Skip to main content
Springer logoLink to Springer
. 2024 Nov 16;30(1):1–16. doi: 10.1007/s10147-024-02651-8

Electronic patient-reported outcomes as digital therapeutics for patients with cancer: a narrative review of current practices and future directions

Ken Yamaguchi 1,, Nozomi Higashiyama 1, Maki Umemiya 1, Yoshihide Inayama 1, Ayami Koike 1, Akihiko Ueda 1, Rin Mizuno 1, Mana Taki 1, Koji Yamanoi 1, Ryusuke Murakami 1, Junzo Hamanishi 1, Masaki Mandai 1
PMCID: PMC11700045  PMID: 39549219

Abstract

Improved cancer treatment outcomes have increased the demand for medical care that considers the quality of life of patients with cancer. Patient-reported outcomes (PROs) help assess the quality of life because they involve direct evaluation of the patients. Recently, electronic PROs (ePROs) have been used in clinical cancer care settings in Europe and the United States. Electronic PROs positively affected communication between patients with cancer and healthcare providers, enhanced education, optimized self-management, contributed to healthcare economics, assisted in monitoring adverse events, and improved prognosis. However, challenges such as adherence, burden on healthcare providers, lack of personalized formats, low digital literacy, and implementation costs remain. Therefore, carefully selecting the items to be recorded by ePROs in alignment with specific objectives is essential. Additionally, developing systems using lifelogs—digital records of daily activities—and creating mechanisms that automatically encourage patient behavioral changes based on the reported data are crucial. This review delineates the advantages and challenges of ePROs according to their history and proposes the prospects of ePRO.

Keywords: Digital therapeutics, Electronic patient-reported outcomes, Digital health, Cancer, Quality of life

Introduction

With advancements in cancer treatment, improving the prognosis and maintaining and enhancing the quality of life (QOL) of patients with cancer have become essential objectives in cancer care [1]. Patient-centered care, patient-driven support, palliative care, and tumor treatment must be comprehensive throughout the diagnosis, treatment, and post-treatment phases to maintain and improve the QOL of patients with cancer [25]. Patient perceptions of adverse events associated with cancer treatment change between therapeutic phases [6]. The physicians assess symptoms using the Common Terminology Criteria for Adverse Events (CTCAE); however, symptoms reported by healthcare providers are reported less frequently and later than those reported directly by patients [710]. Therefore, direct symptom evaluation from patients, known as patient-reported outcomes (PROs), is crucial to accurately capture adverse events and QOL. PROs are defined as self-assessment reports of patients and their disease and treatment without external interpretation [11]. Adverse events and QOL deterioration occur even at home, where medical providers are absent. Although physician assessments using the CTCAE correlate with events such as mortality and hospitalization, PROs reflect the daily health status of patients more accurately. Therefore, the complementary use of both methods can provide clinically important information [12].

Recent advancements in digital communication technology have improved accessibility and the accurate capture of patient needs, thereby reducing healthcare disparities by extending medical services beyond clinical settings [13]. Digital health technologies can be used for cancer screening, patient education, shared decision-making, communication, symptom monitoring, and promoting a healthy lifestyle [13]. Therefore, electronic PROs (ePROs)—components of digital health—play a crucial role in enhancing patient outcomes, streamlining healthcare delivery, and supporting patient-centered care. However, challenges such as maintaining adherence, the strain on healthcare providers, the lack of personalized formats, low levels of digital literacy, and the costs associated with implementation still exist. Therefore, this article reviews evidence on ePROs to propose future perspectives.

History of ePRO clinical studies in oncology

PROs are used in clinical trials for new therapeutic drugs because adverse events and QOL should be evaluated in addition to treatment outcomes in cancer treatment. Between 2007 and 2013, a total of 27% of clinical trials were registered at ClinicalTrials.gov [14]. Review articles on PROs, including ePROs, published between 2013 and 2015 reported improvements in symptom management, supportive care, patient communication, and satisfaction; however, none have reported improvements in QOL [1517]. Since 2014, the QOL of patients with cancer has reportedly improved owing to ePROs [1820]. Therefore, attempts have been made to incorporate ePROs, more easily accessible than PROs, into medical care. A literature review of intervention studies assessing digital health from 1999 to 2021 revealed a rapid increase in articles on ePROs after 2013 [21]. An assessment of ClinicalTrials.gov with “Cancer” in the condition or disease field, “QOL” in other terms, and “digital intervention” in intervention or treatment shows that only one clinical trial on the use of ePROs began in 2012, followed by three to four trials per year until 2016, five to eight trials per year from 2017 to 2020, and over 20 trials per year from 2021 (Fig. 1). In a randomized trial involving 100 postoperative patients with lung cancer, symptom monitoring was conducted twice weekly via automated phone calls over 4 weeks. The system automatically alerted healthcare providers when severe symptoms were detected, facilitating timely intervention and symptom control [22]. This system is possible because ePROs are not paper-based. The first report of improvements in overall survival (OS) because of ePROs was published in 2017 [23, 24]. Furthermore, QOL assessments using ePROs have been performed in cutting-edge clinical genomic research [25]. The increased use of PROs in evaluating artificial intelligence (AI) health technologies also demonstrates the importance of patient-centered care, even in the most technologically advanced healthcare systems [26]. This increased use of PROs may be attributed to a greater awareness of their benefits and guidance from regulatory authorities [26].

Fig. 1.

Fig. 1

The number of clinical trials describing digital interventions regarding quality of life and cancer in ClinicalTrials.gov. The number of clinical trials describing digital interventions for the quality of life and cancer has increased since 2021

However, the various methods by which PRO data are analyzed, presented, and interpreted can lead to potential errors and inconsistencies that could adversely affect patient care and outcomes. Therefore, the Setting International Standards in Analyzing Patient-Reported Outcomes and Quality of Life Endpoints in Cancer Clinical Trials-Innovative Medicines Initiative (SISAQOL-IMI) consortium has established recommendations for designing, analyzing, presenting, and interpreting PRO data in cancer clinical trials based on previous SISAQOL activities [27].

