Abstract
Patient-generated health data (PGHD), or health-related data gathered from patients to help address a health concern, is increasingly used to make regulatory decisions and evaluate quality of care. PGHD includes self-reported health and treatment histories, patient-reported outcomes (PROs), and biometric sensor data. Advances in wireless technology, smartphones, and the Internet of Things (IoT) have facilitated new ways to collect PGHD during clinic visits and in daily life. The goal of the current review is to provide an overview of the current clinical, regulatory, technological, and analytic landscape as it relates to PGHD in oncology research and care. The review begins with a rationale for PGHD as described by the U.S. Food and Drug Administration, Institute of Medicine, and other regulatory and scientific organizations. The evidence base for clinic-based and remote symptom monitoring using PGHD is described, with an emphasis on PROs. An overview is presented of current approaches to digital phenotyping, or device-based, real-time assessment of biometric, behavioral, self-report, and performance data. Analytic opportunities regarding PGHD are envisioned in the context of big data and artificial intelligence in medicine. Finally, challenges and solutions for integration of PGHD into clinical care are presented. Challenges include EMR integration of PROs and biometric data, analysis of large and complex biometric datasets, and potential clinic workflow redesign. In addition, there is currently more limited evidence for use of biometric data relative to PROs. Despite these challenges, the potential benefits of PGHD make it increasingly likely to be integrated into oncology research and clinical care.
The past decade has seen remarkable progress in the translation of biological discoveries into new cancer treatments, such as targeted therapies, immune checkpoint inhibitors, and adaptive cellular therapies. The effect of each new treatment is incremental but cumulatively their impact on survival rates, together with previous advances in early detection, has been unprecedented. Although diagnosis and treatment can still be a lonely and fearful experience for patients, in many cases cancer is becoming a chronic condition rather than a fatal disease. The result is that maintenance of quality of life has become an increasingly important clinical goal.
Quality of life is one aspect of patient-generated health data (PGHD), or health-related data gathered from patients to help address a health concern.1,2 PGHD includes self-reported health and treatment histories, patient-reported outcomes (PROs), and biometric data (see Figure 1). PROs are defined by the U.S. Food and Drug Administration (FDA) as “reports of the status of a patient’s health condition that come directly from the patient, without interpretation of the patient’s response by a clinician or anyone else.”3 PROs can be categorized as disease-related symptoms, side effects of treatment, and quality of life (i.e., how symptoms and side effects impact daily functioning).4 Whereas self-reported health and treatment histories are well-established in the clinical setting and PROs in the research setting, the use of biometric PGHD data is still in early development. Biometric data can include passively-collected data from wearable sensors (e.g., a physical activity tracker) as well as data actively collected by patients through other instruments (e.g., a wireless blood pressure cuff). Advances in wireless technology, smartphones, and the Internet of Things (IoT) have facilitated new ways to collect PGHD during point-of-care clinic visits and in routine daily life. The current article will review opportunities and challenges of PGHD to inform regulatory decisions and cancer care delivery.
Figure 1.
Examples of patient-generated health data as defined by the U.S. Department of Health and Human Services.1
The Need for Patient-Generated Health Data in Cancer Care
Growing interest in PGHD in oncology both reflects and reinforces an increasing role for patient advocates in directing regulatory priorities.5 In 2009 the U.S. FDA released a draft guidance document encouraging patient-focused drug development to ensure that the patient experience is sufficiently represented in benefit-risk assessment.3 Patient-focused drug development has primarily taken the form of collection of PROs as secondary outcomes in Phase III clinical trials,3 although adherence to guidelines for implementation has been sub-optimal.6 With the exception of one trial that collected home-based blood pressure readings,7 remote collection of biometric data has been limited. Nevertheless, collection of PROs is important because there are extensive data that suggest PROs provide information that is complementary to but different than clinician-rated adverse events. While clinicians’ ratings of adverse events are informed by medical knowledge, they tend to significantly underestimate patients’ reports of symptomatology. Severe symptomatology has been shown to be under-reported on clinical trials by up to 76%.8,9 In contrast, data suggest that PROs are more sensitive to treatment-related differences in toxicity than clinician-rated adverse events.10 Notably, patients treated on clinical trials tend to be younger, healthier, and have higher socioeconomic status than patients treated outside of clinical trials in the community setting.11,12 Thus, adverse events reported on clinical trials may not be generalizable to patients receiving the same treatment as standard of care. In contrast, PROs give a voice to patients.13,14 Without PRO data from high-quality studies,15 patients may instead rely on anecdotal information from the internet about what to expect for a given disease and treatment.16 This anecdotal information may be inaccurate or ungeneralizable.16 PRO data are particularly important when patients must decide between two or more treatments that demonstrate similar or modest benefits regarding survival. PRO data can inform decision making in this situation by providing insight into quality of life, such as the ability to maintain one’s roles and responsibilities during treatment (e.g., continuing to work), which can also have significant emotional and financial benefits.16
PGHD also reflects increased awareness of the importance of proactive symptom management in high-quality cancer care.13,17–19 Proactive symptom management is part of a larger trend recommended by the Institute of Medicine (IOM) to engage patients to improve quality of care. As recently as 1999, PROs were framed in terms of understanding tradeoffs between quantity and quality of life.20 However, a series of studies published starting in 2010 demonstrated that early palliative care improved survival by an average of 4.6 months in patients with advanced cancer.21–23 These and other studies showed that early palliative care also improved quality of life and reduced distress in both patients and caregivers.21,23–25 Findings were extended in a recent high-visibility study demonstrating that clinic-based symptom monitoring and management improved quality of life and extended survival by 5 months in cancer patients treated with chemotherapy, perhaps because patients received chemotherapy longer.26,27 Notably, the survival benefits of symptom management compare favorably to U.S. FDA approved anti-cancer agents approved between 2009 and 2013, which demonstrated a median survival benefit of 2.7 months.28 Thus, it has become evident that improving quality of life can also lengthen quantity of life.
Opportunities abound to improve symptom management. Symptomatology has been chronicled most comprehensively by Cancer Care Ontario through systematic administration of the Edmonton Symptom Assessment Scale (ESAS) to oncology patients since 2007.29 In a study of 120,745 patients within 12 months of diagnosis, the most commonly-endorsed moderate to severe symptoms were tiredness (59%), low overall well-being (55%), anxiety (44%), lack of appetite (43%), and pain (37%).29 A diagnosis of respiratory or oropharyngeal cancer, younger age, female sex, lower income, greater comorbidities, and urban residence were associated with significantly higher odds of elevated symptom burden.29 These findings are consistent with meta-analyses estimating the prevalence of common patient-reported symptoms. For example, the prevalence of pain is estimated to be 55% during anticancer treatment; 39% after curative treatment; 66% in advanced, metastatic, or terminal disease; and 50.7% in all cancer stages.30 The prevalence of fatigue is estimated to be 14–27% among breast cancer survivors, 78% among older patients receiving palliative care, and 7% in the general population.31–33 The prevalence of depression is estimated to be 27% during treatment, 21% during the year after diagnosis, 15% after one year following diagnosis, and 12% after two or more years following diagnosis.34,35 In contrast, depression in non-cancer controls assessed similarly is estimated to be 10%.35 The prevalence of anxiety is estimated to be 18% in long-term cancer survivors versus 13% in non-cancer controls assessed similarly.35 A large study found the prevalence of distress to be 46% across the cancer continuum in 55 cancer centers in North America,36 similar to rates reported in Europe.37 Thus, while symptoms of depression and anxiety returned to normative levels in long-term survivorship, the prevalence of pain and fatigue remain high across the survivorship continuum. Rates of these symptoms demonstrate a significant unmet need for symptom monitoring and management, particularly during active treatment and in patients with advanced cancer.
Symptomatology tends to be under-recognized in clinical care, which may contribute to its high prevalence. Outside of the context of oncology, it has been noted that 31% of patients reporting chest pain, 38% reporting dyspnea, and 45% reporting cough on a clinical visit information form did not have documentation of the symptom in electronic medical record (EMR).38 A large multicenter study found that oncologists underestimated rates of patients’ symptoms, particularly among patients with a poor Karnofsky Performance Status or poor Mini Mental State score, or those who were hospitalized, recently diagnosed, or undergoing opioid titration.39 Even on a palliative care inpatient unit, nurses’ ratings of patients’ symptoms were not significantly correlated with patients’ own ratings of their symptoms.40 Providers’ perceptions of symptoms differ not only from patients but also from other providers. In a study of symptoms assessed by pairs of providers approximately one hour apart, intraclass correlations were fair to poor across a range of symptoms including neuropathy, dyspnea, diarrhea, nausea, constipation, fatigue, and vomiting.41,42 These findings demonstrate the need for clear communication and documentation of PROs between patients and providers as well as among members of the treatment team.
PROs are associated with clinically-important events across the disease trajectory. A meta-analysis of 21 studies reported that PROs including symptoms and quality of life were significantly associated with radiographic tumor response to chemotherapy, radiotherapy, and/or targeted therapy.43 Regarding progression, Denis and colleagues have published a series of studies demonstrating that patient-reported symptoms of lung cancer (e.g., fever, cough) can identify cancer progression early (see Figure 2).44–46 A randomized trial showed that screening for these symptoms was associated with a 7 month survival advantage over usual care, which may have been due in part to the fact that patients in the intervention group had better performance status at progression and thus were more likely to receive optimal treatment.47,48 Lastly, several studies have shown that PROs enhance prediction of survival in myelodysplastic syndromes,49 multiple myeloma,50 early stage colorectal cancer,51 advanced breast cancer,52 metastatic castration-resistant prostate cancer,53 metastatic renal cell carcinoma,54 advanced cancers,55,56 and a variety of tumor types.57,58 In fact, some data suggest that PROs predict survival better than provider-rated performance status.59
Figure 2.
A case study by Denis and colleagues47 demonstrating changes in patient-reported outcomes (PROs) during treatment and progression of Stage IV lung adenocarcinoma. Severity of PROs is shown by light green=none, dark green=mild, yellow=moderate, and red=severe. The patient received first-line chemotherapy, then experienced two relapses, after which second-line chemotherapy and immunotherapy were administered, respectively. PROs become more severe and numerous as time goes on, with increases in severity prior to diagnosis of progression.
The Evidence Base for Clinical Monitoring of Patient-Generated Health Data
There is a growing literature base suggesting that incorporating PGHD into clinical care can improve outcomes over standard care.60 Evidence for clinical collection of PGHD comes primarily from studies of symptom monitoring interventions of PROs, which are defined for the purposes of this review as those intended to improve communication between patients and providers regarding patients’ symptoms. Numerous randomized trials have shown that clinic-based symptom monitoring improves patient-provider communication and increases the concordance of their ratings of symptoms and quality of life.61–69 As noted above, these benefits are consistent with additional studies suggesting that symptom management or palliative care improves survival.26,27,61–70 The mechanisms by which these benefits occur are currently unclear, although they may be due to better medication adherence and/or physical functioning, such that patients are able to receive more therapy.47 Notably, improvements in outcomes have generally occurred without lengthening clinic visits.62,64 Findings are less consistent regarding whether clinic-based symptom monitoring improves PROs, although several studies have demonstrated a beneficial effect on quality of life and/or symptomatology.26,64,67,71,72 Velikova and colleagues64 found greater improvement in quality of life for patients in the intervention group when PRO data were explicitly discussed in the clinical encounter than for corresponding patients whose PROs were not explicitly discussed. A similar finding was reported by Carlson and colleagues,73 in which the proportion of lung cancer patients with high distress levels were most impacted when distress screening was combined with telephone triage and resource referral, as opposed to screening alone. In contrast, there were no group differences in the percentage reporting high distress levels among breast cancer patients, who reported less baseline distress than lung cancer patients.73 Similarly, McLachlan and colleagues74 reported that, for patients in their randomized trial who reported moderate to severe depression at baseline, patients receiving clinic-based screening for unmet needs with care coordination exhibited greater reductions in their depression scores compared to patients randomized to conventional clinical care. Thus, clinic-based symptom monitoring may be most beneficial in patients with high symptomatology and when the data collected are discussed during the clinical encounter. Current literature is consistent with numerous quality of care initiatives, such as the Quality Oncology Practice Initiative (QOPI) from the American Society of Clinical Oncology (ASCO) or Electronic Clinical Quality Measures (eCQMs) from Medicare, in which EMR documentation of screening and management of PROs such as pain and distress are indicators of high-quality cancer care.75,76
Results of randomized trials of remote symptom monitoring of PROs have been more equivocal. We define remote symptom monitoring interventions as those intended to improve communication between patients and providers regarding patients’ symptoms when patients are away from the clinic or hospital. Remote symptom monitoring often utilizes ecological momentary assessment, defined as real-time PRO reporting, that may be less subject to recall bias than retrospective questionnaires completed in clinic.77 Although some trials have reported improvements in symptoms and quality of life,78–80 others have not.81–86 Mixed findings may be due to high study heterogeneity in terms of remote monitoring methodology, patient-provider communication, and the patient population. Remote monitoring methodology has included study-initiated calls from an automated telephone system,80 patient-initiated calls to an automated telephone system,78,81,85 study-initiated online questionnaires,25,82 patient-initiated smartphone-based or online questionnaires,83,87 study-initiated telephone calls from a nurse or nurse practitioner,84,86 study-initiated calls from a research coordinator,88 or a paper symptom diary.79 Some studies incorporated automated alerts about severe or worsening symptoms to providers,78,80,81,83,85,89 whereas others encouraged patients to discuss symptoms with their providers.79 Most studies focused on patients receiving chemotherapy or radiation78,81,83,84,90 or post-surgical patients,80,86,89 although some focused instead on advanced cancer patients79,85 or post-treatment cancer survivors.82 Notably, although some trials reported on process variables such as patient uptake of the intervention and provider responses to alerts,80,81,85 no studies systematically examined important health system factors such as type of clinic or provider workflows. Nevertheless, in general, efficacious remote monitoring interventions were those that focused on patients with high unmet needs amenable to intervention, high response rates by providers to alerts, and structured provider responses that focused on symptom reduction. Taken together, these data suggest that thoughtfully-designed remote symptom monitoring interventions may improve outcomes in cancer patients. In addition, numerous studies have shown that remote symptom monitoring is feasible and acceptable to patients and providers.81,85,87,89–94 However, more work is clearly needed to identify characteristics of health systems, patient populations, and intervention designs to ensure that remote symptom monitoring is maximally efficacious.95,96
Digital Phenotyping: Mobile Technology to Collect Patient-Generated Health Data
The limited use of large-scale collection of PGHD for clinical and research purposes is somewhat surprising given the abundance of opportunities to collect such data. Slow uptake may result from challenges in analyzing these data, as described below. Nevertheless, there has been a proliferation of device-based apps to track almost any aspect of health and behavior (i.e., digital phenotyping). Currently, approximately 81% of Americans own a smartphone and 17% own a smart watch.97 Smartphone and smart watch owners tend to be younger and have a higher SES than non-owners.97 Thus, there is concern that reliance on PGHD from these devices could exacerbate health disparities,98,99 an issue which must be considered and addressed as PGHD collection increases. Despite this concern, commercially-available devices provide a robust opportunity to refine collection and analysis of PGHD, which in many cases has comparable reliability and validity to medical-grade biometric sensors. Device-based biometric, behavioral, real-time reporting, and performance measurement approaches are described below.
Biometrics:
The field of device-based biometric sensors is rapidly expanding. Examples include wireless scales, blood pressure cuffs, thermometers, pulse oximeters, heart rate monitors, and blood glucose meters. These typically transmit data to a smartphone app that records the readings. Smart watches are also increasingly incorporating these capabilities, such as electrocardiogram (ECG). Initial published studies have shown good accuracy in comparison to gold standard ambulatory ECG, making them useful for screening of atrial fibrillation and other cardiac issues.100–102 Additional features in development are smartphone-based spirometry, video plethysmography, and detection of seizures.103–105 With the exception of blood glucose monitors which face higher regulatory scrutiny, most products are regulated by the FDA as Class II medical devices, indicating that they are safe to use but not verifying their accuracy. In light of the increasing demand for device-based health monitoring, it is clear that biometric monitoring will continue to evolve in terms of accuracy and functionality.
Physical Activity:
Accelerometers have been used to quantify human physical activity in the context of research since the 1980s.106 However, commercially-available wrist-worn activity trackers have only recently caught up with research-grade devices in terms of data accuracy.107–109 The increasing ubiquity of smartwatches provides new opportunities for passive monitoring of physical activity as a remote indicator of overall health and well-being in cancer patients. Accelerometers located in smartphones can also be used to detect a variety of physical activities including walking, jogging, going up and down stairs, and sitting.110,111 Notably, the acceptability of smartphones has been shown to be superior to that of wrist-based activity trackers.112 Several observational studies have shown that higher levels of physical activity are associated with reduced risk of all-cause mortality and/or cancer-specific mortality in patients with local or regional breast cancer,112–115 colorectal cancer,116,117 locally-advanced non-small cell lung cancer,118 and other cancer types.119,120 Higher levels of physical activity are also associated with reduced risk of several of these cancers, whereas sedentary time is associated with higher risk, independent of physical activity.121 In addition, in a meta-analysis of over one million adults from the general population, higher levels of physical activity mitigated the negative association between sitting time and all-cause mortality.122 Thus, remote monitoring using accelerometers could be used clinically to encourage physical activity among cancer patients.
In contrast, decreases in physical activity may indicate new-onset mental or physical health concerns among cancer patients. For example, physical slowing is a symptom of depression. Research shows that among individuals without cancer, depression is associated with subsequent sedentary behavior and activity disruption.123–126 Sedentary behavior and smartphone use have also been found to be sensitive indicators of stress and depressive mood in non-cancer populations, such as college students.127 Sedentary behavior, together with longer screen time on a smartphone, was found to identify symptom burden among cancer patients with an accuracy of 88%.128 Moreover, sudden decreases in physical activity during active treatment may indicate acute toxicities such as renal insufficiency, pneumonitis, and gastritis, although more research needs to be conducted on these relationships.
Location:
Patients’ community mobility may also indicate overall mental and physical well-being. Several methods have been developed to measure location using smartphones, including global positioning system (GPS), Wi-Fi, and Google Location History.129–132 Location may be a proxy for a diversity of daily activities outside the home that indicate good quality of life, including social, leisure, and work activities as well as activities of daily living (e.g., shopping). Regarding mental health, in a study of adults without cancer, depression was detected with an accuracy of 87% using GPS measurements of movement, mobility between favorite locations, and movement independent of location.133 Another study found that GPS monitoring in combination with location data from the search-and-discovery app Foursquare identified depression and anxiety with accuracy of 88%, better than either alone.134 Regarding physical health, better physical quality of life and less pain have been associated with number of trips outside the home in patients recovering from stroke and spinal surgery, respectively.135,136 GPS can also be used to track outdoor physical activity such as walking, which has been found to occur less often in non-small cell lung cancer patients as compared to similar individuals without cancer.137 Outdoor walking capacity using GPS has found to be strongly associated with clinic-based physical performance measures such as the six-minute walk test.138 Thus, there is significant potential to use location data as a way to remotely monitor cancer patients’ quality of life.
Diet:
Dietary habits have been notoriously difficult to accurately capture due to the use of long retrospective questionnaires that can be susceptible to recall bias and social desirability bias.139 Due to participant burden, dietary habits are typically measured relatively infrequently in epidemiologic studies, which may impact study findings. Web- and app-based dietary measures offer opportunities for improved measurement.140,141 Participant burden can be reduced through use of branching logic for question presentation. Additionally, accuracy of recall may be improved through use of photos of portion sizes that would be difficult to present on paper-and-pencil-based measures.142–144 Although not fully developed yet, the use of a smartphone camera together with machine learning algorithms to identify foods and portions may be a less-burdensome way to collect dietary information in the future.145,146 Nevertheless, studies of device-based measurement of dietary habits tend to suffer from high rates of attrition, which itself may bias results. Assessment of diet is clinically important, however. A diet that is low in saturated- and trans-fat and high in fiber, vegetables, fruits, and other nutrients may reduce risk of all-cause and cancer-specific mortality after diagnosis of breast or colorectal cancer,147–150 although evidence is limited in patients with other cancer types.151,152 Interestingly, patients who improved their diet after colorectal cancer diagnosis also demonstrated lower cancer-specific mortality,150 suggesting that Web- and app-based dietary measurement could be used in the context of clinical intervention to improve cancer outcomes.
Sleep:
There is a large body of literature to support the reliability and validity of sleep assessment using research-grade, wrist-worn accelerometers.153 Outputs include amount of time awake after sleep onset (WASO), sleep efficiency (i.e., percentage of time in bed spent sleeping), and total sleep time. Traditionally, assessing sleep stages (e.g., REM sleep) required polysomnography in sleep laboratories. Over time, the validity of consumer-grade wearable devices has improved, and some devices now include a heart-rate sensor which enables detection of some sleep stages. Some of the more recent devices also demonstrate similar agreement with gold-standard polysomnography and research-grade devices.154–157 However, one drawback of wrist-worn devices is that they will likely need to be recharged during the assessment period. Project Baseline has instead used a piezoelectric sensor placed under the mattress to detect sleep, which has demonstrated high validity and does not require recharging.158 A less accurate, but very low burden, detection method is to use smartphone screen interaction to approximate bedtimes and waking times.159 Smartphones have also shown promise for evaluation of perceived sleep quality using patient reports as well as detection of sleep-disordered breathing using built-in microphones.160–163 In addition, the MyHeart Counts study from Apple uses smartphones to detect ambient light.164 Nevertheless, until battery life and sleep sensor technology improve in commercially-available wearables and smartphones, alternate measures of sleep may need to be utilized.
Real-Time Reporting:
Mobile devices provide an outstanding opportunity to capture real-time patient-reported data. There is a large literature base focused on ecological momentary assessment (EMA) of symptoms such as pain, fatigue, and depression in real-time. Although burdensome for patients relative to passive monitoring of behaviors, EMA can provide important information regarding daily and intra-day variability in symptoms without recall bias.165 For example, using real-time, validated, smartphone-based “brain games,” Small and colleagues166 demonstrated significant intraday cognitive variability in breast cancer patients. Cognitive variability may be a more sensitive indicator of cancer-related cognitive impairment than office-based neuropsychological assessment, as it precedes cognitive decline in normal aging.167,168 Smartphone-based cameras are increasingly used to remotely assess dermatologic conditions or surgical complications,169–172 and have the potential to identify complications early, providing better, lower-cost care. Real-time reporting can facilitate just-in-time or micro-interventions, cognitive-behavioral interventions that can be delivered via smartphone or smartwatch to deliver interventions personalized to an individual’s current circumstances or environment.173,174 Examples include reminders to exercise during periods of inactivity or smoking cessation texts to suggest coping strategies during reported nicotine cravings. Lastly, an innovative clinical trial has used smartphone-based real-time symptom reporting to manage dose-limiting toxicities of cediranib and olaparib for ovarian cancer, a regimen that can cause rapid-onset hypertension and diarrhea.7 There is untapped potential for remote symptom monitoring in clinical trials to keep patients on drug longer, with potential survival benefits.
Functional Status:
A sizable body of research has demonstrated that smartphones can reliably detect gait, physical performance, range of motion, and falls. A smartphone placed in a pants pocket can detect stride times, a clinically-meaningful metric of locomotor control, with comparable accuracy to gold-standard clinic-based instrumentation.175 Interestingly, measurement of gait parameters is equally reliable and valid regardless of whether the smartphone is worn on the body, on a belt, or in a pocket or purse.176,177 Two- and six-minute walk tests can be administered remotely via smartphone with comparable results to clinic-based tests.178–180 The same is true for the Timed Up and Go (TUG) Test, in which a patient is asked to stand up from a chair without using their arms, walk a set distance, return, and sit down again.181,182 Range of motion in the wrist183 and ankle184 can also be captured reliably via smartphone. Additionally, falls can be distinguished from daily actions with an accuracy of 90% using a smartphone-based accelerometer and gyroscope.185,186 Similar to gait detection, the accuracy of fall detection does not appear to differ based on the placement of the smartphone on a belt or in a pocket.187 Use of a smart watch together with a smartphone can significantly decrease the rate of false positives when detecting falls.188 Remote monitoring of functional status may be particularly useful for telemedicine visits and in geriatric populations.
Big Data: Untapped Potential for Patient-Generated Health Data
Digital phenotyping can yield very large datasets amenable to big data analytics. The term “big data” is defined by Google as “extremely large data sets that may be analyzed computationally to reveal patterns, trends, and associations, especially relating to human behavior and interactions.”189 Initial forays into big data in the context of healthcare have been limited primarily to analysis of clinical information, payment and billing data, genomics, and biomarkers. Little has been done specifically in PGHD. Historically, a primary concern regarding sharing of health data has been patient privacy. The U.S. Healthcare Insurance Portability and Accountability Act (HIPAA) of 1996 and later laws focused on regulating electronic storage, transmission, disclosure, and reporting of security breaches of identifiable health information, as well as enforcement mechanisms for healthcare systems that did not comply.190 The U.S. Health Information Technology for Economic and Clinical Health (HITECH) Act of 2009 developed a system of incentives and penalties for healthcare systems to implement electronic medical records (EMR) systems.191 The widespread implementation of EMRs has enabled large-scale pooling of deidentified data from patients outside the context of a clinical trial. Concurrently, it has become increasingly clear that very large datasets would be required to detect and understand the complex genetic and biological mechanisms of cancer.
At the same time, a consensus emerged that the field of medicine had an ethical obligation not just to protect patients’ privacy but also to aggregate and learn from their data. This position was articulated in 2006 in a publication from the IOM entitled “The Learning Healthcare System.”192 Alarmed at the rising costs of healthcare, the IOM recognized an urgent need for data to optimize the effectiveness and value of medical decisions. The existing framework of randomized clinical trials and regulatory approvals was too slow and costly to provide evidence for more than a sliver of decisions made by healthcare providers on a daily basis. As a result, the IOM proposed using real-world evidence to fill in knowledge gaps in the fast-changing healthcare landscape. The IOM recognized that progress towards this goal would rely on “the emergence of linked clinical information systems that might allow information about safety and effectiveness to emerge naturally in the course of care.”192 A follow-up IOM conference193 set forth a rapid-learning systems model specifically for oncology. The 2009 conference provided a more detailed vision of an iterative process in which clinical data are systematically collected and analyzed, with the resulting insights implemented into clinical care. Changes in outcomes are then measured and form the basis for new hypotheses, analyses, and adjustments to care.193 Although rapid-learning systems were described with a focus on clinical data, it is not hard to envision incorporation of PGHD as well. The vision of a rapid learning system relies heavily on advancements in large-scale data aggregation, harmonization, and analysis that are only now starting to be realized.
Recent large-scale data collection initiatives can be seen across government, advocacy, and academic sectors. Regarding government, in 2015 President Obama announced a new Precision Medicine Initiative, one focus of which was to create infrastructure for open data sharing. The 21st Century Cures Act in 2016 provided $1.8 billion in funding to support the Cancer Moonshot Initiative to build a national cancer data infrastructure, establish networks for patients to directly contribute tumor profile and PRO data, and analyze and biospecimens from past clinical trials to predict future patient outcomes. The National Institutes of Health (NIH) is also directing the All of Us research program, an ambitious cohort study of a million or more individuals of diverse backgrounds living in the United States.194 Participants will be followed for ten years; data collected include patient-reported surveys, clinical data from the EMR, and biospecimens.194 The NIH also maintains a wide variety of additional federal health databases of clinical, genomic, imaging and epidemiological data from federally-funded research projects.195 These federal initiatives are consistent with those launched by advocacy groups. One example is ASCO’s CancerLinQ, a platform for sharing and analyzing oncology data from healthcare IT systems.196 Currently consisting of over a million records from 2,000 oncology care providers, the goals of CancerLinQ are to improve quality of care and facilitate research.197 Other advocacy groups, such as the GO2 Foundation for Lung Cancer, have patient registries in which patients can share data with researchers.198 Indeed, cancer patients express willingness to provide PGHD to cancer registries if data are kept confidential.199 Academic institutions are also undertaking large data collection efforts. An example is the Oncology Research Information Exchange Network (ORIEN), a partnership among 19 academic cancer centers to collect and aggregate deidentified molecular, clinical, and epidemiological data.200
The private sector has launched parallel initiatives. For example, Flatiron Health created a computing platform to provide EMR, billing, analytics, and clinical trial screening capabilities for community oncology practices. Curated clinical data from the platform flow into a data warehouse, which can be mined to monitor quality of care, examine patterns of real-world treatment utilization, and create in silico control groups for clinical trials. Another example from the private sector is Verily’s Project Baseline Health Study, a four-year longitudinal study of 10,000 individuals living in the United States. Data to be collected include biometric data (e.g., electrocardiogram data, electrodermal data, heart rate, sleep quality) from Google Watch, genetic information, blood samples, clinical data from annual in-person study visits with healthcare providers, and PROs. This project has recognized the vast analytic possibilities engendered by mobile and wearable devices. Google has also partnered with healthcare providers such as Ascension, an organization consisting of 2,600 hospitals, doctors’ offices, and other facilities, to aggregate patients’ health histories. The goal of this initiative, called Project Nightingale, is to design new artificial intelligence algorithms to support patient care. Similar initiatives are underway at Apple, Microsoft, Amazon, and other companies. An example is the Apple Health Records application programming interface (API), under development as part of HealthKit, an app that aggregates and presents health information from iPhone, Apple Watch, and third-party apps. Health Records can link to the EMR or patient portal and display information on health conditions, lab results, immunizations, medication, procedures, and other data from participating medical providers. Apple also allows users to share health data with researchers. More broadly, APIs allow for data from devices and wearable sensors to be uploaded to the EMR or data warehouses with the patient’s permission. Thus, preliminary infrastructure resources needed for large-scale PGHD collection have been established, from which additional APIs can be developed. In the meantime, most large aggregated datasets are typically comprised of clinical and molecular data with little to no PGHD.
Data Mining, Natural Language Processing, and Artificial Intelligence Using Big Data: A Promising Approach for Analysis of PGHD
The term artificial intelligence was first coined in 1956 as “the conjecture that every aspect of learning or any other feature of intelligence can in principle be so precisely described that a machine can be made to simulate it.”201 Concurrently, there was recognition that algorithmic decision-making might actually surpass the accuracy and reliability of human judgment, an assertion famously proposed by the psychologist Paul Meehl in 1954202 which has been supported by an impressive array of data.203–205 Although AI was dismissed by the New England Journal of Medicine in 1987206 because “the field of medicine is so broad and complex that it is difficult, if not impossible, to capture the relevant information in rules,” it experienced a resurgence in the early 2010’s. This development was due to the convergence of three major trends: 1) improvements in computing power; 2) development of new AI algorithms; and, 3) increases in the number and quality of big data sets for training.207 The first two trends allowed scientists to construct and train dramatically larger and more complex “deep” neural networks. The size and quality of training data, meanwhile, allowed these networks to achieve high accuracy at several narrow-domain tasks. One early application was classification of images,208 followed shortly by labeling and outlining (semantic segmentation) of multiple objects within images.208 These developments were transferred and applied by biomedical scientists to classification of medical images and detection of skin cancer.209,210 There has also been increasing application of machine learning to PGHD. For example, two recent studies have demonstrated that PGHD in the form of internet search logs of symptoms facilitate early identification of cases of pancreatic cancer and lung cancer.211,212 Machine learning has also been applied to PROs to predict the course of multiple sclerosis213,214 and recovery after hip and knee replacement.215
Artificial intelligence is rapidly evolving. For example, in late 2018 scientists at Google published a powerful new algorithm for natural language processing: Bidirectional Encoder Representations from Transformers (BERT),216 a powerful new algorithm for natural language processing (NLP) developed by Google. BERT set new performance records across nearly all NLP benchmarks, and for the first time, achieved human level performance on several of these. BERT and related technologies are being applied to unstructured biomedical217,218 and clinical texts.219,220 Translation of these new technologies into the clinical domain is still in the early stages but progress is accelerating. In 2018 the FDA developed a “fast-track” approval process for AI-based medical technologies.221 By late 2019 the FDA had cleared 26 AI-based tools for marketing and use in the US.222 Although there are no commercial AI tools to our knowledge focused on PGHD in the context of cancer, we expect such tools to appear shortly. Sufficient computing power and algorithms are widely available. These should allow scientists to identify and extract new information from PROs, wearable sensors, and EMR notes.
Challenges and Opportunities for Clinical Integration of Patient-Generated Health Data
Although advancements in technology have the potential to revolutionize medicine through collection and analysis of PGHD, clinical integration of PGHD has lagged behind. Challenges to clinical integration include data linkage and scaling across healthcare systems, provider engagement, and actionability. A significant barrier to data linkage and scaling across healthcare systems is the inflexibility of most EMR platforms. While commonly-used EMRs in oncology such as EPIC are starting to incorporate PRO measures, uptake of both PROs and biometric data has been limited by usability issues of the EMR platforms themselves. As the IOM noted in 2013,223 “Originally designed for billing and coding purposes, health IT systems have not been integrated efficiently into clinical care, do not facilitate the coordination of care, and the need to customize local systems has created a situation where health IT systems cannot communicate with each other… Many of these systems are inflexible and thus are unable to adapt to the changing needs of a modern health care system.” When collected as part of clinical care, PGHD may be buried in the clinical record, with poor visibility and interpretability. For example, Rotenstein and colleagues224 reported barriers to use of PRO data in a large radiation oncology clinic including difficulty accessing the data in the EMR (60%), difficulty bringing the data into the treatment note (48%), too much data (38%), and difficulty interpreting data (26%).224 Notably, these barriers also apply to remotely-collected biometric data and could be addressed through better EMR integration and visualization. In recognition of such barriers to clinical integration, the National Cancer Institute’s Center for Strategic Scientific Initiatives included in its 2020 provocative questions the need to identify new ways to integrate PGHD into the EMR. The development of the Fast Healthcare Interoperability Resources (FHIR) data standard and APIs provides opportunities to flexibly create software that securely pulls discrete data from the EMR into third-party software. FHIR could be used to improve presentation and visualization of PGHD in the form of both PROs and remotely-collected biometric data, although EMR companies must enable FHIR capability. Recent trends in information technology (IT) define FHIR as an approach suitable for citizen developers, in which applications are developed by non-experts with the support and approval of cooperating IT experts.225 Driving this trend is the development of low-code/no-code solutions, which enable users with little to no programming background to create and interact with the applications they need. Numerous potential benefits include rapid development, no need to wait for developers, and solutions that solve the problem at hand. There are also potential drawbacks, including security, scalability, and limited use for complex application. These IT trends may improve the access and impact of the collected data for patients, researchers, and physicians. Developers of such software should draw heavily from research in the fields of user-focused design and human-computer interaction to create intuitive data visualizations that focus on important clinical information and allow providers to identify patterns of data that provide important clinical insights. Although some initial research has been conducted on visualization of PGHD,226–229 transdisciplinary collaborations including citizen developers, graphic designers, cognitive scientists, bioinformaticians, biostatisticians, data scientists, and computer scientists will be needed to create the frictionless user experience necessary for optimal uptake.
Despite challenges with EMR integration, clinical collection of PROs in oncology results in high provider satisfaction.224,230–232 High rates of satisfaction have been reported despite initial provider concerns. These initial concerns commonly focus on the time required to collect and address PROs in clinic, lack of training regarding interpretation and management of some PROs, patient burden, and liability issues if PROs are overlooked.233–235 Thus, an important aspect of successful clinical implementation of PROs is addressing provider concerns, including appropriate provider training and clinical decision support.13,14,19,236,237 Concerns may also be ameliorated in part by communicating observed benefits of PROs reported by other providers. PROs can contribute to shared decision-making and strengthen rapport between patients and providers by facilitating better communication.232 Interestingly, PROs may also reduce provider burnout by contributing to more efficient clinical workflow.232 Because providers arrive to the clinical encounter with information regarding patient concerns, they can spend more time addressing these concerns in a meaningful way rather than running through checklists of questions.232 PRO data may also be less vulnerable than traditional clinical assessments to “white coat syndrome,” in which patients are less likely to report symptoms during an interview than on paper.238 Following clinical rollout of PROs in a radiation oncology setting, over half of the 53 providers surveyed reported no or rare impact of PROs on their ability to see patients on time and a majority would recommend PROs to a colleague at another institution.224
Several steps can be taken to address actionability concerns as well. Perhaps most importantly, collection and use of PGHD to drive clinical decisions should be incorporated into medical boards and continuing education. Training should be extended to physicians, advanced practice professionals, nurses, and medical assistants. Training should include both management of PROs and use of PGHD to support shared decision-making. For example, Thomas Jefferson University has a graduate certificate program in digital health. Organizations such as ASCO and the National Comprehensive Cancer Network (NCCN) provide detailed, evidence-based guidelines for management of PROs including nausea and vomiting, fatigue, distress, and general supportive care. Some PRO data collection apps, such as Carevive, use algorithms based on these guidelines to suggest self-management strategies.239 Measure-specific algorithms for identifying severe symptoms and clinically-significant worsening of symptoms have been well-articulated.240,241 The necessary elements are in place to determine actionability of PROs. Better provider education and well-integrated clinical decision support remain to be addressed, however. Payer reimbursement for symptom management, particularly in the realm of telehealth and digital therapeutics, is also needed. Efforts to address these issues should include the perspectives of multiple stakeholders, including patients, families, providers, payers, EMR companies, and IT developers.242
Summary and Future Directions
The past decade has seen unprecedented progress in the war on cancer in both therapeutic and technological domains. We now have the opportunity to integrate these accomplishments by creating new, data-driven approaches using PGHD to identify and intervene early on clinically-significant events such as toxicity and cancer progression, which may in turn reduce emergency room visits and hospitalizations. At the same time, technology allows closer contact with patients outside of the clinical encounter to facilitate healthy lifestyle behaviors, symptom management, and medication adherence. There are still many technological, analytic, and workflow challenges that need to be overcome to incorporate PGHD into routine research and cancer care, however.13,17,242–244 The evidence base to support use of biometric data is sparse relative to that for PROs. In addition, PGHD must be implemented in a way that prevents exacerbation of health disparities in oncology care. Nevertheless, as with treatment discoveries, although each intervention is incremental, together PHGD-based interventions can dramatically improve both quality and quantity of life.
References
- 1.U. S. Department of Health Human Services. What are patient-generated health data? https://www.healthit.gov/topic/otherhot-topics/what-are-patient-generated-health-data. Published 2019. Accessed October 15, 2019.
- 2.Shapiro M, Johnston D, Wald J, Mon D Patient-generated health data. White paper. In: Research Triangle Park, NC: RTI International. Prepared for Office of Policy and Planning, Office of the National Coordinator for Health Information Technology; 2012: https://www.rti.org/publication/patient-generated-health-data-white-paper. [Google Scholar]
- 3.U.S. Department of Health and Human Services. Guidance for Industry Patient Reported Outcomes Measures: Use in Medical Product Development to Support Labeling Claims. https://www.fda.gov/media/77832/download. Published 2009. Accessed October 19, 2018.
- 4.Kluetz PG SA, Papadopoulos EJ, Johnson LL, Donoghue M, Kwitkowski VE, Chen WH, Sridhara R, Farrell AT, Keegan P, Kim G, Pazdur R. Focusing on Core Patient-Reported Outcomes in Cancer Clinical Trials: Symptomatic Adverse Events, Physical Function, and Disease-Related Symptoms. Clinical Cancer Research. 2016;22(7):1553–1558. [DOI] [PubMed] [Google Scholar]
- 5.Grossman C Patient reported outcomes: Design with the end in mind. Millken Institute; http://milkeninstitute.org/reports/patient-reported-outcomes-design-end-mind. Published 2018. Accessed Oct. 19, 2018. [Google Scholar]
- 6.Rivera SC, Kyte DG, Aiyegbusi OL, Slade AL, McMullan C, Calvert MJ. The impact of patient-reported outcome (PRO) data from clinical trials: a systematic review and critical analysis. Health and Quality of Life Outcomes. 2019;17(1):156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Liu JF, Lee JM, Strock E, et al. Technology Applications: Use of Digital Health Technology to Enable Drug Development. JCO Clin Cancer Inform. 2018;2:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Fromme EK, Eilers KM, Mori M, Hsieh YC, Beer TM. How accurate is clinician reporting of chemotherapy adverse effects? A comparison with patient-reported symptoms from the Quality-of-Life Questionnaire C30. J Clin Oncol. 2004;22(17):3485–3490. [DOI] [PubMed] [Google Scholar]
- 9.Di Maio M, Gallo C, Leighl NB, et al. Symptomatic toxicities experienced during anticancer treatment: agreement between patient and physician reporting in three randomized trials. J Clin Oncol. 2015;33(8):910–915. [DOI] [PubMed] [Google Scholar]
- 10.Dueck AC, Scher HI, Bennett AV, et al. Assessment of Adverse Events From the Patient Perspective in a Phase 3 Metastatic Castration-Resistant Prostate Cancer Clinical Trial. JAMA Oncol. 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Unger JM, Barlow WE, Martin DP, et al. Comparison of survival outcomes among cancer patients treated in and out of clinical trials. J Natl Cancer Inst. 2014;106(3):dju002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Gross CP, Filardo G, Mayne ST, Krumholz HM. The impact of socioeconomic status and race on trial participation for older women with breast cancer. Cancer. 2005;103(3):483–491. [DOI] [PubMed] [Google Scholar]
- 13.LeBlanc TW, Abernethy AP. Patient-reported outcomes in cancer care - hearing the patient voice at greater volume. Nat Rev Clin Oncol. 2017;14(12):763–772. [DOI] [PubMed] [Google Scholar]
- 14.Austin E, LeRouge C, Hartzler AL, Segal C, Lavallee DC. Capturing the patient voice: implementing patient-reported outcomes across the health system. Qual Life Res. 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Rivera SC KD, Aiyegbusi OL, Slade AL, McMullan C, Calvert MJ. The impact of patient-reported outcome (PRO) data from clinical trials: a systematic review and critical analysis. Health and Quality of Life Outcomes. 2019;17(1):156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Basch E, Geoghegan C, Coons SJ, et al. Patient-Reported Outcomes in Cancer Drug Development and US Regulatory Review: Perspectives From Industry, the Food and Drug Administration, and the Patient. JAMA Oncol. 2015;1(3):375–379. [DOI] [PubMed] [Google Scholar]
- 17.Murthy HS, Wood WA. The Value of Patient Reported Outcomes and Other Patient-Generated Health Data in Clinical Hematology. Curr Hematol Malig Rep. 2015;10(3):213–224. [DOI] [PubMed] [Google Scholar]
- 18.Moss HA, Havrilesky LJ. The use of patient-reported outcome tools in Gynecologic Oncology research, clinical practice, and value-based care. Gynecol Oncol. 2018;148(1):12–18. [DOI] [PubMed] [Google Scholar]
- 19.Mooney K, Berry DL, Whisenant M, Sjoberg D. Improving Cancer Care Through the Patient Experience: How to Use Patient-Reported Outcomes in Clinical Practice. Am Soc Clin Oncol Educ Book. 2017;37:695–704. [DOI] [PubMed] [Google Scholar]
- 20.Institute of Medicine. Ensuring Quality Cancer Care. Washington D.C.: National Academies Press; 1999. [PubMed] [Google Scholar]
- 21.Hoerger M, Wayser GR, Schwing G, Suzuki A, Perry LM. Impact of Interdisciplinary Outpatient Specialty Palliative Care on Survival and Quality of Life in Adults With Advanced Cancer: A Meta-Analysis of Randomized Controlled Trials. Ann Behav Med. 2019;53(7):674–685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Bakitas MA, Tosteson TD, Li Z, et al. Early Versus Delayed Initiation of Concurrent Palliative Oncology Care: Patient Outcomes in the ENABLE III Randomized Controlled Trial. J Clin Oncol. 2015;33(13):1438–1445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Temel JS, Greer JA, Muzikansky A, et al. Early palliative care for patients with metastatic non-small-cell lung cancer. N Engl J Med. 2010;363(8):733–742. [DOI] [PubMed] [Google Scholar]
- 24.El-Jawahri A, Traeger L, Greer JA, et al. Effect of Inpatient Palliative Care During Hematopoietic Stem-Cell Transplant on Psychological Distress 6 Months After Transplant: Results of a Randomized Clinical Trial. J Clin Oncol. 2017;35(32):3714–3721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.El-Jawahri A, Greer JA, Pirl WF, et al. Effects of Early Integrated Palliative Care on Caregivers of Patients with Lung and Gastrointestinal Cancer: A Randomized Clinical Trial. Oncologist. 2017;22(12):1528–1534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Basch E, Deal AM, Kris MG, et al. Symptom Monitoring With Patient-Reported Outcomes During Routine Cancer Treatment: A Randomized Controlled Trial. J Clin Oncol. 2016;34(6):557–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Basch E, Deal AM, Dueck AC, et al. Overall Survival Results of a Trial Assessing Patient-Reported Outcomes for Symptom Monitoring During Routine Cancer Treatment. JAMA. 2017;318(2):197–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Davis C, Naci H, Gurpinar E, Poplavska E, Pinto A, Aggarwal A. Availability of evidence of benefits on overall survival and quality of life of cancer drugs approved by European Medicines Agency: retrospective cohort study of drug approvals 2009–13. BMJ. 2017;359:j4530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Bubis LD, Davis L, Mahar A, et al. Symptom Burden in the First Year After Cancer Diagnosis: An Analysis of Patient-Reported Outcomes. J Clin Oncol. 2018;36(11):1103–1111. [DOI] [PubMed] [Google Scholar]
- 30.van den Beuken-van Everdingen MH, Hochstenbach LM, Joosten EA, Tjan-Heijnen VC, Janssen DJ. Update on Prevalence of Pain in Patients With Cancer: Systematic Review and Meta-Analysis. J Pain Symptom Manage. 2016;51(6):1070–1090 e1079. [DOI] [PubMed] [Google Scholar]
- 31.Abrahams HJ, Gielissen MF, Schmits IC, Verhagen CA, Rovers MM, Knoop H. Risk factors, prevalence, and course of severe fatigue after breast cancer treatment: a meta-analysis involving 12 327 breast cancer survivors. Ann Oncol. 2016;27(6):965–974. [DOI] [PubMed] [Google Scholar]
- 32.Van Lancker A, Velghe A, Van Hecke A, et al. Prevalence of symptoms in older cancer patients receiving palliative care: a systematic review and meta-analysis. J Pain Symptom Manage. 2014;47(1):90–104. [DOI] [PubMed] [Google Scholar]
- 33.Ahn SH, Park BW, Noh DY, et al. Health-related quality of life in disease-free survivors of breast cancer with the general population. Annals of Oncology. 2007;18(1):173–182. [DOI] [PubMed] [Google Scholar]
- 34.Krebber AM, Buffart LM, Kleijn G, et al. Prevalence of depression in cancer patients: a meta-analysis of diagnostic interviews and self-report instruments. Psychooncology. 2014;23(2):121–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Mitchell AJ, Ferguson DW, Gill J, Paul J, Symonds P. Depression and anxiety in long-term cancer survivors compared with spouses and healthy controls: a systematic review and meta-analysis. Lancet Oncol. 2013;14(8):721–732. [DOI] [PubMed] [Google Scholar]
- 36.Carlson LE, Zelinski EL, Toivonen KI, et al. Prevalence of psychosocial distress in cancer patients across 55 North American cancer centers. J Psychosoc Oncol. 2019;37(1):5–21. [DOI] [PubMed] [Google Scholar]
- 37.Mehnert A, Hartung TJ, Friedrich M, et al. One in two cancer patients is significantly distressed: Prevalence and indicators of distress. Psychooncology. 2018;27(1):75–82. [DOI] [PubMed] [Google Scholar]
- 38.Pakhomov SV, Jacobsen SJ, Chute CG, Roger VL. Agreement between patient-reported symptoms and their documentation in the medical record. Am J Manag Care. 2008;14(8):530–539. [PMC free article] [PubMed] [Google Scholar]
- 39.Laugsand EA, Sprangers MA, Bjordal K, Skorpen F, Kaasa S, Klepstad P. Health care providers underestimate symptom intensities of cancer patients: a multicenter European study. Health Qual Life Outcomes. 2010;8:104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Rhondali W, Hui D, Kim SH, et al. Association between patient-reported symptoms and nurses’ clinical impressions in cancer patients admitted to an acute palliative care unit. J Palliat Med. 2012;15(3):301–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Cicchetti DV. Guidelines, criteria, and rules of thumb for evaluating normed and standardized assessment instruments in psychology. Psychological Assessment. 1994;6(4):284–290. [Google Scholar]
- 42.Atkinson TM, Li Y, Coffey CW, et al. Reliability of adverse symptom event reporting by clinicians. Qual Life Res. 2012;21(7):1159–1164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Victorson D, Soni M, Cella D. Metaanalysis of the correlation between radiographic tumor response and patient-reported outcomes. Cancer. 2006;106(3):494–504. [DOI] [PubMed] [Google Scholar]
- 44.Denis F, Koontz BF, Letellier C. Application and Benefits of Web-Mediated Symptom Reporting for Patients Undergoing Immunotherapy: A Clinical Example. Case Rep Oncol. 2018;11(3):763–768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Denis F, Viger L, Charron A, Voog E, Letellier C. Detecting lung cancer relapse using self-evaluation forms weekly filled at home: the sentinel follow-up. Support Care Cancer. 2014;22(1):79–85. [DOI] [PubMed] [Google Scholar]
- 46.Denis F, Yossi S, Septans AL, et al. Improving Survival in Patients Treated for a Lung Cancer Using Self-Evaluated Symptoms Reported Through a Web Application. Am J Clin Oncol. 2015. [DOI] [PubMed] [Google Scholar]
- 47.Denis F, Lethrosne C, Pourel N, et al. Randomized Trial Comparing a Web-Mediated Follow-up With Routine Surveillance in Lung Cancer Patients. J Natl Cancer Inst. 2017;109(9). [DOI] [PubMed] [Google Scholar]
- 48.Denis F, Basch E, Septans AL, et al. Two-Year Survival Comparing Web-Based Symptom Monitoring vs Routine Surveillance Following Treatment for Lung Cancer. JAMA. 2019;321(3):306–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Efficace F, Cottone F, Abel G, et al. Patient-reported outcomes enhance the survival prediction of traditional disease risk classifications: An international study in patients with myelodysplastic syndromes. Cancer. 2018;124(6):1251–1259. [DOI] [PubMed] [Google Scholar]
- 50.Viala M, Bhakar AL, de la Loge C, et al. Patient-reported outcomes helped predict survival in multiple myeloma using partial least squares analysis. J Clin Epidemiol. 2007;60(7):670–679. [DOI] [PubMed] [Google Scholar]
- 51.Hsu T, Speers CH, Kennecke HF, Cheung WY. The utility of abbreviated patient-reported outcomes for predicting survival in early stage colorectal cancer. Cancer. 2017;123(10):1839–1847. [DOI] [PubMed] [Google Scholar]
- 52.Smyth EN, Shen W, Bowman L, et al. Patient-reported pain and other quality of life domains as prognostic factors for survival in a phase III clinical trial of patients with advanced breast cancer. Health Qual Life Outcomes. 2016;14:52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Cella D, Traina S, Li T, et al. Relationship between patient-reported outcomes and clinical outcomes in metastatic castration-resistant prostate cancer: post hoc analysis of COU-AA-301 and COU-AA-302. Ann Oncol. 2018;29(2):392–397. [DOI] [PubMed] [Google Scholar]
- 54.Trask PC, Bushmakin AG, Cappelleri JC, et al. Baseline patient-reported kidney cancer-specific symptoms as an indicator for median survival in sorafenib-refractory metastatic renal cell carcinoma. J Cancer Surviv. 2011;5(3):255–262. [DOI] [PubMed] [Google Scholar]
- 55.Popovic G, Harhara T, Pope A, et al. Patient-Reported Functional Status in Outpatients With Advanced Cancer: Correlation With Physician-Reported Scores and Survival. J Pain Symptom Manage. 2018;55(6):1500–1508. [DOI] [PubMed] [Google Scholar]
- 56.Stukenborg GJ, Blackhall LJ, Harrison JH, Dillon PM, Read PW. Longitudinal patterns of cancer patient reported outcomes in end of life care predict survival. Support Care Cancer. 2016;24(5):2217–2224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Quinten C, Maringwa J, Gotay CC, et al. Patient self-reports of symptoms and clinician ratings as predictors of overall cancer survival. J Natl Cancer Inst. 2011;103(24):1851–1858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Quinten C, Martinelli F, Coens C, et al. A global analysis of multitrial data investigating quality of life and symptoms as prognostic factors for survival in different tumor sites. Cancer. 2014;120(2):302–311. [DOI] [PubMed] [Google Scholar]
- 59.Agarwal JP, Chakraborty S, Laskar SG, et al. Prognostic value of a patient-reported functional score versus physician-reported Karnofsky Performance Status Score in brain metastases. Ecancermedicalscience. 2017;11:779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Ishaque S, Karnon J, Chen G, Nair R, Salter AB. A systematic review of randomised controlled trials evaluating the use of patient-reported outcome measures (PROMs). Qual Life Res. 2019;28(3):567–592. [DOI] [PubMed] [Google Scholar]
- 61.Detmar SB, Muller MJ, Schornagel JH, Wever LD, Aaronson NK. Health-related quality-of-life assessments and patient-physician communication: a randomized controlled trial. JAMA. 2002;288(23):3027–3034. [DOI] [PubMed] [Google Scholar]
- 62.Berry DL, Blumenstein BA, Halpenny B, et al. Enhancing patient-provider communication with the electronic self-report assessment for cancer: a randomized trial. J Clin Oncol. 2011;29(8):1029–1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Taenzer P, Bultz BD, Carlson LE, et al. Impact of computerized quality of life screening on physician behaviour and patient satisfaction in lung cancer outpatients. Psychooncology. 2000;9(3):203–213. [DOI] [PubMed] [Google Scholar]
- 64.Velikova G, Booth L, Smith AB, et al. Measuring quality of life in routine oncology practice improves communication and patient well-being: a randomized controlled trial. J Clin Oncol. 2004;22(4):714–724. [DOI] [PubMed] [Google Scholar]
- 65.Hilarius DL, Kloeg PH, Gundy CM, Aaronson NK. Use of health-related quality-of-life assessments in daily clinical oncology nursing practice: a community hospital-based intervention study. Cancer. 2008;113(3):628–637. [DOI] [PubMed] [Google Scholar]
- 66.Nicklasson M, Elfstrom ML, Olofson J, Bergman B. The impact of individual quality of life assessment on psychosocial attention in patients with chest malignancies: a randomized study. Support Care Cancer. 2013;21(1):87–95. [DOI] [PubMed] [Google Scholar]
- 67.Ruland CM, Holte HH, Roislien J, et al. Effects of a computer-supported interactive tailored patient assessment tool on patient care, symptom distress, and patients’ need for symptom management support: a randomized clinical trial. J Am Med Inform Assoc. 2010;17(4):403–410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Takeuchi EE, Keding A, Awad N, et al. Impact of patient-reported outcomes in oncology: a longitudinal analysis of patient-physician communication. J Clin Oncol. 2011;29(21):2910–2917. [DOI] [PubMed] [Google Scholar]
- 69.Read PWBLJS, G. J; Harrison J; Barclay J; Dillon PM; Wilson DD; Showalter TN; Handsfield LL; Chen Q; Larner J. Outcomes of a Re-engineered Palliative Care and Radiation Therapy Care Model. International Journal of Radiation Oncology Biology Physics. 2016;94(1):2–3. [Google Scholar]
- 70.Barbera LC, Sutradhar R, Earle C, Mittmann N, Seow H, Howell D, Li Q, Deva T The impact of routine ESAS use on overall survival: Results of a population-based retrospective matched cohort analysis. Journal of Clinical Oncology 2019;37(15_suppl):6509–6509. [Google Scholar]
- 71.Ganz PA, Greendale GA, Petersen L, Zibecchi L, Kahn B, Belin TR. Managing menopausal symptoms in breast cancer survivors: results of a randomized controlled trial. J Natl Cancer Inst. 2000;92(13):1054–1064. [DOI] [PubMed] [Google Scholar]
- 72.Klinkhammer-Schalke M, Koller M, Steinger B, et al. Direct improvement of quality of life using a tailored quality of life diagnosis and therapy pathway: randomised trial in 200 women with breast cancer. Br J Cancer. 2012;106(5):826–838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Carlson LE, Groff SL, Maciejewski O, Bultz BD. Screening for distress in lung and breast cancer outpatients: a randomized controlled trial. J Clin Oncol. 2010;28(33):4884–4891. [DOI] [PubMed] [Google Scholar]
- 74.McLachlan SA, Allenby A, Matthews J, et al. Randomized trial of coordinated psychosocial interventions based on patient self-assessments versus standard care to improve the psychosocial functioning of patients with cancer. J Clin Oncol. 2001;19(21):4117–4125. [DOI] [PubMed] [Google Scholar]
- 75.Pirl WF, Fann JR, Greer JA, et al. Recommendations for the implementation of distress screening programs in cancer centers: report from the American Psychosocial Oncology Society (APOS), Association of Oncology Social Work (AOSW), and Oncology Nursing Society (ONS) joint task force. Cancer. 2014;120(19):2946–2954. [DOI] [PubMed] [Google Scholar]
- 76.Centers for Medicare and Medicaid Services. Clinical Quality Measures Basics. https://www.cms.gov/Regulations-and-Guidance/Legislation/EHRIncentivePrograms/ClinicalQualityMeasures. Published 2019. Accessed November 17, 2019.
- 77.Phillips KM, Faul LA, Small BJ, Jacobsen PB, Apte SM, Jim HS. Comparing the retrospective reports of fatigue using the Fatigue Symptom Index with daily diary ratings in women receiving chemotherapy for gynecologic cancer. J Pain Symptom Manage. 2013;46(2):282–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Mooney KH, Beck SL, Wong B, et al. Automated home monitoring and management of patient-reported symptoms during chemotherapy: results of the symptom care at home RCT. Cancer Med. 2017;6(3):537–546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Hoekstra J, de Vos R, van Duijn NP, Schade E, Bindels PJ. Using the symptom monitor in a randomized controlled trial: the effect on symptom prevalence and severity. J Pain Symptom Manage. 2006;31(1):22–30. [DOI] [PubMed] [Google Scholar]
- 80.Cleeland CS, Wang XS, Shi Q, et al. Automated symptom alerts reduce postoperative symptom severity after cancer surgery: a randomized controlled clinical trial. J Clin Oncol. 2011;29(8):994–1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Mooney KH, Beck SL, Friedman RH, Farzanfar R, Wong B. Automated monitoring of symptoms during ambulatory chemotherapy and oncology providers’ use of the information: a randomized controlled clinical trial. Support Care Cancer. 2014;22(9):2343–2350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Wheelock AE, Bock MA, Martin EL, et al. SIS.NET: a randomized controlled trial evaluating a web-based system for symptom management after treatment of breast cancer. Cancer. 2015;121(6):893–899. [DOI] [PubMed] [Google Scholar]
- 83.Kearney N, McCann L, Norrie J, et al. Evaluation of a mobile phone-based, advanced symptom management system (ASyMS) in the management of chemotherapy-related toxicity. Support Care Cancer. 2009;17(4):437–444. [DOI] [PubMed] [Google Scholar]
- 84.Traeger L, McDonnell TM, McCarty CE, Greer JA, El-Jawahri A, Temel JS. Nursing intervention to enhance outpatient chemotherapy symptom management: Patient-reported outcomes of a randomized controlled trial. Cancer. 2015;121(21):3905–3913. [DOI] [PubMed] [Google Scholar]
- 85.Yount SE, Rothrock N, Bass M, et al. A randomized trial of weekly symptom telemonitoring in advanced lung cancer. J Pain Symptom Manage. 2014;47(6):973–989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Young JM, Butow PN, Walsh J, et al. Multicenter randomized trial of centralized nurse-led telephone-based care coordination to improve outcomes after surgical resection for colorectal cancer: the CONNECT intervention. J Clin Oncol. 2013;31(28):3585–3591. [DOI] [PubMed] [Google Scholar]
- 87.Falchook AD, Tracton G, Stravers L, et al. Use of mobile device technology to continuously collect patient-reported symptoms during radiation therapy for head and neck cancer: A prospective feasibility study. Adv Radiat Oncol. 2016;1(2):115–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Kornblith AB, Dowell JM, Herndon JE 2nd, et al. Telephone monitoring of distress in patients aged 65 years or older with advanced stage cancer: a cancer and leukemia group B study. Cancer. 2006;107(11):2706–2714. [DOI] [PubMed] [Google Scholar]
- 89.Cowan RA, Suidan RS, Andikyan V, et al. Electronic patient-reported outcomes from home in patients recovering from major gynecologic cancer surgery: A prospective study measuring symptoms and health-related quality of life. Gynecol Oncol. 2016;143(2):362–366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Judson TJ, Bennett AV, Rogak LJ, et al. Feasibility of long-term patient self-reporting of toxicities from home via the Internet during routine chemotherapy. J Clin Oncol. 2013;31(20):2580–2585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Andikyan V, Rezk Y, Einstein MH, et al. A prospective study of the feasibility and acceptability of a Web-based, electronic patient-reported outcome system in assessing patient recovery after major gynecologic cancer surgery. Gynecol Oncol. 2012;127(2):273–277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.McCann L, Maguire R, Miller M, Kearney N. Patients’ perceptions and experiences of using a mobile phone-based advanced symptom management system (ASyMS) to monitor and manage chemotherapy related toxicity. Eur J Cancer Care (Engl). 2009;18(2):156–164. [DOI] [PubMed] [Google Scholar]
- 93.Vickers AJ, Savage CJ, Shouery M, Eastham JA, Scardino PT, Basch EM. Validation study of a web-based assessment of functional recovery after radical prostatectomy. Health Qual Life Outcomes. 2010;8:82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Zylla DM, Gilmore GE, Steele GL, et al. Collection of electronic patient-reported symptoms in patients with advanced cancer using Epic MyChart surveys. Support Care Cancer. 2019. [DOI] [PubMed] [Google Scholar]
- 95.Wilson SR, Cram P. Another sobering result for home telehealth-and where we might go next. Arch Intern Med. 2012;172(10):779–780. [DOI] [PubMed] [Google Scholar]
- 96.Anatchkova M, Donelson SM, Skalicky AM, McHorney CA, Jagun D, Whiteley J. Exploring the implementation of patient-reported outcome measures in cancer care: need for more real-world evidence results in the peer reviewed literature. J Patient Rep Outcomes. 2018;2(1):64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Sim I Mobile Devices and Health. N Engl J Med. 2019;381(10):956–968. [DOI] [PubMed] [Google Scholar]
- 98.Hoogland A, Mansfield J, LaFranchise E, Bulls H, Johnstone P, H J. eHealth literacy in older adults with cancer. J Geriatr Oncol. 2020;In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Rathod KS, Wragg A. Do patient-reported outcome measures speak for all patient subgroups: is everyone included? Eur Heart J Qual Care Clin Outcomes. 2018;4(2):79–80. [DOI] [PubMed] [Google Scholar]
- 100.Nelson BW, Allen NB. Accuracy of Consumer Wearable Heart Rate Measurement During an Ecologically Valid 24-Hour Period: Intraindividual Validation Study. JMIR Mhealth Uhealth. 2019;7(3):e10828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Thomson EA, Nuss K, Comstock A, et al. Heart rate measures from the Apple Watch, Fitbit Charge HR 2, and electrocardiogram across different exercise intensities. J Sports Sci. 2019;37(12):1411–1419. [DOI] [PubMed] [Google Scholar]
- 102.Haberman ZC, Jahn RT, Bose R, et al. Wireless Smartphone ECG Enables Large-Scale Screening in Diverse Populations. J Cardiovasc Electrophysiol. 2015;26(5):520–526. [DOI] [PubMed] [Google Scholar]
- 103.Joo S, Lee K, Song C. A Comparative Study of Smartphone Game with Spirometry for Pulmonary Function Assessment in Stroke Patients. Biomed Res Int. 2018;2018:2439312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Alafeef M Smartphone-based photoplethysmographic imaging for heart rate monitoring. J Med Eng Technol. 2017;41(5):387–395. [DOI] [PubMed] [Google Scholar]
- 105.Ahmed A, Ahmad W, Khan MJ, Siddiqui SA, Cheema HM. A wearable sensor based multi-criteria-decision-system for real-time seizure detection. Conf Proc IEEE Eng Med Biol Soc. 2017;2017:2377–2380. [DOI] [PubMed] [Google Scholar]
- 106.Klesges RC, Klesges LM, Swenson AM, Pheley AM. A validation of two motion sensors in the prediction of child and adult physical activity levels. Am J Epidemiol. 1985;122(3):400–410. [DOI] [PubMed] [Google Scholar]
- 107.Breteler MJ, Janssen JH, Spiering W, Kalkman CJ, van Solinge WW, Dohmen DA. Measuring Free-Living Physical Activity With Three Commercially Available Activity Monitors for Telemonitoring Purposes: Validation Study. JMIR Form Res. 2019;3(2):e11489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Zhang P, Godin SD, Owens MV. Measuring the validity and reliability of the Apple Watch as a physical activity monitor. J Sports Med Phys Fitness. 2019;59(5):784–790. [DOI] [PubMed] [Google Scholar]
- 109.Bai Y, Hibbing P, Mantis C, Welk GJ. Comparative evaluation of heart rate-based monitors: Apple Watch vs Fitbit Charge HR. J Sports Sci. 2018;36(15):1734–1741. [DOI] [PubMed] [Google Scholar]
- 110.Wu W, Dasgupta S, Ramirez EE, Peterson C, Norman GJ. Classification accuracies of physical activities using smartphone motion sensors. J Med Internet Res. 2012;14(5):e130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Nolan M, Mitchell JR, Doyle-Baker PK. Validity of the Apple iPhone(R) /iPod Touch(R) as an accelerometer-based physical activity monitor: a proof-of-concept study. J Phys Act Health. 2014;11(4):759–769. [DOI] [PubMed] [Google Scholar]
- 112.Hardy J, Veinot TC, Yan X, et al. User acceptance of location-tracking technologies in health research: Implications for study design and data quality. J Biomed Inform. 2018;79:7–19. [DOI] [PubMed] [Google Scholar]
- 113.Irwin ML, McTiernan A, Manson JE, et al. Physical activity and survival in postmenopausal women with breast cancer: results from the women’s health initiative. Cancer Prev Res (Phila). 2011;4(4):522–529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Holick CN, Newcomb PA, Trentham-Dietz A, et al. Physical activity and survival after diagnosis of invasive breast cancer. Cancer Epidemiol Biomarkers Prev. 2008;17(2):379–386. [DOI] [PubMed] [Google Scholar]
- 115.Holmes MD, Chen WY, Feskanich D, Kroenke CH, Colditz GA. Physical activity and survival after breast cancer diagnosis. JAMA. 2005;293(20):2479–2486. [DOI] [PubMed] [Google Scholar]
- 116.Arem H, Pfeiffer RM, Engels EA, et al. Pre- and postdiagnosis physical activity, television viewing, and mortality among patients with colorectal cancer in the National Institutes of Health-AARP Diet and Health Study. J Clin Oncol. 2015;33(2):180–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Ratjen I, Schafmayer C, di Giuseppe R, et al. Postdiagnostic physical activity, sleep duration, and TV watching and all-cause mortality among long-term colorectal cancer survivors: a prospective cohort study. BMC Cancer. 2017;17(1):701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Ohri N, Halmos B, Bodner WR, et al. Daily Step Counts: A New Prognostic Factor in Locally Advanced Non-Small Cell Lung Cancer? Int J Radiat Oncol Biol Phys. 2019;105(4):745–751. [DOI] [PubMed] [Google Scholar]
- 119.Arem H, Pfeiffer RM, Moore SC, Brinton LA, Matthews CE. Body mass index, physical activity, and television time in relation to mortality risk among endometrial cancer survivors in the NIH-AARP Diet and Health Study cohort. Cancer Causes Control. 2016;27(11):1403–1409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Arem H, Park Y, Pelser C, et al. Prediagnosis body mass index, physical activity, and mortality in endometrial cancer patients. J Natl Cancer Inst. 2013;105(5):342–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Patel AV, Friedenreich CM, Moore SC, et al. American College of Sports Medicine Roundtable Report on Physical Activity, Sedentary Behavior, and Cancer Prevention and Control. Med Sci Sports Exerc. 2019;51(11):2391–2402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Ekelund U, Steene-Johannessen J, Brown WJ, et al. Does physical activity attenuate, or even eliminate, the detrimental association of sitting time with mortality? A harmonised meta-analysis of data from more than 1 million men and women. Lancet. 2016;388(10051):1302–1310. [DOI] [PubMed] [Google Scholar]
- 123.Roshanaei-Moghaddam B, Katon WJ, Russo J. The longitudinal effects of depression on physical activity. Gen Hosp Psychiatry. 2009;31(4):306–315. [DOI] [PubMed] [Google Scholar]
- 124.Low CA, Stanton AL. Activity disruption and depressive symptoms in women living with metastatic breast cancer. Health Psychol. 2015;34(1):89–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Canzian M,M,. Trajectories of depression: unobtrusive monitoring of depressive states by means of smartphone mobility traces analysis. ACM International Joint Conference on Pervasive and Ubiquitous Computing (UbiComp ‘15); 2015. [Google Scholar]
- 126.Canzian L, Musolesi M. Trajectories of depression: unobtrusive monitoring of depressive states by means of smartphone mobility traces analysis. Paper presented at: Association for Computing Machinery International Joint Conference on Pervasive and Ubiquitous Computing2015; Osaka. [Google Scholar]
- 127.Sano A, Taylor S, McHill AW, et al. Identifying Objective Physiological Markers and Modifiable Behaviors for Self-Reported Stress and Mental Health Status Using Wearable Sensors and Mobile Phones: Observational Study. J Med Internet Res. 2018;20(6):e210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Low CA, Dey AK, Ferreira D, et al. Estimation of Symptom Severity During Chemotherapy From Passively Sensed Data: Exploratory Study. J Med Internet Res. 2017;19(12):e420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Wind DK, Sapiezynski P, Furman MA, Lehmann S. Inferring Stop-Locations from WiFi. PLoS One. 2016;11(2):e0149105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Geyer K, Ellis DA, Piwek L. A simple location-tracking app for psychological research. Behav Res Methods. 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Zhou X, Li D. Quantifying multi-dimensional attributes of human activities at various geographic scales based on smartphone tracking. Int J Health Geogr. 2018;17(1):11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Ruktanonchai NW, Ruktanonchai CW, Floyd JR, Tatem AJ. Using Google Location History data to quantify fine-scale human mobility. Int J Health Geogr. 2018;17(1):28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Saeb S, Zhang M, Karr CJ, et al. Mobile Phone Sensor Correlates of Depressive Symptom Severity in Daily-Life Behavior: An Exploratory Study. J Med Internet Res. 2015;17(7):e175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Saeb S, Lattie EG, Kording KP, Mohr DC. Mobile Phone Detection of Semantic Location and Its Relationship to Depression and Anxiety. JMIR Mhealth Uhealth. 2017;5(8):e112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Tsuruda JS, Bradley WG. MR detection of intracranial calcification: a phantom study. AJNR Am J Neuroradiol. 1987;8(6):1049–1055. [PMC free article] [PubMed] [Google Scholar]
