Completeness of data collection75
Japan [37]4430Completeness of data collection 90
France [38]4430Completeness of data collection 86
Ireland [39] 241268Completeness of data collection 89
The United States of America [40]521249Completeness of data collection65
Switzerland [41]121230Completeness of data collection and qualitative analysis of
interviews
83
The United States of America [42]101059Completeness of data collection 100
The United States of America [43] 121250Completeness of data collection 94
Australia [44] 241234Based on participants who completed the study criterion, which is a minimum of 1000 steps or more denoted per day. 82
Western Australia [45] 121268Completeness of data collection94
The United States of America [46]121241Completeness of data
collection
81
South Korea [47] 1212102Completeness of data collection74
The United States of America [48] 121244Completeness of data collection 88
Taiwan [49] 1112Completeness of data collection 100
The United States of America [50] 1139Completeness of data collection 67
The United States of America [51]3330Completeness of data collection60
South Korea [52] 5252555Completeness of data collection100
The United States of America [53] 3341Based on the rate of data collected during chemotherapy63
Taiwan [54] 4443Completeness of data collection 100
South Korea [55] 121237Equivalent to the completion of the exercise program 84
The United States of America [56] 2212Completeness of data collection92
Japan [57] 121228Completeness of data collection 75
The United States of America [58]3360In-person interviews to examine the acceptability of the device and analysis of qualitative data100
Table 3. Reported clinical outcomes in the cancer population using wearable devices.
Table 3. Reported clinical outcomes in the cancer population using wearable devices.
Country of StudyCancer Type Purpose Reported Clinical Outcomes
The United States of America [21]Breast Cancer Behavioral health
management (PA/QoL and
fatigue)
High engagement among
hospitalized patients and
increased energy
expenditure among cancer
survivors. Outcomes depend on
numerous factors related to users and their needs.
Australia [22]Breast Cancer Behavioral health
management (sleep quality)
Changes in actigraphy (sleep
efficiency) and PSQI global and subscales favored the intervention arm. Findings were not significant or clinically meaningful.
The United States of America [23]Breast Cancer Behavioral health
management (physical activity/BMI/QoL/fatigue/
fitness/self-regulation and self-efficacy related to PA)
Self-monitoring, goal setting, and self-efficacy were significantly
correlated with activity levels. Increased improvement in health was noted with an increase in PA.
Australia [24]Breast Cancer Behavioral health management (MVPA/Sedentary
Behavior)
The intervention resulted in increases in MVPA and MVPA accrued in bouts of at least 10 consecutive min while reducing total and prolonged sitting times. A significant difference in MVPA was noted between groups at T2, favoring the
intervention arm.
The United States of America [25]Breast Cancer Behavioral health management (PA- MVPA,
Sedentary/physiological/
psychosocial/QoL variables)
No significant group differences were observed for changes over time for any variable. Both groups showed increased mean daily MVPA, light PA, energy
expenditure, and steps/day.
Canada [26]Breast Cancer Behavioral health
management (PA-MVPA, LIPA, Sedentary
Behavior/Sleep quality/health-related Fitness Markers)
Increases in moderate-to-vigorous intensity PA and decreases in sedentary time were significantly greater in the lower-intensity PA group versus the control group at 12 weeks. Increases in V˙O2 max at 12 weeks in both intervention groups were significantly greater than the changes in the control group. Changes in PA and V˙O2 max
remained at 24 weeks but
differences between the
intervention and control groups were not significant.
The Netherlands [27]Breast Cancer Behavioral health
management (PA-Sedentary behavior)
The activity tracker motivated women to be physically active and increased their awareness of their sedentary lifestyle. Wearing an
activity tracker raised lifestyle awareness in patients with breast cancer.
United Kingdom [28]Breast Cancer Behavioral health
management (Upper Limb Function)
WAM improved on the surgical side of the upper limb with an
increment in PA for the first week and showed a good correlation with DASH (0.0506)
India [29]Breast Cancer Behavioral health
management
(Fatigue/QoL//Functional
Capacity/PA/Body Composition)
At the end of the 7-week
intervention, functional capacity, quality of life, and skeletal mass were significantly improved, whereas fatigue and changes in
total fat improved nonsignificantly.
The United States of America [30]Breast Cancer Behavioral health management (PA/MVPA/SB/Cognitive functions)Participants decreased their activity from pre- to post-chemotherapy by 1 h/week in MVPA and 8 h/week in TPA during the decline. This is
useful for determining the stage of chemotherapy in which PA starts to decline and patients need extra
support for their care.