Advantages of ePRO in oncology

Communication between patients and healthcare providers

ePRO systems enhance communication between patients and healthcare providers, enabling effective symptom management and timely interventions [28, 29]. A randomized controlled trial (RCT) demonstrated that using ePROs resulted in a 20% increase in patient-provider communication regarding symptoms and side effects [28, 29]. The impact of using the Electronic Self-Report Assessment–Cancer (ESRA-C) in outpatient settings to facilitate discussions between clinicians and patients with cancer regarding symptoms and QOL issues was investigated [30]. The discussions during outpatient visits were more frequent when symptoms and QOL issues were initially reported and clinicians indicated that summaries provided by the ESRA-C were valuable [30]. In the Patient-Reported Outcomes To Enhance Cancer Treatment (PRO-TECT) trial, patients, clinicians, and nurses experienced improved discussions [31].

Educating patients and caregivers

ePRO systems provide valuable educational resources, aiding patients and caregivers in understanding and managing their condition, potentially leading to improved health outcomes [32]. ePROs are associated with increased patient empowerment and self-management capabilities, as patients using ePROs require more disease-specific content [32]. A total of 97% of the clinicians noted that ePROs using the PRO-TECT system helped provide the patients with information [31].

Patient condition monitoring, self-management, and healthcare costs

ePRO systems facilitate continuous monitoring and support for patient self-management, allowing the early detection of issues and timely interventions. The benefits of ePRO measurement include greater patient preference and acceptability, similar or faster completion times, higher data quality and response rates, and facilitated symptom management [33]. Patient self-management as a component of self-management interventions has been demonstrated to contribute to enhanced self-efficacy. Moreover, increased patient self-efficacy has been associated with enhancements in health-related QOL [34].

eRAPID, which was developed in the United Kingdom, is an online eHealth system for patients with cancer to self-manage adverse events during and after treatment [35]. A phase-III trial evaluated the impact of eRAPID on symptom control, healthcare utilization, patient self-efficacy, and QOL in patients receiving chemotherapy for colorectal, breast, or gynecological cancers. Patients using eRAPID reported higher self-efficacy and better health status scores on the EQ5D-VAS at 18 weeks than patients who did not use eRAPID. Patient adherence correlated with clinical data usage and was associated with improved QOL at 12 weeks [36].

Furthermore, ePRO implementation can reduce costs by decreasing emergency visits and hospital admissions through improved symptom management [4, 37, 38].

Managing adverse events and QOL

The regular use of ePRO systems is associated with better management of adverse events and increased patient QOL during treatment [39, 40]. The ESRA-C intervention group demonstrated significantly improved symptom relief compared to the control group [18]. Another randomized study compared a control group that solely used a fully automated ESRA-C with an intervention group that received results delivered to their homes or hospitals. The intervention group showed an estimated 1.53-point reduction in Symptom Distress Scale-15 (SDS-15) scores compared to the control group [41]. A multicenter RCT using Electronic Monitoring of Symptoms and Syndromes Associated with Cancer (E-MOSAIC) showed that overall QOL—which was assessed by using items from the EORTC-QLQ-C30—showed favorable trends in the intervention group, with significant improvement in symptom management. Cancérologie Parcours Région Ile de France-Remote Patient Monitoring Systems (CAPRI-PRMS) is a nurse navigator-led program developed in France [42]. A phase-III trial randomly assigned patients receiving approved oral anticancer agents or molecular targeted therapies to either an intervention group, which received nurse navigator-led follow-up through a combination of a web portal and smartphone application in addition to standard care for 6 months, or a control group, which underwent regular symptom monitoring by treatment providers. The intervention did not significantly alter the EORTC-QLQ-C30 Global Health Score. However, it significantly improved the Patient Assessment of Chronic Illness Care score (2.94 vs. 2.67), reduced hospitalization days (2.82 vs. 4.44 days) and decreased treatment-related Grade 3 or higher toxicities (27.6 vs. 36.9%) [43]. eRAPID showed that the intervention group had improved FACT-PHYSICAL WELLBEING at 6 and 12 weeks compared with the control group. Symptom Tracking and Reporting (STAR) e-mail nurses and patients alerts for severe symptoms or deterioration. Compared to the baseline, the intervention group showed a significantly improved QOL compared to the control group (7.1 points vs. 1.4 points in EQ-5D-5L), lower emergency room visit rates (45 vs. 49%), and prolonged chemotherapy duration (8.2 vs. 6.3 months). In the PRO-TECT trial, a multicenter cluster randomized trial, the EORTC-QLQ-C30 scores in the intervention group compared to the control group at 3 months showed significant improvements with approximately 2.5 times higher QOL [44]. Moreover, the probability of experiencing clinically meaningful benefits favored the PRO group regarding physical functioning, symptom control, and health-related QOL (HRQOL; odds ratios 1.35, 1.50, and 1.41, respectively) compared to the standard care group [44]. In an intention-to-treat analysis assessing CANKADO PRO-React, the cumulative incidence of QOL deterioration was approximately 1.4 times lower in the CANKADO treatment group than in the control group [45]. Mika is a digital therapy aimed at alleviating pain associated with cancer and its treatments, to improve the QOL of patients [46]. Compared with the control group, the intervention group showed significant reductions in pain, depression, anxiety, and fatigue [46].

A meta-analysis conducted from 2016 to 2021 involving 2058 adult patients showed that remote patient monitoring software improved QOL and reduced physical symptom burden during cancer treatment [13]. Furthermore, a meta-analysis of 32 RCTs involving 7888 patients demonstrated that digital self-management symptom interventions significantly reduced symptom burden (effect size [ES] = − 0.230), pain (ES = − 0.292), fatigue (ES = − 0.417), anxiety (ES = − 0.320), and depression (ES = − 0.261) [13].

These studies highlight the significant potential of ePROs in enhancing symptom management, improving QOL, and reducing the burden of physical and psychological symptoms in patients with cancer.