- 136.Cote DJ, Barnett I, Onnela JP, Smith TR. Digital Phenotyping in Patients with Spine Disease: A Novel Approach to Quantifying Mobility and Quality of Life. World Neurosurg. 2019;126:e241–e249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Granger CL, Denehy L, McDonald CF, Irving L, Clark RA. Physical activity measured using global positioning system tracking in non-small cell lung cancer: an observational study. Integr Cancer Ther. 2014;13(6):482–492. [DOI] [PubMed] [Google Scholar]
- 138.Wevers LE, Kwakkel G, van de Port IG. Is outdoor use of the six-minute walk test with a global positioning system in stroke patients’ own neighbourhoods reproducible and valid? J Rehabil Med. 2011;43(11):1027–1031. [DOI] [PubMed] [Google Scholar]
- 139.Shim JS OK, Kim HC. Dietary assessment methods in epidemiologic studies. Epidemiology and Health. 2014:e2014009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Carter MC, Albar SA, Morris MA, et al. Development of a UK Online 24-h Dietary Assessment Tool: myfood24. Nutrients. 2015;7(6):4016–4032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Liu B, Young H, Crowe FL, et al. Development and evaluation of the Oxford WebQ, a low-cost, web-based method for assessment of previous 24 h dietary intakes in large-scale prospective studies. Public Health Nutr. 2011;14(11):1998–2005. [DOI] [PubMed] [Google Scholar]
- 142.Kristal AR, Kolar AS, Fisher JL, et al. Evaluation of web-based, self-administered, graphical food frequency questionnaire. J Acad Nutr Diet. 2014;114(4):613–621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Gemming L, Utter J, Ni Mhurchu C. Image-assisted dietary assessment: a systematic review of the evidence. J Acad Nutr Diet. 2015;115(1):64–77. [DOI] [PubMed] [Google Scholar]
- 144.Beasley JM, Davis A, Riley WT. Evaluation of a web-based, pictorial diet history questionnaire. Public Health Nutr. 2009;12(5):651–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.BVR ES, Rad MG, Cui J, McCabe M, Pan K. A Mobile-Based Diet Monitoring System for Obesity Management. J Health Med Inform. 2018;9(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Lo FP, Sun Y, Qiu J, Lo B. Food Volume Estimation Based on Deep Learning View Synthesis from a Single Depth Map. Nutrients. 2018;10(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Beasley JM, Newcomb PA, Trentham-Dietz A, et al. Post-diagnosis dietary factors and survival after invasive breast cancer. Breast Cancer Res Treat. 2011;128(1):229–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.McEligot AJ, Largent J, Ziogas A, Peel D, Anton-Culver H. Dietary fat, fiber, vegetable, and micronutrients are associated with overall survival in postmenopausal women diagnosed with breast cancer. Nutr Cancer. 2006;55(2):132–140. [DOI] [PubMed] [Google Scholar]
- 149.Van Blarigan EL, Fuchs CS, Niedzwiecki D, et al. Association of Survival With Adherence to the American Cancer Society Nutrition and Physical Activity Guidelines for Cancer Survivors After Colon Cancer Diagnosis: The CALGB 89803/Alliance Trial. JAMA Oncol. 2018;4(6):783–790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Guinter MA, McCullough ML, Gapstur SM, Campbell PT. Associations of Pre- and Postdiagnosis Diet Quality With Risk of Mortality Among Men and Women With Colorectal Cancer. J Clin Oncol. 2018:JCO1800714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Jochems SHJ, Van Osch FHM, Bryan RT, et al. Impact of dietary patterns and the main food groups on mortality and recurrence in cancer survivors: a systematic review of current epidemiological literature. BMJ Open. 2018;8(2):e014530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Karavasiloglou N, Pestoni G, Wanner M, Faeh D, Rohrmann S. Healthy lifestyle is inversely associated with mortality in cancer survivors: Results from the Third National Health and Nutrition Examination Survey (NHANES III). PLoS One. 2019;14(6):e0218048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Zinkhan M, Kantelhardt JW. Sleep Assessment in Large Cohort Studies with High-Resolution Accelerometers. Sleep medicine clinics. 2016;11(4):469–488. [DOI] [PubMed] [Google Scholar]
- 154.Lee XK, Chee NI, Ong JL, et al. Validation of a Consumer Sleep Wearable Device With Actigraphy and Polysomnography in Adolescents Across Sleep Opportunity Manipulations. Journal of Clinical Sleep Medicine. 2019;15(09):1337–1346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Haghayegh S, Khoshnevis S, Smolensky MH, Diller KR, Castriotta RJ. Performance assessment of new-generation Fitbit technology in deriving sleep parameters and stages. Chronobiology international. 2019:1–13. [DOI] [PubMed] [Google Scholar]
- 156.Rieck TM, Gaz DV, Peterson NW, et al. Comparison of Commercially-Available Sleep Tracking Devices With Sleep Diary and Actigraphy. Sleep. 2019;42(Supplement_1):A404–A405. [Google Scholar]
- 157.Chinoy ED, Huwa KE, Snider MN, et al. Examination of Wearable and Non-Wearable Consumer Sleep-Tracking Devices Versus Polysomnography. Sleep. 2019;42(Supplement_1):A403–A404. [Google Scholar]
- 158.Tal A, Shinar Z, Shaki D, Codish S, Goldbart A. Validation of Contact-Free Sleep Monitoring Device with Comparison to Polysomnography. J Clin Sleep Med. 2017;13(3):517–522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Ciman M, Wac K. Smartphones as Sleep Duration Sensors: Validation of the iSenseSleep Algorithm. JMIR Mhealth Uhealth. 2019;7(5):e11930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Narayan S, Shivdare P, Niranjan T, Williams K, Freudman J, Sehra R. Noncontact identification of sleep-disturbed breathing from smartphone-recorded sounds validated by polysomnography. Sleep Breath. 2019;23(1):269–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Kim T, Kim JW, Lee K. Detection of sleep disordered breathing severity using acoustic biomarker and machine learning techniques. Biomed Eng Online. 2018;17(1):16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Nakano H, Hirayama K, Sadamitsu Y, et al. Monitoring sound to quantify snoring and sleep apnea severity using a smartphone: proof of concept. J Clin Sleep Med. 2014;10(1):73–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Shin H, Cho J. Unconstrained snoring detection using a smartphone during ordinary sleep. Biomed Eng Online. 2014;13:116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Hershman SG, Bot BM, Shcherbina A, et al. Physical activity, sleep and cardiovascular health data for 50,000 individuals from the MyHeart Counts Study. Sci Data. 2019;6(1):24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Kampshoff CS, Verdonck-de Leeuw IM, van Oijen MG, Sprangers MA, Buffart LM. Ecological momentary assessments among patients with cancer: A scoping review. Eur J Cancer Care (Engl). 2019;28(3):e13095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Small BJ, Jim HSL, Eisel SL, Jacobsen PB, Scott SB. Cognitive performance of breast cancer survivors in daily life: Role of fatigue and depressed mood. Psychooncology. 2019;28(11):2174–2180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Diehl M, Hooker K, Sliwinski MJ. Handbook of intraindividual variability across the life span. New York, NY: Routledge; 2015. [Google Scholar]
- 168.MacDonald SWS, Nyberg L, Backman L. Intra-individual variability in behavior: links to brain structure, neurotransmission and neuronal activity. Trends Neurosci. 2006;29(8):474–480. [DOI] [PubMed] [Google Scholar]
- 169.Kummerow Broman K, Gaskill CE, Faqih A, et al. Evaluation of Wound Photography for Remote Postoperative Assessment of Surgical Site Infections. JAMA Surg. 2019;154(2):117–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Tolins ML, Hippe DS, Morse SC, Evans HL, Lober WB, Vrablik MC. Wound Care Follow-Up From the Emergency Department Using a Mobile Application: A Pilot Study. J Emerg Med. 2019;57(5):629–636. [DOI] [PubMed] [Google Scholar]
- 171.Gunter RL, Fernandes-Taylor S, Rahman S, et al. Feasibility of an Image-Based Mobile Health Protocol for Postoperative Wound Monitoring. J Am Coll Surg. 2018;226(3):277–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Gunter R, Fernandes-Taylor S, Mahnke A, et al. Evaluating Patient Usability of an Image-Based Mobile Health Platform for Postoperative Wound Monitoring. JMIR Mhealth Uhealth. 2016;4(3):e113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Muller AM, Blandford A, Yardley L. The conceptualization of a Just-In-Time Adaptive Intervention (JITAI) for the reduction of sedentary behavior in older adults. Mhealth. 2017;3:37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Burns MN, Begale M, Duffecy J, et al. Harnessing context sensing to develop a mobile intervention for depression. J Med Internet Res. 2011;13(3):e55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Manor B, Yu W, Zhu H, et al. Smartphone App-Based Assessment of Gait During Normal and Dual-Task Walking: Demonstration of Validity and Reliability. JMIR Mhealth Uhealth. 2018;6(1):e36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Pepa L, Verdini F, Spalazzi L. Gait parameter and event estimation using smartphones. Gait Posture. 2017;57:217–223. [DOI] [PubMed] [Google Scholar]
- 177.Silsupadol P, Teja K, Lugade V. Reliability and validity of a smartphone-based assessment of gait parameters across walking speed and smartphone locations: Body, bag, belt, hand, and pocket. Gait Posture. 2017;58:516–522. [DOI] [PubMed] [Google Scholar]
- 178.Capela NA, Lemaire ED, Baddour NC. A smartphone approach for the 2 and 6-minute walk test. Conf Proc IEEE Eng Med Biol Soc. 2014;2014:958–961. [DOI] [PubMed] [Google Scholar]
- 179.Capela NA, Lemaire ED, Baddour N. Novel algorithm for a smartphone-based 6-minute walk test application: algorithm, application development, and evaluation. J Neuroeng Rehabil. 2015;12:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Brooks GC, Vittinghoff E, Iyer S, et al. Accuracy and Usability of a Self-Administered 6-Minute Walk Test Smartphone Application. Circ Heart Fail. 2015;8(5):905–913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Galan-Mercant A, Baron-Lopez FJ, Labajos-Manzanares MT, Cuesta-Vargas AI. Reliability and criterion-related validity with a smartphone used in timed-up-and-go test. Biomed Eng Online. 2014;13:156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Mellone S, Tacconi C, Chiari L. Validity of a Smartphone-based instrumented Timed Up and Go. Gait Posture. 2012;36(1):163–165. [DOI] [PubMed] [Google Scholar]
- 183.Santos C, Pauchard N, Guilloteau A. Reliability assessment of measuring active wrist pronation and supination range of motion with a smartphone. Hand Surg Rehabil. 2017;36(5):338–345. [DOI] [PubMed] [Google Scholar]
- 184.Cox RW, Martinez RE, Baker RT, Warren L. Validity of a Smartphone Application for Measuring Ankle Plantar Flexion. J Sport Rehabil. 2018;27(3). [DOI] [PubMed] [Google Scholar]
- 185.Ning Y, Hu S, Nie X, Liang S, Li H, Zhao G. Real-time Action Recognition and Fall Detection Based on Smartphone. Conf Proc IEEE Eng Med Biol Soc. 2018;2018:4418–4422. [DOI] [PubMed] [Google Scholar]
- 186.De Cillisy F, De Simioy F, Guidoy F, Incalzi RA, Setolay R. Fall-detection solution for mobile platforms using accelerometer and gyroscope data. Conf Proc IEEE Eng Med Biol Soc. 2015;2015:3727–3730. [DOI] [PubMed] [Google Scholar]
- 187.Vermeulen J, Willard S, Aguiar B, De Witte LP. Validity of a Smartphone-Based Fall Detection Application on Different Phones Worn on a Belt or in a Trouser Pocket. Assist Technol. 2015;27(1):18–23. [DOI] [PubMed] [Google Scholar]
- 188.Casilari E, Oviedo-Jimenez MA. Automatic Fall Detection System Based on the Combined Use of a Smartphone and a Smartwatch. PLoS One. 2015;10(11):e0140929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Google. Big Data. https://www.google.com/search?client=safari&rls=en&q=big+data+definition&ie=UTF-8&oe=UTF-8. Published 2020. Accessed Jan. 2, 2020.
- 190.Department of Health and Human Services. Summary of the HIPAA Privacy Rule. https://www.hhs.gov/hipaa/for-professionals/privacy/laws-regulations/index.html. Published 2013. Accessed November 27, 2019.
- 191.Department of Health and Human Services. HITECH Act Enforcement Interim Final Rule. https://www.hhs.gov/hipaa/for-professionals/special-topics/hitech-act-enforcement-interim-final-rule/index.html. Published 2017. Accessed November 27, 2019.
- 192.Institute of Medicine. The Learning Healthcare System: Workshop Summary (2007). Washington D.C.: National Academies Press; 2007. [PubMed] [Google Scholar]
- 193.Abernethy AP, Etheredge LM, Ganz PA, et al. Rapid-learning system for cancer care. J Clin Oncol. 2010;28(27):4268–4274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Sankar PL, Parker LS. The Precision Medicine Initiative’s All of Us Research Program: an agenda for research on its ethical, legal, and social issues. Genet Med. 2017;19(7):743–750. [DOI] [PubMed] [Google Scholar]
- 195.Moraska AR, Sood A, Dakhil SR, et al. Phase III, randomized, double-blind, placebo-controlled study of long-acting methylphenidate for cancer-related fatigue: North Central Cancer Treatment Group NCCTG-N05C7 trial. J Clin Oncol. 2010;28(23):3673–3679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Schilsky RL, Michels DL, Kearbey AH, Yu PP, Hudis CA. Building a rapid learning health care system for oncology: the regulatory framework of CancerLinQ. J Clin Oncol. 2014;32(22):2373–2379. [DOI] [PubMed] [Google Scholar]
- 197.American Society of Clinical Oncology. ASCO CancerLinQ. https://www.cancerlinq.org/index.php/solutions/oncology-practices. Published 2019. Accessed November 17, 2019.
- 198.The GO2 Foundation for Lung Cancer. Lung cancer registry. https://www.lungcancerregistry.org. Published 2019. Accessed Jan. 2, 2020.
- 199.Smith TG, Dunn ME, Levin KY, et al. Cancer survivor perspectives on sharing patient-generated health data with central cancer registries. Qual Life Res. 2019;28(11):2957–2967. [DOI] [PubMed] [Google Scholar]
- 200.Oncology Research Information Exchange Network. ORIEN. http://www.oriencancer.org. Published 2019. Accessed November 1, 2019.
- 201.McCarthy J, Minsky M, Rochester N, Shannon CE. A Proposal for the Dartmouth Summer Research Project on Artificial Intelligence. http://raysolomonoff.com/dartmouth/boxa/dart564props.pdf. Published 1995. Accessed November 1, 2019. [Google Scholar]
- 202.Meehl PE. Clinical versus statistical prediction: a theoretical analysis and a review of evidence. Echo Pint Books & Media; 2013. [Google Scholar]
- 203.Bornstein BH, Emler AC. Rationality in medical decision making: a review of the literature on doctors’ decision-making biases. J Eval Clin Pract. 2001;7(2):97–107. [DOI] [PubMed] [Google Scholar]
- 204.Saposnik G, Redelmeier D, Ruff CC, Tobler PN. Cognitive biases associated with medical decisions: a systematic review. BMC Med Inform Decis Mak. 2016;16(1):138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Dawes RM, Faust D, Meehl PE. Clinical versus actuarial judgment. Science. 1989;243(4899):1668–1674. [DOI] [PubMed] [Google Scholar]
- 206.Schwartz WB, Patil RS, Szolovits P. Artificial intelligence in medicine. Where do we stand? N Engl J Med. 1987;316(11):685–688. [DOI] [PubMed] [Google Scholar]
- 207.Goodfellow I, Bengio Y, Courville A. Deep Learning. Cambridge MA: MIT Press; 2016. [Google Scholar]
- 208.Krizhevsky A, Sutskever I, Hinton GE. ImageNet classification with deep convolutional neural networks. Commun ACM. 2017;60(6):84–90. [Google Scholar]
- 209.Ronneberger O, Fischer P, Brox T. U-Net: Convolutional Networks for Biomedical Image Segmentation. 2015; Cham. [Google Scholar]
- 210.Esteva A, Kuprel B, Novoa RA, et al. Dermatologist-level classification of skin cancer with deep neural networks. Nature. 2017;542(7639):115–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.White RW, Horvitz E. Evaluation of the Feasibility of Screening Patients for Early Signs of Lung Carcinoma in Web Search Logs. JAMA Oncol. 2017;3(3):398–401. [DOI] [PubMed] [Google Scholar]
- 212.Paparrizos J, White RW, Horvitz E. Screening for Pancreatic Adenocarcinoma Using Signals From Web Search Logs: Feasibility Study and Results. J Oncol Pract. 2016. [DOI] [PubMed] [Google Scholar]
- 213.Brichetto G, Monti Bragadin M, Fiorini S, et al. The hidden information in patient-reported outcomes and clinician-assessed outcomes: multiple sclerosis as a proof of concept of a machine learning approach. Neurol Sci. 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Fiorini S, Verri A, Tacchino A, Ponzio M, Brichetto G, Barla A. A machine learning pipeline for multiple sclerosis course detection from clinical scales and patient reported outcomes. Conf Proc IEEE Eng Med Biol Soc. 2015;2015:4443–4446. [DOI] [PubMed] [Google Scholar]
- 215.Huber M, Kurz C, Leidl R. Predicting patient-reported outcomes following hip and knee replacement surgery using supervised machine learning. BMC Med Inform Decis Mak. 2019;19(1):3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Devlin J, Chang M, Lee K, Toutanova K. BERT: Pre-training of Deep Bidirectional Transformers for Language Understanding. Paper presented at: Proceedings of the 2019 Conference of the North American Chapter of the Association for Computational Linguistics: Human Language Technologies, Volume 1 (Long and Short Papers)\; jun\, 2019; Minneapolis, Minnesota\. [Google Scholar]
- 217.Lee J, Yoon W, Kim S, et al. BioBERT: a pre-trained biomedical language representation model for biomedical text mining. Bioinformatics. 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Moradi M, Dorffner G, Samwald M. Deep contextualized embeddings for quantifying the informative content in biomedical text summarization. Comput Methods Programs Biomed. 2019;184:105117. [DOI] [PubMed] [Google Scholar]
- 219.Li F, Jin Y, Liu W, Rawat BPS, Cai P, Yu H. Fine-Tuning Bidirectional Encoder Representations From Transformers (BERT)-Based Models on Large-Scale Electronic Health Record Notes: An Empirical Study. JMIR Med Inform. 2019;7(3):e14830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Zhang X, Zhang Y, Zhang Q, et al. Extracting comprehensive clinical information for breast cancer using deep learning methods. Int J Med Inform. 2019;132:103985. [DOI] [PubMed] [Google Scholar]
- 221.Topol EJ. High-performance medicine: the convergence of human and artificial intelligence. Nat Med. 2019;25(1):44–56. [DOI] [PubMed] [Google Scholar]
- 222.U.S. Department of Health Human Services. FDA Approved AI Algorithms. https://www.acrdsi.org/DSI-Services/FDA-Approved-AI-Algorithms. Published 2019. Accessed November 22, 2019.
- 223.Medicine). IIo. Delivering high-quality cancer care: Charting a new course for a system in crisis. Washington, DC: The National Academies Press.; 2013. [PubMed] [Google Scholar]
- 224.Rotenstein LS, Agarwal A, O’Neil K, et al. Implementing patient-reported outcome surveys as part of routine care: lessons from an academic radiation oncology department. J Am Med Inform Assoc. 2017;24(5):964–968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Totterdale RL. Case study: The utlization of low-code development technology to support research data collection. Issues in Information Systems. 2018;19(2):132–139. [Google Scholar]
- 226.Yanez B, Bouchard LC, Cella D, et al. Patient-centered engagement and symptom/toxicity monitoring in the new era of tumor next-generation sequencing and immunotherapy: The OncoTool and OncoPRO platforms. Cancer. 2019;125(14):2338–2344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Snyder C, Smith K, Holzner B, et al. Making a picture worth a thousand numbers: recommendations for graphically displaying patient-reported outcomes data. Qual Life Res. 2019;28(2):345–356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Gensheimer SG, Wu AW, Snyder CF, Group P-EUGS, Group P-EUGW. Oh, the Places We’ll Go: Patient-Reported Outcomes and Electronic Health Records. Patient. 2018;11(6):591–598. [DOI] [PubMed] [Google Scholar]
- 229.Bell ML, Fiero MH, Dhillon HM, Bray VJ, Vardy JL. Statistical controversies in cancer research: using standardized effect size graphs to enhance interpretability of cancer-related clinical trials with patient-reported outcomes. Ann Oncol. 2017;28(8):1730–1733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Kallen MA, Yang D, Haas N. A technical solution to improving palliative and hospice care. Support Care Cancer. 2012;20(1):167–174. [DOI] [PubMed] [Google Scholar]
- 231.Snyder CF, Blackford AL, Wolff AC, et al. Feasibility and value of PatientViewpoint: a web system for patient-reported outcomes assessment in clinical practice. Psychooncology. 2013;22(4):895–901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Rotenstein LS, Huckman RS, Wagle NW. Making Patients and Doctors Happier - The Potential of Patient-Reported Outcomes. N Engl J Med. 2017;377(14):1309–1312. [DOI] [PubMed] [Google Scholar]
- 233.Howell D, Molloy S, Wilkinson K, et al. Patient-reported outcomes in routine cancer clinical practice: a scoping review of use, impact on health outcomes, and implementation factors. Ann Oncol. 2015;26(9):1846–1858. [DOI] [PubMed] [Google Scholar]
- 234.Luckett T, Butow PN, King MT. Improving patient outcomes through the routine use of patient-reported data in cancer clinics: future directions. Psychooncology. 2009;18(11):1129–1138. [DOI] [PubMed] [Google Scholar]
- 235.Snyder CF, Jensen RE, Geller G, Carducci MA, Wu AW. Relevant content for a patient-reported outcomes questionnaire for use in oncology clinical practice: Putting doctors and patients on the same page. Qual Life Res. 2010;19(7):1045–1055. [DOI] [PubMed] [Google Scholar]
- 236.Hsiao CJ, Dymek C, Kim B, Russell B. Advancing the use of patient-reported outcomes in practice: understanding challenges, opportunities, and the potential of health information technology. Qual Life Res. 2019;28(6):1575–1583. [DOI] [PubMed] [Google Scholar]
- 237.Warsame R, D’Souza A. Patient Reported Outcomes Have Arrived: A Practical Overview for Clinicians in Using Patient Reported Outcomes in Oncology. Mayo Clin Proc. 2019;94(11):2291–2301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Bowling A Mode of questionnaire administration can have serious effects on data quality. J Public Health (Oxf). 2005;27(3):281–291. [DOI] [PubMed] [Google Scholar]
- 239.Brant JM, Hirschman KB, Keckler SL, Dudley WN, Stricker C. Patient and Provider Use of Electronic Care Plans Generated From Patient-Reported Outcomes. Oncol Nurs Forum. 2019;46(6):715–726. [DOI] [PubMed] [Google Scholar]
- 240.Barbera L, Moody L. A Decade in Review: Cancer Care Ontario’s Approach to Symptom Assessment and Management %U https://journals.lww.com/lww-medicalcare/Fulltext/2019/05001/A_Decade_in_Review__Cancer_Care_Ontario_s_Approach.14.aspx. Medical Care. 2019;57:S80–S84. [DOI] [PubMed] [Google Scholar]
- 241.Blackford AL, Wu AW, Snyder C. Interpreting and Acting on PRO Results in Clinical Practice: Lessons Learned From the PatientViewpoint System and Beyond %U https://journals.lww.com/lww-medicalcare/Fulltext/2019/05001/Interpreting_and_Acting_on_PRO_Results_in_Clinical.9.aspx. Medical Care. 2019;57:S46–S51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Stover AM, Tompkins Stricker C, Hammelef K, et al. Using Stakeholder Engagement to Overcome Barriers to Implementing Patient-reported Outcomes (PROs) in Cancer Care Delivery: Approaches From 3 Prospective Studies. Med Care. 2019;57 Suppl 5 Suppl 1:S92–S99. [DOI] [PubMed] [Google Scholar]
- 243.Wood WA, Bennett AV, Basch E. Emerging uses of patient generated health data in clinical research. Mol Oncol. 2015;9(5):1018–1024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Bennett AV, Jensen RE, Basch E. Electronic patient-reported outcome systems in oncology clinical practice. CA Cancer J Clin. 2012;62(5):337–347. [DOI] [PubMed] [Google Scholar]