The United States of America [31]Breast Cancer Behavioral health
management (PA/Sleep
Metrics)
Overall step count decreased by an average of 54 steps per day from baseline during treatment. Although differences in step count,
calories expended, and miles walked throughout the RT were minimal, they were significant because of the substantial number of events
Germany [32] Breast Cancer Behavioral health
management (PA)
Coherence between self-reported and device data was strong (r = 0.566). Neither treatment nor week nor their interaction had effects on step counts. Self-reported activity time was lower for patients receiving chemotherapy than for those not
receiving chemotherapy and lower in the 18th week than in the 3rd week
Central China [33]Mixed CancerBehavioral health
management (Asleep + QoL)
The baseline measurement was not significantly different among the three groups. However, after the
intervention, a significant difference between the experimental and
control groups was noted. Sleep quality and PA improved
significantly but not the secondary
outcomes.
The United States of America [34]Mixed CancerBehavioral health
management (Unplanned Healthcare Encounter/PA)
Kinematic features associated with physical activity showed a positive correlation. Chair-to-table
kinematics are good predictors of unexpected hospitalization. Get-
up-and-walk kinematics are good predictors of low physical activity
The United States of America [35]Mixed CancerBehavioral health
management (Unplanned Healthcare Encounter/PA)
This study demonstrated the
feasibility of an outpatient wearable activity tracker. The results revealed a 50% disagreement with no
association of these disagreements with UHEs and no correlation
between the UHEs and ECOG scores. A correlation between (1)
average METs and UHEs and (2) no sedentary physical activity hours and UHEs was noted
The United States of America [36]Mixed CancerBehavioral health
management (PA/QoL)
Significant improvements across all eight dimensions of HRQOL; most patients (85%) reported that they enjoyed wearing the Fitbit. Most felt that the Fitbit helped them to be more active (79%), whereas a minority (18%) felt their activity level was the same, and none
reported becoming less active.
Japan [37]Mixed Cancer Behavioral health
management (PA/Symptom Burden Assessment/Sleep/Fatigue)
Use of a wearable activity tracker for collecting PGHD in real time
according to the protocol was
feasible. With respect to adherence, the result was significant. The
correlation between the assessed data was not significant
France [38]Mixed Cancer Behavioral health management (PA/Sleep)Results provide evidence for both the feasibility and relevance of the combined objective and subjective remote monitoring of sleep and other symptoms in patients with cancer with single-night precision. This dynamic approach can help the development of novel therapeutics whose testing is warranted in
patients with cancer
Ireland [39] Mixed CancerBehavioral health management (MVPA/Cardiovascular risk factors and sedentary behavior)The estimated difference between groups at 24 weeks supported higher MVPA; no change in MVPA in the intervention group was
observed during the 12-week
follow-up period, indicating a
positive correlation with
the improvement in cardiovascular risk factors.
The United States of America [40]Mixed CancerBehavioral health
Management (MVPA/QoL/
Fatigue/Fitness/Sedentary
Behavior)
Results of the studies revealed some promising improvements in
muscular strength that aligned with the intervention’s focus on strength training.
Switzerland [41]Mixed CancerBehavioral health
management (Symptom Analysis)
Remote monitoring of healthcare status in patients receiving
palliative care with a limited life
expectancy is feasible, and patients can handle the smartphone and
sensor-equipped bracelet. Feedback toward the use of this monitoring system was mostly positive.
The United States of America [42]Mixed Cancer Behavioral health management (PA-SB and MVPA/QoL)Intervention participants had a lower-than-expected engagement in the Facebook group component, (passive instead of active engagement); MVPA and sedentary time showed no significant difference b/w gaps
The United States of America [43] Mixed Cancer Behavioral health management (PA-MVPA)Increased physical activity among cancer survivors was noted: the intervention group increased their daily steps. Moderate-to-vigorous-intensity activity performed in 10 min bouts increased, but no significant group-by-time differences for either light- or vigorous-intensity activity were noted
Australia [44] Colorectal and endometrial Cancer Behavioral health management (PA -Steensma)Fitbit wear time (percentage of valid wear days = adherence) was consistent with a median adherence score of 100%. Comparison and
correlation with actigraphy (MVPA) show that both devices are not
correlated and do not show any type of association.
Western Australia [45] Colorectal Cancer Behavioral health management (MVPA/Cardiovascular Risk)Despite a significant increase in MVPA, the change in the proportion of participants meeting the