Therapeutic outcomes

Digital therapeutics (DTx) and ePROs increase treatment adherence and enhance medical safety and quality, thereby improving treatment outcomes [4, 38, 40, 47]. The physician-reported symptoms such as fatigue, nausea, and constipation have been significantly associated with mortality and emergency room visits compared with those reported through ePROs [7], suggesting that physician-interpreted symptom reporting may better capture the more severe conditions of patients. ePRO-driven DTx can reduce symptom burden, unplanned hospitalizations, and medication non-adherence and improve adverse event management, QOL, and survival rates [4, 38]. The CAPRI-PRMS trial indicated that the relative intensity of treatment dosing was higher in the intervention group than in the control group (93.4 vs. 89.4%) [42].

Using ePROs to improve OS was first reported in 2017 [23, 24]. The STAR clinical trial is a randomized comparative trial with QOL as the primary outcome, and retrospective prognostic results have been reported [20]. The STAR group observed a significant improvement in median OS by approximately 5 months (median OS: intervention group 31.2 months vs. usual care group 26 months) [23]. Based on these findings, the PRO-TECT trial was a large-scale randomized trial conducted across multiple centers [44]. An electronic follow-up application (e-FAP) has proven highly reliable, detecting recurrences an average of 5 weeks earlier than regular imaging diagnostics [48, 49]. Subsequently, an RCT validating its prognosis revealed a median OS of 19 months (95% confidence interval [CI] 12.5 to not calculable) in the e-FAP group compared to 12 months (95% CI 8.6–16.4) in the control group (hazard ratio [HR] = 0.32, 95% CI 0.15–0.67) [24]. Subsequent 2-year follow-up reports added a median OS of 22.5 months in the e-FAP group vs. 14.9 months in the control group (without crossover termination; HR = 0.59 [95% CI 0.37–0.96]). Upon terminating crossover, the median OS was 22.5 months in the e-FAP group vs. 13.5 months in the control group (HR = 0.50 [95% CI 0.31–0.81]) [50].

Challenges of ePROs

Adherence

Adherence is a barrier to using ePROs. The barriers to ePRO use include patients, clinicians, and health system levels. Thirty percent of patients consistently failed to use ePRO systems because of adherence barriers. Additionally, adherence rates varied substantially based on demographic factors, with lower adherence observed among older adults and those with a lower socioeconomic status. Patient-level barriers include education, digital literacy, language barriers, and lack of understanding of the necessity of ePRO use. Physician-level barriers include being busy, difficulty making final decisions based on ePRO results, and uncertainty about integrating ePRO results into clinical practice. System-level barriers include a lack of technical support within hospitals and economic constraints [51].

Overburden on healthcare providers

Using ePRO systems can increase the workload of healthcare providers, who must review and act on the data, potentially leading to burnout and decreased efficiency [52, 53]. Although ePROs can facilitate comprehensive healthcare by assessing QOL and directly treating the primary condition, the healthcare provider burden remains an issue. The PRO-TECT trial highlighted challenges in the workload of nurses receiving alerts, prompting the development of algorithms to identify alerts with a high likelihood of non-urgency [52]. Ninety-one percent of the patients responded to weekly surveys, with alerts generated in 34% of the surveys and immediate nursing actions were taken in 59% of the alerts. Patients perceived 10% of the alerts as urgent. Nurses often classified alerts as urgent when patients reported worse symptoms than the previous week. The developed algorithm identified 38% of alerts as likely nonurgent [52].

Lack of formats in recent technologies

Numerous PROs exist, including those that report QOL and adverse events. The Patient-Reported Outcomes version of the Common Terminology Criteria for Adverse Events (PRO-CTCAE®), developed by the National Cancer Institute [54], is frequently used to report adverse events. Recording PROs on days when deterioration occurs can quickly convey the symptoms to healthcare providers because the side effects of cancer treatment can fluctuate daily. However, the recommended recall period for the PRO-CTCAE® is 1 week [55], limiting the real-time monitoring of adverse events in clinical practice. PRO tools such as the EORTC-QLQ and FACT are commonly used to evaluate the QOL of patients with cancer. However, these tools are copyrighted and not available for commercial use. Although paper-based PROs are relatively easy to collect, patients must answer numerous questions, making frequent data collection, and remote monitoring challenging.

Lack of technical support in hospitals

Many healthcare systems lack the necessary technical infrastructure and support to effectively implement and maintain ePRO systems [51]. Forty percent of healthcare facilities reported insufficient technical support as a significant barrier to the successful deployment of ePRO systems [51], hindering the integration of ePROs into routine clinical practice.

Economic concerns

The costs associated with implementing and maintaining ePRO systems can be a substantial barrier, particularly in resource-limited settings where financial constraints are a primary concern [51]. The initial setup and ongoing maintenance costs of ePRO systems were excessive, posing a barrier to widespread adoption [51]. Additionally, economic constraints are key challenges in sustaining these systems in low-income healthcare settings [51].

Future prospects and proposal of new technologies and approaches

Symptoms that ePRO should monitor

Early ePROs often used validated QOL scales. The EORTC-QLQ-C30 and Hospital Anxiety and Depression Scale used touchscreen technology in a 2004 intervention trial [56]. The ESRA-C also uses validated measures such as the SDS-15 and EORTC-QLQ-C30 [18]. However, minimizing the number of recorded symptoms is crucial for adherence (Fig. 2). Table 1 summarizes RCTs using the original symptom records via ePRO. Some ePROs allow for selecting and recording 11–17 symptoms from a broader list. Although specific symptoms vary according to disease and study objective, dyspnea and fatigue were consistently incorporated into four or more ePROs among 10 ePROs, followed by appetite loss, diarrhea, nausea, depression, pain, constipation, fever, vomiting, insomnia, anxiety, and neuropathy (Table 2). These symptoms are presumed to affect the QOL of patients across various cancer types.

Fig. 2.

Fig. 2

Patient care using new technologies and approaches. The number of clinical trials describing digital interventions for the quality of life and cancer has increased since 2021

Table 1.

List of randomized clinical trials using electronic patient-reported outcomes that record selected symptoms

ePRO or ClinicalTrials.gov Patient population Trial design Sample size Study arms Number of recorded symptoms Primary outcome Summarized findings Publication, trial setting
E-MOSAIC Patients receiving anticancer treatment in palliative intention RCT 102 E-MOSAIC intervention with the longitudinal monitoring sheet given to physicians vs. standard care 9–12 QOL Favor of the intervention for QOL, symptoms, communication, and coping Strasser et al. [19], Switzerland
NCT02004496 Patients receiving chemotherapy for breast cancer RCT 127 Mobile or web-based app with physician supervision vs. mobile or web-based app without physician supervision vs. standard care Selection from 48 QOL

Declined functional status was reported in the control and unsupervised app-using groups; however, no significant decline was observed in the supervised app-using group

More symptoms were reported in the app-using group than in the control group. More symptoms were reported in the unsupervised group than in the supervised group

Higher satisfaction with care was reported in the supervised app-using group than in the other groups

Egbring et al. [9], Switzerland
STAR Patients receiving chemotherapy for advanced solid cancer and metastatic disease RCT 766 Weekly patient-adapted CTCAE assessments vs. usual care 12 QOL

Improved HRQOL was reported among patients who completed the patient-adapted CTCAE assessments

Significantly lower emergency room admission for patients in the digital therapeutics group than in the usual care group

Longer chemotherapy periods in the digital therapeutics group than in the usual care group

Improved OS

Basch et al. JCO 2016

Basch et al. JAMA 2017, US

e-FAP

Moovcare

Patients with advanced-stage lung cancer undergoing systemic therapy RCT 121 Weekly electronic follow-up application (e-FAP) vs. usual care Weight and 11 symptoms OS Improved OS and higher performance status were observed at relapse in the intervention group than in the usual care group

Denis et al. JNCI [24]

Denis F et al. JAMA [50], France

Oncokompas Post-treatment patients with head and neck cancer, colorectal cancer, breast cancer, Hodgkin’s lymphoma, or non-Hodgkin’s lymphoma after treatment with curative intent (all treatment modalities) RCT 625 “Oncokompas” app vs. standard care 18 physical, four psychological, eight social, six lifestyle, one existential questions, and tumor-specific modules (6–11) Patient activation (knowledge, skills, and confidence for self-management

Better QOL was observed in the app-using group than in the control group at 6 months of follow-up

Less pain in the mouth, coughing, swallowing difficulty, and more social eating were observed in head and neck cancer survivors in the app-using group

Less weight change was observed in colorectal cancer survivors in the app-using group than in the control group

Less emotional impact was observed in lymphoma survivors in the app-using group than in the control group

No difference in reported symptoms was observed in breast cancer survivors

No significant difference was observed in the intervention and control groups regarding health management knowledge, attitude, self-efficacy, and supportive care needs

More app usage was reported in patients with higher education, having a partner, and currently working

Hout et al., 2019,

Hout et al. Acta Oncol 2021,

Netherlands

Interaktor Patients scheduled to undergo pancreaticoduodenectomy due to a suspected malignancy RCT 59 “Interaktor” app vs. standard care 14 QOL, self-care activity

Higher emotional function, less nausea, pain, appetite loss, constipation, pancreatic pain, flatulence, and weight loss. Worries and fewer hepatic symptoms were observed in the app-using group than in the control group at six weeks post-operation

No significant difference was observed between the app-using group and the control on engagement in self-care activities

Gustavell et al., 2019, Sweden
ChemOtheRapy Assistant (CORA) Patients with diverse malignancies who were prescribed oral therapy for cancer RCT 181 CORA app vs. standard care 17 Adherence, symptom burden, QOL

No significant difference was observed in all study measurements

Higher improvement in adherence rate in app-using participants with higher anxiety and poor baseline medication adherence were observed

Greer et al., 2020, US
BPSS Patients with breast cancer who had undergone anthracycline- or taxane-based chemotherapy regimens RCT 95 “BPSS” web app vs. standard care 12 QOL (psychological distress)

No significant difference between app-using and control groups was observed on depression subscales or health literacy skills

Less anxiety was observed in the app-using group than in the control group

The medical staff underestimated some grade 3 symptoms compared to the patient’s records

Handa et al. [10], Japan
SOFIA Patients who received immune checkpoint inhibition therapy RCT 71 Intervention group (SOFIA) vs. usual care Eleven physical and up to nine mental ePROs Feasibility of SOFIA

Higher feasibility and acceptance in the SOFIA group than in the control group

Better HRQOL and role functioning and less depression, distress, and appetite loss were observed in the SOFIA group post-intervention

No significant differences were observed regarding medical data, the utilization of supportive care services, or survival

Sauer et al. Cancer 2020, Germany
Msymptom Patients with breast cancer receiving adjuvant and neoadjuvant chemotherapy treatment for the first time RCT 57 “Msymptom” app vs. standard care Symptom control, QOL

Lower physical symptom burden was observed in the app-using group than in the control group

Higher scores were observed on the EORTC QOL-C30 symptom scale and nausea/vomiting score in the app-using group

Higher scores on EORTC QOL-BR23 sexual function and sexual pleasure subscales were observed in the app-using group

Öztürk et al., 2021, Turkey
eRAPID Patients with colorectal, breast, or gynecological cancer undergoing chemotherapy RCT 508 Weekly eRAPID vs. usual care 17 Symptom control

Improved physical well-being and self-efficacy were observed in the eRAPID group

Higher confidence in health management and self-rated overall health were observed in the app-using group

Absolom et al. JCO [36], UK
PRO-TECT Patients with metastatic cancer undergoing systemic therapy Cluster RCT 1191 Weekly PRO-CTCAE assessments vs. usual care 11 OS Improved physical function, symptom control, and HRQOL (OS pending) by weekly PROCTCAE assessments Basch et al. JAMA [44], US
CANKADO PRO-React Patients with HR+/HER2 locally advanced or metastatic breast cancer RCT 499 CANKADO-active arm vs. CONKADO-inactive inform arm 13 QOL (psychological distress) Favorable QOL in CANKADO-active arm Harbeck et al. Ann Oncol [45], Germany
Mika Adult patients with malignant tumors RCT 218 Mika-App and usual care vs. usual care Selection from 58 options QOL Higher reductions were observed in distress, depression, anxiety, and fatigue were observed in the intervention group than in the control group Spring et al. J Med Internet Res [46], Germany

CTCAE Common Terminology Criteria for Adverse Events, e-FAP electronic follow-up application, HRQOL health-related quality of life, QOL quality of life, OS overall survival, RCT randomized controlled trial

Table 2.

Selected symptoms that were applied in the electronic patient-reported outcomes

Total E-MOSAIC STAR e-FAP Oncokompas Interaktor BPSS SOFIA eRAPID PRO-TECT CANKADO
Dyspnea/shortness of breath 9
Fatigue 9
Appetite loss 8
Diarrhea 8
Nausea 8
Depression 6
Pain 6
Constipation 6
Fever/chills 6
Vomiting 6
Insomnia/drowsiness 5
Anxiety 4
Neuropathy 4
Cough/dry cough 3
Mucositis/oral problems 3
Dysuria/reduced urinary output 2
Hot flashes/night sweats 2
Joint pain/muscle pain 2
Rash/skin toxicity 2
Skin changes 2
Visual disturbances 2
An occurrence of a lump under the skin 1
Appearance or increase of blood in sputum 1
Bleeding 1
Falls 1
Financial challenges 1
Flu-like symptoms 1
Hair loss 1
Headache 1
Melaena 1
Nose bleeds 1
Overall well-being 1
Palmar-plantar erythema 1
Performance status 1
Psychological distress 1
Stomatitis 1
Sudden face swelling 1
Swelling/pain/redness in the arm 1
Thrombocytic purpura 1
Voice changing 1
Weakness 1
Weight 1
Yellow coloring 1
Others 3

e-FAP electronic follow-up application, E-MOSAIC Electronic Monitoring of Symptoms and Syndromes Associated with Cancer, PRO-TECT Patient-Reported Outcomes To Enhance Cancer Treatment, STAR Symptom Tracking and Reporting

Use of lifelogs

Methods unrelated to patient adherence should be used and digital information related to patients’ daily lives (lifelogs) helps evaluate QOL (Fig. 2). Using lifelogs can minimize the influence of adherence and language barriers. Integrating real-world big data in a reliable cancer research environment may assist clinicians and researchers in developing new models for predictive and preventive algorithms and personalized care [57]. Diverse lifelogs were collected from approximately 100 patients with gynecological cancer, and associations with QOL-related lifelogs were observed, particularly with heart rate variability (unpublished). Heart rate variability, measured from R waves and their fluctuations, can be assessed using a smartphone camera. Thus, leveraging digital information from wearable devices and mobile applications enables cancer healthcare management without the patient recording the data, thereby representing a future direction of cancer treatment. Illustrating how real-world big data can contribute to the development of new models for predictive and preventive algorithms and personalized care is crucial.

Automatic alert systems

In 2022, the European Society for Medical Oncology released clinical practice guidelines for patient-reported outcome measurements [58]. These guidelines specifically address the recommendations of ePRO for patients with cancer. The automatic alert functions that notify clinicians of severe or worsening symptoms are recommended for patients undergoing active cancer treatment [58]. However, overburdening healthcare providers with automatic alert functions is challenging for clinicians. Therefore, an automatic alert system for patients can be developed to reduce the burden on healthcare providers (Fig. 2).

CANKADO PRO-React represents an advanced next-generation ePRO, suggesting actions to patients automatically rather than alerting healthcare providers. These recommendations encourage behavioral changes in patients categorized into observation until the next visit (Level 1), prompt contact with the center (Level 2), or immediate contact or emergency visit (Level 3) [45]. Mika is a next-generation ePRO comprising three modules: Check-up, Discover, and Journeys [46]. The Check-up module monitors pain and symptoms, enabling electronic outcomes reported by patients to be shared and discussed with healthcare providers. The Discover module provides coaching through articles and videos on cancer types, treatments, psychological health, physical activity, diet, and socioeconomic issues. In the Journeys module, AI algorithms in the Mika application customize content for each patient based on cancer type, treatment stage, and the reported symptom nature and severity, prompting behavior change and ensuring personalized support for each individual [46]. Oncokompas, a Dutch-approved ePRO, is divided into three components: Measure, Learner, and Act components [59]. However, no significant differences were observed in patient activation (knowledge, skills, or confidence in self-management) between the intervention and control groups [59]. Nevertheless, it suggested effectiveness for HRQOL in cancer survivors with moderate self-efficacy, high personal control, and high health literacy scores, particularly for specific symptoms such as mouth pain in head and neck cancer, social eating, swallowing, coughing, and weight in colorectal cancer [60].

ePROs have been developed to promote behavioral changes in patients and reduce the burden on healthcare providers. However, patients expressed a desire for clinicians to acknowledge the results of an ePRO system called MyChristie-MyHealth [61], indicating the crucial role of physicians in patient care. ePRO-promoting behavioral change in patients has demonstrated improvement in symptoms and QOL enhancement. However, safety evaluations and patient satisfaction are crucial and future research should verify their impact on treatment outcomes.

Conclusions

ePROs enhance patient–provider communication, educate patients and caregivers, facilitate the monitoring of patient conditions and self-management, and improve adverse event management and QOL, thereby potentially improving treatment outcomes and reducing healthcare expenditures. To fully capitalize on the advantages of ePROs, future advancements should prioritize developing lifelogs independent of adherence to assess patient symptoms and QOL and create automatic alert systems for patients to alleviate the burden on healthcare providers while rigorously evaluating their safety and efficacy.

Acknowledgements

The authors thank Editage (https://www.editage.jp) for the English language editing.

Author contributions

All authors contributed to the study conception and design. Conceptualization was provided by Ken as provided by Ken Yamaguchi. Material preparation, data collection and analysis were performed by Ken Yamaguchi, Nozomi Higashiyama, Maki Umamiya, Yoshihide Inayama, and Ayami Koike. The first draft of the manuscript was written by Ken Yamaguchi and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript. Masaki Mandai supervised this study.

Funding

The authors did not receive support from any organization for the submitted work.

Declarations

Conflict of interest

K. Yamaguchi reports personal fees and other support from DUMSCO Inc. and personal fees from Takeda Pharmaceutical Company Limited, Eisai Co., Ltd., Daiichi Sankyo, Inc., and Merck & Co., Inc. outside the submitted work. M. Mandai reports support from DUMSCO, Inc. outside the submitted work. N Higashiyama reports personal fees from DUMSCO Inc. outside the submitted work. No competing interests have been reported by the other authors.

Footnotes

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

  • 1.Leung KKY, Ray S, Chan GCF et al (2022) Functional outcomes at PICU discharge in hemato-oncology children at a tertiary oncology center in Hong Kong. Int J Clin Oncol 27(12):1904–1915. 10.1007/s10147-022-02244-3 [DOI] [PubMed] [Google Scholar]
  • 2.Jordan K, Aapro M, Kaasa S et al (2018) European society for medical oncology (ESMO) position paper on supportive and palliative care. Ann Oncol 29(1):36–43. 10.1093/annonc/mdx757 [DOI] [PubMed] [Google Scholar]
  • 3.Kaasa S, Loge JH, Aapro M et al (2018) Integration of oncology and palliative care: a lancet oncology commission. Lancet Oncol 19(11):e588–e653. 10.1016/S1470-2045(18)30415-7 [DOI] [PubMed] [Google Scholar]
  • 4.Gussoni G, Ravot E, Zecchina M et al (2022) Digital therapeutics in oncology: findings, barriers and prospects. A narrative review. Ann Res Oncol 2(1):55–69. 10.4828/aro.2022.39 [Google Scholar]
  • 5.Inayama Y, Mizuno K, Yamaguchi K et al (2023) Real-world practice of estrogen therapy after surgery for endometrial cancer: a descriptive study using a Japanese claims database. Int J Clin Oncol 28(3):445–453. 10.1007/s10147-022-02289-4 [DOI] [PubMed] [Google Scholar]
  • 6.Sasaki H, Tamura K, Naito Y et al (2017) Patient perceptions of symptoms and concerns during cancer chemotherapy: 'affects my family’ is the most important. Int J Clin Oncol 22(4):793–800. 10.1007/s10147-017-1117-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Basch E, Jia X, Heller G et al (2009) Adverse symptom event reporting by patients vs clinicians: relationships with clinical outcomes. J Natl Cancer Inst 101(23):1624–1632. 10.1093/jnci/djp386 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Basch E (2010) The missing voice of patients in drug-safety reporting. N Engl J Med 362(10):865–869. 10.1056/NEJMp0911494 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Egbring M, Far E, Roos M et al (2016) A mobile app to stabilize daily functional activity of breast cancer patients in collaboration with the physician: a randomized controlled clinical trial. J Med Internet Res 18(9):e238. 10.2196/jmir.6414 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Handa S, Okuyama H, Yamamoto H et al (2020) Effectiveness of a smartphone application as a support tool for patients undergoing breast cancer chemotherapy: a randomized controlled trial. Clin Breast Cancer 20(3):201–208. 10.1016/j.clbc.2020.01.004 [DOI] [PubMed] [Google Scholar]
  • 11.Guidance for industry patient-reported outcome measures: use in medical product development to support labeling claims. (2009). https://www.fda.gov/media/77832/download. Accessed on 30 July 2024 [DOI] [PMC free article] [PubMed]
  • 12.Di Maio M, Basch E, Bryce J et al (2016) Patient-reported outcomes in the evaluation of toxicity of anticancer treatments. Nat Rev Clin Oncol 13(5):319–325. 10.1038/nrclinonc.2015.222 [DOI] [PubMed] [Google Scholar]
  • 13.Wang T, Ho MH, Tong MCF et al (2023) Effects of patient-reported outcome tracking and health information provision via remote patient monitoring software on patient outcomes in oncology care: a systematic review and meta-analysis. Semin Oncol Nurs 39(5):151473. 10.1016/j.soncn.2023.151473 [DOI] [PubMed] [Google Scholar]
  • 14.Vodicka E, Kim K, Devine EB et al (2015) Inclusion of patient-reported outcome measures in registered clinical trials: evidence from ClinicalTrials.gov (2007–2013). Contemp Clin Trials 43:1–9. 10.1016/j.cct.2015.04.004 [DOI] [PubMed] [Google Scholar]
  • 15.Chen J, Ou L, Hollis SJ (2013) A systematic review of the impact of routine collection of patient reported outcome measures on patients, providers and health organisations in an oncologic setting. BMC Health Serv Res 13:211. 10.1186/1472-6963-13-211 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kotronoulas G, Kearney N, Maguire R et al (2014) What is the value of the routine use of patient-reported outcome measures toward improvement of patient outcomes, processes of care, and health service outcomes in cancer care? A systematic review of controlled trials. J Clin Oncol 32(14):1480–1501. 10.1200/JCO.2013.53.5948 [DOI] [PubMed] [Google Scholar]
  • 17.Howell D, Molloy S, Wilkinson K et al (2015) Patient-reported outcomes in routine cancer clinical practice: a scoping review of use, impact on health outcomes, and implementation factors. Ann Oncol 26(9):1846–1858. 10.1093/annonc/mdv181 [DOI] [PubMed] [Google Scholar]
  • 18.Berry DL, Hong F, Halpenny B et al (2014) Electronic self-report assessment for cancer and self-care support: results of a multicenter randomized trial. J Clin Oncol 32(3):199–205. 10.1200/JCO.2013.48.6662 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Strasser F, Blum D, von Moos R et al (2016) The effect of real-time electronic monitoring of patient-reported symptoms and clinical syndromes in outpatient workflow of medical oncologists: E-MOSAIC, a multicenter cluster-randomized phase III study (SAKK 95/06). Ann Oncol 27(2):324–332. 10.1093/annonc/mdv576 [DOI] [PubMed] [Google Scholar]
  • 20.Basch E, Deal AM, Kris MG et al (2016) Symptom monitoring with patient-reported outcomes during routine cancer treatment: a randomized controlled trial. J Clin Oncol 34(6):557–565. 10.1200/JCO.2015.63.0830 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Lee K, Kim S, Kim SH et al (2023) Digital health interventions for adult patients with cancer evaluated in randomized controlled trials: scoping review. J Med Internet Res 25:e38333. 10.2196/38333 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Cleeland CS, Wang XS, Shi Q et al (2011) Automated symptom alerts reduce postoperative symptom severity after cancer surgery: a randomized controlled clinical trial. J Clin Oncol 29(8):994–1000. 10.1200/JCO.2010.29.8315 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Basch E, Deal AM, Dueck AC et al (2017) Overall survival results of a trial assessing patient-reported outcomes for symptom monitoring during routine cancer treatment. JAMA 318(2):197–198. 10.1001/jama.2017.7156 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Denis F, Lethrosne C, Pourel N et al (2017) Randomized trial comparing a web-mediated follow-up with routine surveillance in lung cancer patients. J Natl Cancer Inst. 10.1093/jnci/djx029 [DOI] [PubMed] [Google Scholar]
  • 25.Hashimoto T, Nakamura Y, Oki E et al (2024) Bridging horizons beyond CIRCULATE-Japan: a new paradigm in molecular residual disease detection via whole genome sequencing-based circulating tumor DNA assay. Int J Clin Oncol 29(5):495–511. 10.1007/s10147-024-02493-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Pearce FJ, Cruz Rivera S, Liu X et al (2023) The role of patient-reported outcome measures in trials of artificial intelligence health technologies: a systematic evaluation of ClinicalTrials.gov records (1997–2022). Lancet Digit Health 5(3):e160–e167. 10.1016/S2589-7500(22)00249-7 [DOI] [PubMed] [Google Scholar]
  • 27.Pe M, Alanya A, Falk RS et al (2023) Setting international standards in analyzing patient-reported outcomes and quality of life endpoints in cancer clinical trials-innovative medicines initiative (SISAQOL-IMI): stakeholder views, objectives, and procedures. Lancet Oncol 24(6):e270–e283. 10.1016/S1470-2045(23)00157-2 [DOI] [PubMed] [Google Scholar]
  • 28.Yang A, Moore TC, Iacovella CR et al (2020) Examining tail and headgroup effects on binary and ternary gel-phase lipid bilayer structure. J Phys Chem B 124(15):3043–3053. 10.1021/acs.jpcb.0c00490 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Knowles H, Swoboda TK, Sandlin D et al (2023) The association between electronic health information usage and patient-centered communication: a cross sectional analysis from the health information national trends survey (HINTS). BMC Health Serv Res 23(1):1398. 10.1186/s12913-023-10426-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Berry DL, Blumenstein BA, Halpenny B et al (2011) Enhancing patient-provider communication with the electronic self-report assessment for cancer: a randomized trial. J Clin Oncol 29(8):1029–1035. 10.1200/JCO.2010.30.3909 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Basch E, Stover AM, Schrag D et al (2020) Clinical Utility and user perceptions of a digital system for electronic patient-reported symptom monitoring during routine cancer care: findings from the PRO-TECT Trial. JCO Clin Cancer Inform 4:947–957. 10.1200/CCI.20.00081 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Gvozdanovic A, Jozsa F, Fersht N et al (2022) Integration of a personalised mobile health (mHealth) application into the care of patients with brain tumours: proof-of-concept study (IDEAL stage 1). BMJ Surg Interv Health Technol 4(1):e000130. 10.1136/bmjsit-2021-000130 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Meirte J, Hellemans N, Anthonissen M et al (2020) Benefits and disadvantages of electronic patient-reported outcome measures: systematic review. JMIR Perioper Med 3(1):e15588. 10.2196/15588 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Kawaguchi K, Kawazoe H, Sakurai T et al (2020) Effect of general self-efficacy on promoting health-related quality of life during recovery from radical prostatectomy: a 1-year prospective study. Int J Clin Oncol 25(12):2122–2129. 10.1007/s10147-020-01765-z [DOI] [PubMed] [Google Scholar]
  • 35.Holch P, Warrington L, Bamforth LCA et al (2017) Development of an integrated electronic platform for patient self-report and management of adverse events during cancer treatment. Ann Oncol 28(9):2305–2311. 10.1093/annonc/mdx317 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Absolom K, Warrington L, Hudson E et al (2021) Phase III randomized controlled trial of eRAPID: eHealth Intervention during chemotherapy. J Clin Oncol 39(7):734–747. 10.1200/JCO.20.02015 [DOI] [PubMed] [Google Scholar]
  • 37.Kruse CS, Goswamy R, Raval Y et al (2016) Challenges and opportunities of big data in health care: a systematic review. JMIR Med Inform 4(4):e38. 10.2196/medinform.5359 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Aapro M, Bossi P, Dasari A et al (2020) Digital health for optimal supportive care in oncology: benefits, limits, and future perspectives. Support Care Cancer 28(10):4589–4612. 10.1007/s00520-020-05539-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kenney LB, Antal Z, Ginsberg JP et al (2018) Improving male reproductive health after childhood, adolescent, and young adult cancer: progress and future directions for survivorship research. J Clin Oncol 36(21):2160–2168. 10.1200/JCO.2017.76.3839 [DOI] [PubMed] [Google Scholar]
  • 40.Kolb NA, Smith AG, Singleton JR et al (2018) Chemotherapy-related neuropathic symptom management: a randomized trial of an automated symptom-monitoring system paired with nurse practitioner follow-up. Support Care Cancer 26(5):1607–1615. 10.1007/s00520-017-3970-7 [DOI] [PubMed] [Google Scholar]
  • 41.Berry DL, Blonquist TM, Patel RA et al (2015) Exposure to a patient-centered, Web-based intervention for managing cancer symptom and quality of life issues: impact on symptom distress. J Med Internet Res 17(6):e136. 10.2196/jmir.4190 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ferrua M, Minvielle E, Fourcade A et al (2020) How to design a remote patient monitoring system? A French case study. BMC Health Serv Res 20(1):434. 10.1186/s12913-020-05293-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Mir O, Ferrua M, Fourcade A et al (2022) Digital remote monitoring plus usual care versus usual care in patients treated with oral anticancer agents: the randomized phase 3 CAPRI trial. Nat Med 28(6):1224–1231. 10.1038/s41591-022-01788-1 [DOI] [PubMed] [Google Scholar]
  • 44.Basch E, Schrag D, Henson S et al (2022) Effect of electronic symptom monitoring on patient-reported outcomes among patients with metastatic cancer: a randomized clinical trial. JAMA 327(24):2413–2422. 10.1001/jama.2022.9265 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Harbeck N, Fasching PA, Wuerstlein R et al (2023) Significantly longer time to deterioration of quality of life due to CANKADO PRO-React eHealth support in HR+ HER2- metastatic breast cancer patients receiving palbociclib and endocrine therapy: primary outcome analysis of the multicenter randomized AGO-B WSG PreCycle trial. Ann Oncol 34(8):660–669. 10.1016/j.annonc.2023.05.003 [DOI] [PubMed] [Google Scholar]
  • 46.Springer F, Maier A, Friedrich M et al (2024) Digital therapeutic (Mika) targeting distress in patients with cancer: results from a nationwide waitlist randomized controlled trial. J Med Internet Res 26:e51949. 10.2196/51949 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ferretti A, Ronchi E, Vayena E (2019) From principles to practice: benchmarking government guidance on health apps. Lancet Digit Health 1(2):e55–e57. 10.1016/S2589-7500(19)30027-5 [DOI] [PubMed] [Google Scholar]
  • 48.Denis F, Viger L, Charron A et al (2014) Detecting lung cancer relapse using self-evaluation forms weekly filled at home: the sentinel follow-up. Support Care Cancer 22(1):79–85. 10.1007/s00520-013-1954-9 [DOI] [PubMed] [Google Scholar]
  • 49.Denis F, Viger L, Charron A et al (2014) Detection of lung cancer relapse using self-reported symptoms transmitted via an internet web-application: pilot study of the sentinel follow-up. Support Care Cancer 22(6):1467–1473. 10.1007/s00520-013-2111-1 [DOI] [PubMed] [Google Scholar]
  • 50.Denis F, Basch E, Septans AL et al (2019) Two-year survival comparing web-based symptom monitoring vs routine surveillance following treatment for lung cancer. JAMA 321(3):306–307. 10.1001/jama.2018.18085 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Narra LR, Verdini N, Lapen K et al (2023) Patient-reported outcomes in clinical trials: from an endpoint to an intervention in cancer care. Semin Radiat Oncol 33(4):358–366. 10.1016/j.semradonc.2023.07.002 [DOI] [PubMed] [Google Scholar]
  • 52.Mazza GL, Dueck AC, Ginos B et al (2024) Optimization of alert notifications in electronic patient-reported outcome (ePRO) remote symptom monitoring systems (AFT-39). Qual Life Res 33(7):1985–1995. 10.1007/s11136-024-03675-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kiderlen TR, Schnack A, de Wit M (2022) Essential barriers and considerations for the implementation of electronic patient-reported outcome (ePRO) measures in oncological practice: contextualizing the results of a feasibility study with existing literature. J Public Health. 10.1007/s10389-022-01767-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Basch E, Reeve BB, Mitchell SA et al (2014) Development of the national cancer institute’s patient-reported outcomes version of the common terminology criteria for adverse events (PRO-CTCAE). J Natl Cancer Inst. 10.1093/jnci/dju244 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Mendoza TR, Dueck AC, Bennett AV et al (2017) Evaluation of different recall periods for the US national cancer institute’s PRO-CTCAE. Clin Trials 14(3):255–263. 10.1177/1740774517698645 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Velikova G, Booth L, Smith AB et al (2004) Measuring quality of life in routine oncology practice improves communication and patient well-being: a randomized controlled trial. J Clin Oncol 22(4):714–724. 10.1200/JCO.2004.06.078 [DOI] [PubMed] [Google Scholar]
  • 57.Recommended dietary allowances for the elderly (1986). N Engl J Med 314 (26):1707–1710. 10.1056/NEJM198606263142612 [DOI] [PubMed]
  • 58.Di Maio M, Basch E, Denis F et al (2022) The role of patient-reported outcome measures in the continuum of cancer clinical care: ESMO clinical practice guideline. Ann Oncol 33(9):878–892. 10.1016/j.annonc.2022.04.007 [DOI] [PubMed] [Google Scholar]
  • 59.van der Hout A, van Uden-Kraan CF, Holtmaat K et al (2020) Role of eHealth application Oncokompas in supporting self-management of symptoms and health-related quality of life in cancer survivors: a randomized, controlled trial. Lancet Oncol 21(1):80–94. 10.1016/S1470-2045(19)30675-8 [DOI] [PubMed] [Google Scholar]
  • 60.van der Hout A, Holtmaat K, Jansen F et al (2021) The eHealth self-management application ‘Oncokompas’ that supports cancer survivors to improve health-related quality of life and reduce symptoms: which groups benefit most? Acta Oncol 60(4):403–411. 10.1080/0284186X.2020.1851764 [DOI] [PubMed] [Google Scholar]
  • 61.Shipman LA, Price J, Abdulwahid D et al (2024) Service evaluation of MyChristie-MyHealth, an electronic patient-reported outcome measure integrated into clinical cancer care. JCO Clin Cancer Inform 8:e2300162. 10.1200/CCI.23.00162 [DOI] [PubMed] [Google Scholar]

Articles from International Journal of Clinical Oncology are provided here courtesy of Springer

RESOURCES