guidelines in relation to MV10 did not significantly differ by group. Reduction in DBP among
intervention participants that were hypertensive. Fitbit was promising for low-intensity interventions.
The United States of America [46]Colorectal Cancer Behavioral health
management (PA-MVPA/
Adverse events)
Intervention arm increased its MVPA by 13 min per day more than the control arm. Larger studies should be conducted to determine whether the intervention increases physical activity.
South Korea [47] Colorectal Cancer Behavioral health
management (PA/QoL/Nutritional Status/Physical Performance)
Lower-extremity strength and
cardiorespiratory endurance were significantly improved. Fatigue and nausea/vomiting symptoms were significantly relieved after the program. Most of the functional scales showed improvements,
although the changes were not
significant.
The United States of America [48] Colorectal Cancer Behavioral health
management (PA/)
Pilot data show a nonsignificant
decrease in moderate activity
accumulated in bouts of at least 10 min in both arms (16–21 min per week).
Taiwan [49] Lung
Cancer
Behavioral health
management (CRF)
The LF to HF ratio is highly
correlated with the subjective BFI,
particularly when measured during sleep time. Analytical results revealed that this ratio can be used to
evaluate cancer fatigue because of a 3% mapping error in the BFI
The United States of America [50] Lung
Cancer
Behavioral health
management (Steps/Day and MVPA/Sedentary
Behavior/Cardiorespiratory Fitness)
Participants who received surgery in the spring, summer, autumn, and winter seasons, respectively, had lower PA and CRF than those who received surgery in other seasons. These results were consistent among all study subgroups.
The United States of America [51]Lung
Cancer
Behavioral health management (PA-Steps/QoL/
Symptoms/Functional
Status/Dyspepsia)
Improved PA was associated with the early discharge of patients with GC undergoing gastrectomy. This was because patients with improved PA had resumed physical function, which was the main factor
evaluated if patients were qualified to be discharged.
South
Korea [52]
Lung
Cancer
Behavioral health
management (MVPA/Aerobic Capacity)
Eight (47%) of the seventeen
participants demonstrated a
clinically significant improvement of 14 m or more. The average
improvement in aerobic capacity (13.8 m) was close to the minimum threshold for a clinically
meaningful improvement of 14 m
The United States of America [53] Gastric cancer Behavioral health
management (PA and
Symptom Burden)
This study’s results indicate
significant correlations between the number of the step count and two common performance statuses, which is consistent with previous research findings. Questionnaire findings
indicated that active patients have a lower burden of symptoms.
Taiwan [54] Gastric Cancer Behavioral health management (PA/Sleep Metrics)Results provide evidence for both the feasibility and relevance of
the combined objective and subjective remote monitoring of sleep and other symptoms in patients with cancer with single-night precision. This dynamic approach can guide the development of novel therapeutic concepts whose testing is warranted in patients with cancer
South Korea [55] Liver
Cancer
Behavioral health
management (Exercise
Capacity/PA/QoL/Body
Composition and
Biochemical)
Compared with baseline, significant improvements were found in
physical fitness measures, body composition, self-reported amount of physical activity, and pain. All symptoms improved, as observed in the QoL scales (i.e., EORTC-QLQ C30).
The United States of America [56] Blood Cancer Behavioral health
management (PA/Sleep)
This study demonstrates the
feasibility of collecting sleep data through actigraphy among
hospitalized adults. Actigraphy measures suggested poor sleep.
Japan [57] Urothelial Carcinoma Behavioral health
management (PA/QoL/
Adverse Events)
Significant correlations were noted between measurements performed using an oscillometer and a Fitbit during chemotherapy for patients. The measurement of fatigue using Fitbit was effective
The United States of America [58]Skin
Cancer
Preventive care No differences in baseline knowledge or attitudes regarding sun exposure or protection were noted between the two groups.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Huang, Y.; Upadhyay, U.; Dhar, E.; Kuo, L.-J.; Syed-Abdul, S. A Scoping Review to Assess Adherence to and Clinical Outcomes of Wearable Devices in the Cancer Population. Cancers 2022, 14, 4437. https://doi.org/10.3390/cancers14184437

AMA Style

Huang Y, Upadhyay U, Dhar E, Kuo L-J, Syed-Abdul S. A Scoping Review to Assess Adherence to and Clinical Outcomes of Wearable Devices in the Cancer Population. Cancers. 2022; 14(18):4437. https://doi.org/10.3390/cancers14184437

Chicago/Turabian Style

Huang, Yaoru, Umashankar Upadhyay, Eshita Dhar, Li-Jen Kuo, and Shabbir Syed-Abdul. 2022. "A Scoping Review to Assess Adherence to and Clinical Outcomes of Wearable Devices in the Cancer Population" Cancers 14, no. 18: 4437. https://doi.org/10.3390/cancers14184437

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop