Abstract
As new cancer therapies emerge and evolve, there is a need for a better understanding of their safety and tolerability. Safety and tolerability are distinct, albeit related, constructs; an unsafe treatment cannot be considered tolerable, whereas safe treatments may not be tolerable to some patients. Cancer treatment tolerability is a multidimensional construct, and is influenced both by the profile of adverse events and by whole‐person factors. This commentary provides definitions of safety, tolerability, adverse events, and toxicity by relating these constructs to the adverse event reporting paradigm. Measures, including summary indicators, that reflect the tolerability of cancer treatments are also identified. The commentary concludes with a discussion of how evaluations of tolerability may be meaningfully incorporated into the design and interpretation of future cancer treatment trials.
Keywords: cancer treatment, Common Terminology Criteria for Adverse Events (CTCAE), patient‐reported outcomes, Patient‐Reported Outcomes version of the Common Terminology Criteria for Adverse Events (PRO‐CTCAE), tolerability, toxicity
Short abstract
An understanding of treatment tolerability includes both the profile of adverse events and summary indicators of tolerability, interpreted in the context of whole‐person factors. Tolerability measurements that incorporate both clinician‐ and patient‐reported outcomes support the most complete understanding of the benefits and risks of a cancer treatment, and are essential for shared decision‐making and the provision of optimized supportive care.
EVOLVING PERSPECTIVES ON CANCER TREATMENT TOLERABILITY
Safety and tolerability are fundamental to drawing conclusions about the effectiveness of cancer therapies, including comparative effectiveness, and complement clinical trial end points such as progression‐free survival, overall survival, and clinical benefit. 1 All cancer‐directed therapies have the potential to cause toxicities that can result in early treatment discontinuation, dose reductions, or patient nonadherence to therapy and, as a consequence, inferiorities in cancer treatment outcomes 2 , 3 and impairments in health‐related quality of life (HRQOL) 4 and functional status. 5 , 6
In this commentary, we outline key concepts to be considered when evaluating cancer treatment tolerability. We begin by offering definitions of tolerability and related concepts such as adverse events (AEs), toxicity, and safety by situating these concepts within the AE monitoring paradigm. This is followed by a summary of direct and indirect approaches to the measurement of tolerability, and a brief consideration of the ways in which whole‐person factors may meaningfully shape our interpretations and conclusions about cancer treatment tolerability, at both the group/trial level and the person/individual level. We conclude by highlighting selected tolerability‐related considerations in cancer treatment trial design.
GAUGING SAFETY AND TOLERABILITY VIA AE MONITORING
AEs, toxicity, safety, and tolerability
A comprehensive understanding of tolerability must also incorporate an understanding of related constructs including AEs, toxicity, and safety and the related constructs of dose‐limiting toxicity, serious adverse events (SAEs), and maximum tolerated dose (MTD). Table 1 presents definitions of these key constructs drawn from the pharmacovigilance literature. AEs are harmful, unfavorable occurrences that arise during medical treatment or the conduct of a clinical trial, regardless of whether the event is attributable to the treatment. AEs that are determined to be probably or possibly related to a treatment are considered to represent the toxicities or side effects of that treatment (see Table 1).
TABLE 1.
Concepts and definitions.
| Concept | Definition |
|---|---|
| Adverse event | Any unexpected, harmful, or unfavorable occurrences, medical conditions, symptoms, or signs that happen during medical treatment or a clinical trial. The occurrence of an AE does not necessarily mean that the event is definitively attributable to the treatment being studied in a trial |
| Dose‐limiting toxicity | Describes side effects of a treatment that are serious enough to prevent an increase in the dose level of that treatment |
| Maximum tolerated dose | The highest dose of a treatment that does not cause unacceptable side effects. The maximum tolerated dose is determined in clinical trials by testing increasing doses on different groups of people until the highest dose with acceptable side effects is found |
| Serious adverse event | A serious adverse event refers to any expected or unexpected AE, related or unrelated to the therapy being studied, occurring at any agent dose, any phase of product or procedure testing, that results in death, a life‐threatening AE, requirement of inpatient hospitalization or prolongation of existing hospitalization, or a persistent or significant disability or incapacity |
| Safety | Safety is an evaluation process designed to detect, assess, monitor, prevent, and understand AEs experienced by patients receiving a treatment regimen. An evaluation of safety yields an understanding of the risks, harms, and benefits of a treatment, medication, or therapeutic regimen. Safety includes the frequency, seriousness, severity, reversibility, duration, and consequences of observed AEs and reactions |
| Toxicity | A potentially harmful, untoward, or unpleasant reaction that results from or is associated with an intervention or medicinal product, and that may be (1) associated with a clinical sign or syndrome, (2) reflected in a laboratory test or clinical investigation, and/or (3) experienced by the patient as a sign or symptom |
| Tolerability | The degree to which AEs resulting from or associated with a treatment affect the ability or desire of the patient to adhere to the planned dose and/or schedule of a regimen or therapy. A complete understanding of tolerability should include direct measurement from the patient of how they are feeling and functioning while receiving the treatment |
Note: Based on information from Aronson 2023, 7 Friends of Cancer Research 2025, 8 National Cancer Institute 2025, 9 National Center for Advancing Translational Sciences 2025, 142 Peipert & Smith 2022, 10 Ray 2018, 11 Shader 2018, 12 Singh & Loke 2012, 13 and Stanulović 2022. 14
Abbreviation: AE, adverse event.
Safety, with respect to the development and testing of new medical treatments, is an evaluation process designed to detect, assess, monitor, prevent, and understand AEs, including SAEs, experienced by patients receiving a treatment regimen. Tolerability is the degree to which AEs affect the ability or desire of the patient to adhere to the planned dose and/or schedule of a treatment regimen. On the basis of the expectation that chemotherapy agents have a monotonically increasing dose–efficacy relationship, conventional phase 1/2 clinical trial designs have focused on identifying the MTD. Determinations about the recommended phase 2 dose (for use in subsequent trials) are then determined on the basis of the highest dose that will minimize SAEs and cumulative toxicity and provide long‐term tolerability. 15
However, newer cancer treatment approaches, such as molecularly targeted agents and immunotherapies, are often not dose dependent. Thus, cancer treatment trial designs are emerging that do not escalate to the MTD, in favor of finding the dose or range of doses that optimize the effectiveness of an agent. This paradigm shift has stimulated the use of novel trial designs that identify the dose and/or schedule of a drug that is tolerable without sacrificing efficacy. 16 , 17 It has also accelerated the imperative for the inclusion of patient‐centered indicators of treatment tolerability in early‐ and later phase trials of all new investigational cancer therapies. 18 , 19 , 20
Although the terms “safety” and “tolerability” are sometimes used interchangeably, they represent somewhat different concepts. 14 , 21 For example, an agent may be considered tolerable, whereas laboratory testing or other evaluations show it to be unsafe (e.g., producing irreversible renal insufficiency). Conversely, a treatment may be safe on the basis of objective clinician assessments, including relevant laboratory or diagnostic studies, and yet that treatment may produce toxicities that are unacceptable to the patient or that warrant dose interruption or discontinuation. Conclusions about safety and tolerability should be based on a systematic and generalizable evaluation of the absence or presence of harms, as determined by relevant questioning, observation, and testing. 12
Safety and tolerability are considered in terms of the risks and the magnitude of harm, and particularly with respect to tolerability, there is a value judgment of the acceptability of those risks based on their probability of occurring and their effect on patients. 22 The health condition for which the treatment is being given is also relevant because judgments about acceptable risk depend on the type and severity of the underlying health problem being addressed, the treatment alternatives, and the magnitude of potential benefit. Relevant contextual factors in drawing conclusions about safety and tolerability also include the population (and any inherent subgroup characteristics that might increase susceptibility to adverse effects) and the specific conditions of use (e.g., research settings vs. nonresearch settings). 12 , 21 , 23 This judgment about the acceptability of the risks must be deliberated from the perspectives of patients, clinicians, society, and other appropriate decision makers. 24 , 25 , 26
It is important to note that the concept of tolerability should be considered as both a between‐group and a within‐person construct. 10 , 27 Interpretation at the group or trial level allows for generalizable comparisons and conclusions about the aggregated tolerability of a specific treatment regimen being tested in a trial. Within‐person interpretations of treatment tolerability can be applied to inform patient–clinician shared decision‐making and to individualize the provision of toxicity‐directed interventions and self‐management support. 28 , 29
AE‐monitoring paradigm
AE monitoring, severity rating, and reporting are essential components of any clinical trial evaluating a new cancer therapy. AE monitoring is the process of systematically identifying and tracking unfavorable clinical outcomes associated with a treatment regimen to draw conclusions about its safety and tolerability. Therapeutic response to a new cancer therapy is determined on the basis of outcomes such as survival, disease modification, clinicians’ global impression of treatment benefit, and patient‐reported outcomes (PROs) such as HRQOL gathered at periodic prespecified intervals. In contrast, safety and tolerability determinations require continuous surveillance and monitoring for AEs across the course of a trial, together with a standardized approach to categorization and severity rating. Important characteristics to understand about AEs include not only their specific nature (e.g., neuropathy, nausea, acute kidney injury, noninfectious pneumonia, gait disturbance, and ileus) but also their severity, reversibility, consequences, chronicity, and pattern of onset, offset, accumulation, and co‐occurrence. 30 , 31 AE monitoring ensures that individual study participants are not harmed by treatment, and allows for generalizable conclusions to be drawn at the trial level regarding a regimen’s safety and tolerability and the anticipated profile of treatment‐related toxicities. A well‐characterized AE profile derived at the trial level also informs individual‐level provision of anticipatory guidance and the delivery of targeted supportive care to mitigate and manage adverse effects and deleterious outcomes.
MEASURING THE MULTIDIMENSIONAL ASPECTS OF TOLERABILITY
Over the past several years, there has been growing momentum to more systematically incorporate the patient’s perspective into AE reporting and the evaluation of cancer treatment tolerability. 20 , 32 , 33 This has been motivated in part by the greater use of novel immunologic and molecularly targeted agents that are chronically administered by patients in their homes, often over months or years. 34 These novel agents may also have a more subtle, distinctive, and/or enduring toxicity profile, 35 and chronic, lower grade but still bothersome toxicities have been implicated in elective treatment discontinuation and patient nonadherence to therapy. 36 , 37 A growing consensus that patients can best gauge outcomes such as symptoms, function, and HRQOL has accompanied this trend, 38 , 39 together with an expanding interest in using comparative effectiveness methods 40 and value frameworks 41 , 42 to improve the patient centeredness of care and provide tailored, personalized treatment. The definition of tolerability offered in Table 1 emphasizes the patient centeredness of this construct, in which tolerability reflects the extent to which the toxicities and harms associated with a treatment regimen can be accepted by a patient.
In addition to PROs, other indicators of tolerability include rates of Common Terminology Criteria for Adverse Events (CTCAE) grade 2 or 3 AEs, dose reductions/dose interruptions and treatment discontinuation due to AEs, hospitalizations, and elective patient discontinuations due to moderate or severe toxicity. Although tolerability is directly related to the profile of treatment‐related AEs, it is also influenced by patient‐ and system‐level factors, including comorbidities, treatment preferences, physiologic resilience, psychosocial health, health behaviors, age/developmental stage, lifestyle, geographic environment, socioeconomic status, and social support. 10 , 38 , 43 Thus, a full understanding of tolerability requires a comprehensive ascertainment of the profile of toxicities considered in the context of whole‐person factors that may influence an individual patient’s capacity, desire, or willingness to persist with treatment, although not all of these domains will be measured in any given trial.
As shown in Figure 1, this tolerability framework situates the domains articulated by the Food and Drug Administration’s core PROs in cancer trials draft guidance, such as symptomatic AEs, overall side effect impact summary measures, and physical and role functioning, 44 within the broader AE‐reporting paradigm (which encompasses a comprehensive AE profile as well as summary indicators of tolerability, as discussed below). It serves as a unifying conceptual perspective via which trialists, PRO‐focused investigators, patient advocates, and regulators can consider what tolerability domains to measure and how to simultaneously interpret both clinician‐ and patient‐reported tolerability indicators.
FIGURE 1.

Multidimensional aspects of tolerability.
Profiling of AEs
Monitoring and characterization of AEs are essential aspects of every cancer clinical trial, which ensure that individual study participants are not harmed by treatment and allow for valid, reliable, and generalizable conclusions about a regimen’s safety and tolerability to be made at the trial level. The standard system for defining, classifying, reporting, and monitoring AEs in cancer clinical trials is the National Cancer Institute’s CTCAE, now in version 5. 45 The CTCAE was designed for evaluating safety. CTCAE toxicities are graded by health care providers on the basis of the clinician’s review of systems, physical examination, and review of data derived from laboratory and diagnostic studies. Clinicians incorporate patient reporting of their experiences into the assignment of CTCAE grades; however, notably, these experiences are filtered through the interpretations of a clinician. The CTCAE provides for standardization and efficiency across studies in the identification and severity grading of AEs, including asymptomatic AEs.
There are several limitations to measuring tolerability via the CTCAE alone. First, although high‐grade AEs (grade 3 [medically significant], grade 4 [life threatening], and grade 5 [death]) have important implications for treatment safety, lower grade AEs (grades 1 and 2) can be underreported yet still affect tolerability. 46 Second, CTCAE severity grades of symptomatic AEs such as fatigue, pain, nausea, and shortness of breath may not fully correspond to the frequency, severity, or interference scores obtained via direct patient self‐report with PROs. 32 , 47 Clinician AE grading also generally occurs in association with clinic visits, and thus AEs that occur between visits may be missed. As a result of these gaps, we may lack a full understanding of the severity, chronicity, and impact of AEs in a clinical trial. 48 , 49 This incomplete picture of toxicities hinders the drug development process, and at the individual patient level may limit optimal supportive care to mitigate toxicities and improve tolerability. The systematic assessment of symptomatic AEs via PROs provides additional information about tolerability that is complementary to the assessments captured by clinicians using the CTCAE. The inclusion of patient self‐report in the assessment of AEs also has the potential to improve patient satisfaction and communication with their clinicians and to foster early detection of potentially serious AEs. 50
To support the inclusion of the patient experience of adverse treatment effects in evaluations of cancer treatment tolerability and to complement the CTCAE, the National Cancer Institute developed and rigorously validated the PRO‐CTCAE measurement system. 51 , 52 , 53 , 54 , 55 , 56 The PRO‐CTCAE is designed to profile the symptomatic AEs of cancer treatment, and has an item structure that allows the ascertainment of the full range of patient‐reportable treatment AEs, including AEs that are not typically represented on other symptom inventories, such as nail dystrophy, photosensitivity, bruising, chills, and fecal incontinence. 57 , 58 As with all AE reporting, capturing both expected and unexpected events is critically important. A set of relevant AEs for routine surveillance by clinicians using the CTCAE should be identified in the Comprehensive Adverse Event and Potential Risks (CAEPR) or AE‐monitoring section of the study protocol; other AEs that arise are graded as unexpected events. Similarly, the symptomatic AEs selected should be those likely to be associated with the regimen under study on the basis of the mechanism of action and early clinical data, 32 or symptoms that might be expected to be present at baseline due to prior cancer treatments and/or comorbid conditions. 59 Symptomatic toxicities selected for surveillance should match the CAEPR, and in a multiarm trial all participants should report on the same PRO‐CTCAE items across the different trial arms to reduce reporting bias. Additionally, the capture of unsolicited symptomatic AEs via a write‐in or verbatim text feature helps to minimize possible biases in item selection, and may identify early signals of previously unrecognized AEs. 60 , 61
The assessment schedule and intervals for symptomatic AE assessment with PROs must be appropriate for the regimen, patient population, and expected trajectory of AE onset and duration. Frequency and timing of patient reporting may vary on the basis of the specific trial design; however, there is evidence that sparse assessment intervals, particularly when coupled with a 24‐h recall period, may result in inaccuracies in symptomatic AE detection. 62 , 63 In all trials, a baseline assessment is essential to evaluate the prevalence and severity of symptoms before the study‐specific intervention is administered. Baseline values have interpretive value in AE attribution by examining group imbalance in multiarm trials, by gauging exacerbation in established toxicities caused by prior therapies, and as a covariate in longitudinal models. In some trial contexts, symptomatic AEs may also be captured via PROs after treatment discontinuation. 64 The study design and analysis plan should incorporate published guidelines for studies that include a PRO end point. 65 , 66
PRO‐CTCAE scores are intended for the descriptive reporting of symptomatic AEs and their direct effects on functioning in daily life. Clinician grading with the CTCAE remains the basis for determining protocol eligibility, dose modifications or interruptions, treatment discontinuation, identification of dose‐limiting AEs, and safety reporting to regulatory agencies. 67 The integration of both clinician‐ and patient‐reported measures supports a comprehensive understanding of treatment tolerability.
Summary indicators of treatment tolerability
Although a precise and accurate profiling of AEs via the CTCAE and PRO‐CTCAE is essential, other indicators can quantitatively summarize the extent and clinical impact of AEs, and thereby contribute additional insights into treatment tolerability. Such summary indicators include dose preservation, adherence and persistence, 68 health services utilization, self‐reported measures of HRQOL and physical functioning, 69 , 70 single‐item measures of overall side effect burden, 71 performance‐based and instrumented measures such as the 6‐min walk, 72 and actigraphy. 73
Hospitalizations, unscheduled health care visits, the inability to receive the full dose and duration of a planned treatment, elective treatment discontinuation, and nonadherence are all indirect indicators of treatment tolerability. 14 , 36 , 74 However, the interpretation of these metrics as indicators of tolerability also requires that investigators address confounding introduced by chronic conditions 75 , 76 and the side effects of noncancer medications. 77 Measures of HRQOL and well‐being such as the Functional Assessment of Cancer Therapy–General (FACT‐G), European Organisation for Research and Treatment of Cancer (EORTC) Core Quality of Life Questionnaire C30, and self‐reported health status, physical function, and activities of daily living 38 are particularly helpful in gauging the cumulative and whole‐person impact of multiple symptomatic AEs. Measures of HRQOL used in conjunction with the PRO‐CTCAE may yield additional insights into the impact of AEs, especially those that are low grade or may exert differential impacts at the individual level among those with organ system compromise, limited functional reserve, multimorbidity, and unmet social needs. Patient‐reported physical function may be a particularly important summary indicator of treatment tolerability, particularly in older adults. 78 , 79
There is evidence that single‐item measures of overall side effect burden—such as FACT GP5 (“I am bothered by the side effects of treatment”) and EORTC Q168 (“To what extent have you been troubled with side effects from your treatment?”)—and indicators of the patient’s willingness to stay on treatment even while enduring side effects—such as the Patient Acceptable Symptom State 80 —are valid and reliable, and may have predictive value and utility for stratification and subgroup analyses. 81 , 82 , 83 , 84 Strengths of single‐item summary measures of side effect burden include their brevity, face validity, and ease of interpretation. However, as with many single‐item measures, they may demonstrate insensitivity to between‐person differences, ceiling effects, and nonresponsiveness to change, particularly in the setting of missing assessments. 85
Whole‐person factors influencing treatment tolerability
There is evidence that patient preferences, 26 health status and functional reserve, 79 , 86 experiences of care, 87 and social determinants of health such as social support, 88 poverty, 89 or rurality 90 may directly or indirectly influence experiences of tolerability. For example, two patients who have been prescribed the same regimen may have differing experiences of tolerability on the basis of their specific circumstances. This might be because one patient has pain from a spine injury that occurred 10 years ago, has a stressful job with a supervisor who does not support the need for periodic absence to attend clinic appointments, and is a sole provider for two young children with little community support, whereas another patient has no chronic conditions, is not currently working, and has a strong support system and abundant financial resources. Although both patients may experience a similar AE profile, their overall experience of tolerability may be quite different. There is empirical evidence of individual differences in tolerability based on socioeconomic, environmental, cultural, psychosocial, and behavioral factors. 87 , 91 , 92 , 93 , 94 , 95 , 96 Evidence also points to several whole‐person factors that place patients at risk for more frequent AEs, 92 greater symptom burden, 97 , 98 and inferiorities in HRQOL during and after cancer treatment. 99 , 100 Financial hardship, deficiencies in patient–provider communication and rapport, low health literacy, comorbid conditions, and barriers to accessing needed care can all influence tolerability and adversely affect effective self‐management. 101 , 102 These whole‐person factors can be targeted with proactive, multilevel, and interdisciplinary interventions designed to improve treatment tolerability, preserve dose density, and optimize recovery and overall well‐being. 103 , 104 , 105 , 106
TOLERABILITY MEASUREMENT CONSIDERATIONS IN CANCER TREATMENT TRIAL DESIGN
A thoroughly characterized AE profile is especially important in phase 1 trials, where AEs are the primary end point and sample sizes may be small. It is similarly important in phase 2 trials, in which the goal is to gauge preliminary signals about relative benefits versus risk. In early‐phase trials, the inclusion of both the PRO‐CTCAE and summary indicators of tolerability sets the stage for more focused and efficient tolerability evaluations in later phase trials, and may help to define the supportive care that should be included as part of the standard of care in subsequent trials. 107 The PRO‐CTCAE and summary indicators of tolerability may also have interpretive value in the context of dose‐optimizing study designs 108 , 109 by helping to define candidate dose ranges/schedules to be studied in the expansion cohort and confirming improved tolerability in dose comparison designs. 18 , 110 , 111 , 112
In later phases of therapy development, the emphasis in understanding tolerability shifts from defining the MTD or exploring the AE profile associated with a range of potentially efficacious dose levels to evaluating combination or sequential multiagent regimens 113 or comparing two or more effective treatment approaches in larger samples. 114 Patient‐reported AEs supplemented by summary indicators of tolerability may also provide a sensitive secondary end point in comparative effectiveness studies or studies of new approaches to supportive cancer care where toxicity reduction and improving tolerability is a goal of treatment. 95
Precise characterization of the profiles of clinician‐ and patient‐reported AEs, including low‐grade AEs, may be especially important in prevention, adjuvant, and maintenance therapy trials. Moreover, with many of these therapies, patients’ adherence and persistence over time is fundamental to treatment effectiveness. As such, comparatively mild toxicities, especially experienced over an extended period, may contribute to nonadherence or lead patients to request early treatment discontinuation. 68
Systematically characterizing baseline symptoms at study entry complements baseline assessments of vital organ functioning, and can result in a more accurate and efficient attribution of symptomatic AEs 115 and inform prediction of favorable 116 and unfavorable outcomes. 117 The trajectory of symptomatic AEs may also provide additional information about the duration 118 and chronicity 119 , 120 of toxicities. Symptomatic AEs may also be important covariates in analyses to understand change over time (both improvements and worsening) in HRQOL outcomes. Identifying subgroups experiencing distinct toxicity profiles on the basis of the awareness that whole‐person factors influence treatment tolerability can also be informative for hypothesis generation about the mechanisms underlying specific AEs. This knowledge can be integrated with biomarker and omic profiles to improve AE risk prediction. 121 Patient self‐report of symptomatic AEs may be particularly useful in identifying subgroups who are at greatest risk for adverse treatment outcomes, and would therefore benefit from tailored supportive care management.
Despite the importance of understanding cancer treatment tolerability, challenges in capturing, analyzing, and reporting symptomatic AE data have been documented. 122 , 123 A range of resources and methodological tools exists to support early‐phase trial study teams in implementing best practices during trial design, 124 protocol development, 125 PRO data collection and study implementation, 65 , 126 , 127 and analysis and interpretation. 128 However, investments are needed to strengthen trials’ research infrastructure, 66 , 127 scale up overall system capacity for the collection of PRO data and address implementation barriers and facilitators, 129 , 130 support method development, 131 and create and sustain partnerships among trialists, PRO researchers, clinicians, regulators, and patient advocates, 127 , 132 particularly in the context of early‐phase clinical trials. Research is ongoing to evaluate alternative methods for AE reporting, including electronic PRO collection and alerts, 127 , 133 automated extraction from the electronic health record, 134 , 135 implementation of triggers and alerts to improve data quality, 49 novel tolerability indicators that incorporate patient‐generated health data, 136 and analytic methods to understand the longitudinal profile and cumulative impact of nonsymptomatic and symptomatic AEs rather than only the highest AE grade. 137 , 138 Efforts are also ongoing to examine interventions to improve health care professionals’ practices in reporting AEs. 139
Conclusions
In this commentary, we have outlined the key concepts to be considered when evaluating cancer treatment tolerability, describing direct and indirect indicators of tolerability and emphasizing the important role of PRO measures in a wide range of tolerability‐focused study designs. We have also highlighted the ways in which whole‐person factors meaningfully influence the generalizability of conclusions about tolerability, particularly with respect to understanding the experiences of distinct subgroups or the factors that can be addressed to improve the tolerability of cancer treatments.
Strengthening the measurement and interpretation of tolerability enhances our understanding of patient risk, benefit, and outcomes in both cancer treatment trials and comparative effectiveness research conducted in real‐world settings. A more precise and comprehensive measurement of tolerability is essential for shared decision‐making, and can inform the provision of tailored supportive care. To address the need to better integrate and analyze different measures of tolerability, the National Cancer Institute funded the Cancer Treatment Tolerability Consortium. 140 , 141 Ultimately, these insights can inform future clinical trials and advance the delivery of individualized cancer care.
AUTHOR CONTRIBUTIONS
Sandra A. Mitchell: Conceptualization; writing—original draft; and writing—review and editing. Rachel D. Altshuler: Conceptualization; writing—original draft; and writing—review and editing. Diane C. St. Germain: Conceptualization; writing—original draft; and writing—review and editing. Brennan Parmelee Streck: Writing—original draft; conceptualization; and writing—review and editing. Alice P. Chen: Conceptualization; writing—original draft; and writing—review and editing. Lori M. Minasian: Conceptualization; writing—original draft; and writing—review and editing.
CONFLICT OF INTEREST STATEMENT
Brennan Parmelee Streck reports holding stock with Gilead Sciences, Johnson & Johnson, and Pfizer. The other authors declare no conflicts of interest.
ACKNOWLEDGMENTS
We thank the ICF International for their support in editing the manuscript. We also acknowledge with gratitude Dr. Ashley Wilder Smith, Ms. Kate Castro, and Ms. Diane Ng, who provided helpful comments on an earlier version of the manuscript. This material should not be interpreted as representing the viewpoint of the US Department of Health and Human Services, the National Institutes of Health, or the National Cancer Institute.
Mitchell SA, Altshuler RD, St. Germain DC, Streck BP, Chen AP, Minasian LM. Measuring the multidimensional aspects of tolerability. Cancer. 2025;e70085. doi: 10.1002/cncr.70085
We thank the members of the Tolerability Consortium and its working groups for their contributions. We also thank ICF International for supporting the Tolerability Consortium activities, including this supplement.
The findings and conclusions in this Supplement are those of the authors and do not necessarily reflect the official position of the American Cancer Society, John Wiley & Sons, Inc., or the opinions of the journal editors.
The first two authors contributed equally to this article.
DATA AVAILABILITY STATEMENT
The original contributions are included in the article; further inquiries can be directed to the corresponding author.
REFERENCES
- 1. Delgado A, Guddati AK. Clinical endpoints in oncology—a primer. Am J Cancer Res. 2021;11(4):1121‐1131. [PMC free article] [PubMed] [Google Scholar]
- 2. Lasala R, Santoleri F. Association between adherence to oral therapies in cancer patients and clinical outcome: a systematic review of the literature. Br J Clin Pharmacol. 2022;88(5):1999‐2018. doi: 10.1111/bcp.15147 [DOI] [PubMed] [Google Scholar]
- 3. Sandström N, Leppälä E, Jekunen A, Johansson M, Andersén H. Role of patient characteristics in adherence to first‐line treatment guidelines in breast, lung and prostate cancer: insights from the Nordic healthcare system. BMJ Open. 2024;14(4):e084689. doi: 10.1136/bmjopen-2024-084689 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Lee EM, Jiménez‐Fonseca P, Galán‐Moral R, et al. Toxicities and quality of life during cancer treatment in advanced solid tumors. Curr Oncol. 2023;30(10):9205‐9216. doi: 10.3390/curroncol30100665 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Nekhlyudov L, Campbell GB, Schmitz KH, et al. Cancer‐related impairments and functional limitations among long‐term cancer survivors: gaps and opportunities for clinical practice. Cancer. 2022;128(2):222‐229. doi: 10.1002/cncr.33913 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Culakova E, Mohamed M, Flannery M, et al. Relationships between patient‐reported and clinician‐rated toxicities and daily functioning in older adults with advanced cancer undergoing systemic therapy. Cancer. 2025;131(4):e35766. doi: 10.1002/cncr.35766 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Aronson JK. When I use a word … medical definitions: adverse events, effects, and reactions. BMJ. 2023;381:917. doi: 10.1136/bmj.p917 [DOI] [PubMed] [Google Scholar]
- 8. Broadening the Definition of Tolerability in Cancer Clinical Trials to Better Measure the Patient Experience. Friends of Cancer Research . Accessed March 1, 2025. https://friendsofcancerresearch.org/wp‐content/uploads/Comparative‐Tolerability‐Whitepaper_FINAL.pdf
- 9. NCI Dictionary of Cancer Terms. National Cancer Institute . Accessed June 13, 2025. https://www.cancer.gov/publications/dictionaries/cancer‐terms
- 10. Peipert JD, Smith ML. Reconsidering tolerability of cancer treatments: opportunities to focus on the patient. Support Care Cancer. 2022;30(5):3661‐3663. doi: 10.1007/s00520-021-06700-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Ray SD, Black AT, Shah SK, Stohs SJ. ADRs, ADEs and SEDs: a bird's eye view. In: Ray SD, ed. Side Effects of Drugs Annual. Elsevier; 2018:xxvii‐xliv. [Google Scholar]
- 12. Shader RI. Safety versus tolerability. Clin Ther. 2018;40(5):672‐673. doi: 10.1016/j.clinthera.2018.04.003 [DOI] [PubMed] [Google Scholar]
- 13. Singh S, Loke YK. Drug safety assessment in clinical trials: methodological challenges and opportunities. Trials. 2012;13(1):138. doi: 10.1186/1745-6215-13-138 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Stanulović V, Hodolic M, Mitsikostas DD, Papadopoulos D. Drug tolerability: how much ambiguity can be tolerated? A systematic review of the assessment of tolerability in clinical studies. Br J Clin Pharmacol. 2022;88(2):551‐565. doi: 10.1111/bcp.15016 [DOI] [PubMed] [Google Scholar]
- 15. Manavi MA, Fathian Nasab MH, Mohammad Jafari R, Dehpour AR. Mechanisms underlying dose‐limiting toxicities of conventional chemotherapeutic agents. J Chemother. 2024;36(8):623‐653. doi: 10.1080/1120009x.2023.2300217 [DOI] [PubMed] [Google Scholar]
- 16. Liao JJZ, Asatiani E, Liu Q, Hou K. Three steps toward dose optimization for oncology dose finding. Contemp Clin Trials Commun. 2024;40:101329. doi: 10.1016/j.conctc.2024.101329 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Li M, Guo Z, Qiu Y. UNITED: a unified transparent and efficient phase I/II trial design for dose optimization accounting for ordinal graded, continuous and mixed toxicity and efficacy endpoints. Stat Med. 2025;44(10‐12):e70098. doi: 10.1002/sim.70098 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Alger E, Lee SM, Cheung YK, Yap C. U‐PRO‐CRM: designing patient‐centred dose‐finding trials with patient‐reported outcomes. ESMO Open. 2024;9(7):103626. doi: 10.1016/j.esmoop.2024.103626 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Alger E, Van Zyl M, Aiyegbusi OL, et al. Patient and public involvement and engagement in the development of innovative patient‐centric early phase dose‐finding trial designs. Res Involv Engagem. 2024;10(1):63. doi: 10.1186/s40900-024-00599-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Byrom B, Everhart A, Cordero P, Garratt C, Meyer T. Leveraging patient‐reported outcome measures for optimal dose selection in early phase cancer trials. JMIR Cancer. 2025;11:e64611. doi: 10.2196/64611 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Prajapati S, Tao R, Feldman SR. Drug tolerability versus drug safety. J Dermatol Treat. 2023;34(1):2202786. doi: 10.1080/09546634.2023.2202786 [DOI] [PubMed] [Google Scholar]
- 22. Hansson SO. Safety is an inherently inconsistent concept. Saf Sci. 2012;50(7):1522‐1527. doi: 10.1016/j.ssci.2012.03.003 [DOI] [Google Scholar]
- 23. Cohen A. Should we tolerate tolerability as an objective in early drug development? Br J Clin Pharmacol. 2007;64(3):249‐252. doi: 10.1111/j.1365-2125.2007.03023.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Pignatti F, Wilking U, Postmus D, Wilking N, Delgado J, Bergh J. The value of anticancer drugs—a regulatory view. Nat Rev Clin Oncol. 2022;19(3):207‐215. doi: 10.1038/s41571-021-00584-z [DOI] [PubMed] [Google Scholar]
- 25. Loeser A, Kim JS, Peppercorn J, et al. The right dose: results of a patient advocate‐led survey of individuals with metastatic breast cancer regarding treatment‐related side effects and views about dosage assessment to optimize quality of life. JCO Oncol Pract. 2024;20(7):972‐983. doi: 10.1200/op.23.00539 [DOI] [PubMed] [Google Scholar]
- 26. Brédart A, Bodson S, Le Tourneau C, et al. Patients' perceived tolerance of side effects in phase I cancer clinical trials: a qualitative study. Eur J Cancer Care. 2017;26(6):e12596. doi: 10.1111/ecc.12596 [DOI] [PubMed] [Google Scholar]
- 27. Dahabreh IJ, Hayward R, Kent DM. Using group data to treat individuals: understanding heterogeneous treatment effects in the age of precision medicine and patient‐centred evidence. Int J Epidemiol. 2016;45(6):2184‐2193. doi: 10.1093/ije/dyw125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Shickh S, Leventakos K, Lewis MA, Bombard Y, Montori VM. Shared decision making in the care of patients with cancer. Am Soc Clin Oncol Educ Book. 2023;43:e389516. doi: 10.1200/edbk_389516 [DOI] [PubMed] [Google Scholar]
- 29. Rothausen CS, Dieperink KB, Ruhlmann CH, Pappot H, Tolstrup LK. Patient and caregiver education to support self‐efficacy and self‐management during immunotherapy—an integrative review. Psychooncology. 2025;34(3):e70100. doi: 10.1002/pon.70100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Lopes GS, Tournigand C, Olswold CL, et al. Adverse event load, onset, and maximum grade: a novel method of reporting adverse events in cancer clinical trials. Clin Trials. 2021;18(1):51‐60. doi: 10.1177/1740774520959313 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Lee SM, Hershman DL, Miao J, Zhong X, Unger JM, Cheung YKK. Estimating global treatment toxicity burden from adverse‐event data. Cancer. 2018;124(4):858‐864. doi: 10.1002/cncr.31107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Minasian LM, O'Mara A, Mitchell SA. Clinician and patient reporting of symptomatic adverse events in cancer clinical trials: using CTCAE and PRO‐CTCAE® to provide two distinct and complementary perspectives. Patient Relat Outcome Meas. 2022;13:249‐258. doi: 10.2147/prom.S256567 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Kluetz PG, Chingos DT, Basch EM, Mitchell SA. Patient‐reported outcomes in cancer clinical trials: measuring symptomatic adverse events with the National Cancer Institute's Patient‐Reported Outcomes version of the Common Terminology Criteria for Adverse Events (PRO‐CTCAE). Am Soc Clin Oncol Educ Book. 2016;35:67‐73. doi: 10.1200/EDBK_159514 [DOI] [PubMed] [Google Scholar]
- 34. Sotelo‐Rodríguez DC, Ruíz‐Patiño A, Ricaurte L, Arrieta O, Zatarain‐Barrón ZL, Cardona AF. Challenges and shifting paradigms in clinical trials in oncology: the case for immunological and targeted therapies. Ecancermedicalscience. 2019;13:936. doi: 10.3332/ecancer.2019.936 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Thanarajasingam G, Minasian LM, Bhatnagar V, et al. Reaching beyond maximum grade: progress and future directions for modernising the assessment and reporting of adverse events in haematological malignancies. Lancet Haematol. 2022;9(5):e374‐e384. doi: 10.1016/s2352-3026(22)00045-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Palwe V, Patil R, Pandit P, Nagarkar R. Factors influencing non‐adherence to radiotherapy: a retrospective audit of 1,548 patients from a tertiary cancer centre. J Radiother Pract. 2020;19(4):359‐364. doi: 10.1017/S1460396919000840 [DOI] [Google Scholar]
- 37. Hershman DL, Neugut AI, Moseley A, et al. Patient‐reported outcomes and long‐term nonadherence to aromatase inhibitors. J Natl Cancer Inst. 2021;113(8):989‐996. doi: 10.1093/jnci/djab022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Flannery MA, Culakova E, Canin BE, Peppone L, Ramsdale E, Mohile SG. Understanding treatment tolerability in older adults with cancer. J Clin Oncol. 2021;39(19):2150‐2163. doi: 10.1200/jco.21.00195 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Retzer A, Aiyegbusi OL, Rowe A, et al. The value of patient‐reported outcomes in early‐phase clinical trials. Nat Med. 2022;28(1):18‐20. doi: 10.1038/s41591-021-01648-4 [DOI] [PubMed] [Google Scholar]
- 40. Ijzerman MJ, Manca A, Keizer J, Ramsey SD. Implementation of comparative effectiveness research in personalized medicine applications in oncology: current and future perspectives. Comp Eff Res (Auckl). 2015;5:65‐72. doi: 10.2147/cer.s92212 [DOI] [Google Scholar]
- 41. Schnipper LE, Davidson NE, Wollins DS, et al. Updating the American Society of Clinical Oncology value framework: revisions and reflections in response to comments received. J Clin Oncol. 2016;34(24):2925‐2934. doi: 10.1200/jco.2016.68.2518 [DOI] [PubMed] [Google Scholar]
- 42. Cherny NI, Sullivan R, Dafni U, et al. ESMO—Magnitude of Clinical Benefit Scale v.1.0 questions and answers. ESMO Open. 2016;1(5):e000100. doi: 10.1136/esmoopen-2016-000100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Unger JM, Vaidya R, Albain KS, et al. Sex differences in risk of severe adverse events in patients receiving immunotherapy, targeted therapy, or chemotherapy in cancer clinical trials. J Clin Oncol. 2022;40(13):1474‐1486. doi: 10.1200/jco.21.02377 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Food and Drug Administration . Core Patient‐Reported Outcomes in Cancer Clinical Trials: Guidance for Industry. Food and Drug Administration; 2024. [Google Scholar]
- 45. National Cancer Institute . Common Terminology Criteria for Adverse Events (CTCAE) Version 5.0. National Cancer Institute. Accessed September 10, 2025. https://dctd.cancer.gov/research/ctep‐trials/for‐sites/adverse‐events [Google Scholar]
- 46. O'Connell NS, Zhao F, Lee JW, et al. Importance of low‐ and moderate‐grade adverse events in patients' treatment experience and treatment discontinuation: an analysis of the E1912 trial. J Clin Oncol. 2024;42(3):266‐272. doi: 10.1200/jco.23.00377 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Atkinson TM, Dueck AC, Satele DV, et al. Clinician vs patient reporting of baseline and postbaseline symptoms for adverse event assessment in cancer clinical trials. JAMA Oncol. 2020;6(3):437‐439. doi: 10.1001/jamaoncol.2019.5566 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Edgerly M, Fojo T. Is there room for improvement in adverse event reporting in the era of targeted therapies? J Natl Cancer Inst. 2008;100(4):240‐242. doi: 10.1093/jnci/djm324 [DOI] [PubMed] [Google Scholar]
- 49. Miller TP, Aplenc R. Evolution of hematology clinical trial adverse event reporting to improve care delivery. Curr Hematol Malig Rep. 2021;16(2):126‐131. doi: 10.1007/s11899-021-00627-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Kennedy F, Shearsmith L, Ayres M, et al. Online monitoring of patient self‐reported adverse events in early phase clinical trials: views from patients, clinicians, and trial staff. Clin Trials. 2021;18(2):168‐179. doi: 10.1177/1740774520972125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Atkinson TM, Hay JL, Dueck AC, et al. What do “none,” “mild,” “moderate,” “severe,” and “very severe” mean to patients with cancer? Content validity of PRO‐CTCAE™ response scales. J Pain Symptom Manage. 2018;55(3):e3‐e6. doi: 10.1016/j.jpainsymman.2017.10.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Bennett AV, Dueck AC, Mitchell SA, et al. Mode equivalence and acceptability of tablet computer‐, interactive voice response system‐, and paper‐based administration of the U.S. National Cancer Institute's Patient‐Reported Outcomes version of the Common Terminology Criteria for Adverse Events (PRO‐CTCAE). Health Qual Life Outcomes. 2016;14:24. doi: 10.1186/s12955-016-0426-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Dueck AC, Mendoza TR, Mitchell SA, et al. Validity and reliability of the US National Cancer Institute's Patient‐Reported Outcomes version of the Common Terminology Criteria for Adverse Events (PRO‐CTCAE). JAMA Oncol. 2015;1(8):1051‐1059. doi: 10.1001/jamaoncol.2015.2639 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Hay JL, Atkinson TM, Reeve BB, et al. Cognitive interviewing of the US National Cancer Institute's Patient‐Reported Outcomes version of the Common Terminology Criteria for Adverse Events (PRO‐CTCAE). Qual Life Res. 2014;23(1):257‐269. doi: 10.1007/s11136-013-0470-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Mendoza TR, Dueck AC, Bennett AV, et al. Evaluation of different recall periods for the US National Cancer Institute's PRO‐CTCAE. Clin Trials. 2017;14(3):255‐263. doi: 10.1177/1740774517698645 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Lee MK, Basch E, Mitchell SA, et al. Reliability and validity of PRO‐CTCAE® daily reporting with a 24‐hour recall period. Qual Life Res. 2023;32(7):2047‐2058. doi: 10.1007/s11136-023-03374-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Patient‐Reported Outcomes Version of the Common Terminology Criteria for Adverse Events (PRO‐CTCAE®). National Cancer Institute. Accessed January 26, 2025. https://healthcaredelivery.cancer.gov/pro‐ctcae/ [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Basch E, Reeve BB, Mitchell SA, et al. 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. 2014;106(9):dju244. doi: 10.1093/jnci/dju244 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Feldman E, Pos FJ, Smeenk RJ, et al. Selecting a PRO‐CTCAE‐based subset for patient‐reported symptom monitoring in prostate cancer patients: a modified Delphi procedure. ESMO Open. 2023;8(1):100775. doi: 10.1016/j.esmoop.2022.100775 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Friese CR, Fauer AJ, Kuisell C, et al. Patient‐reported outcomes collected in ambulatory oncology practices: feasibility, patterns, and correlates. Health Serv Res. 2020;55(6):966‐972. doi: 10.1111/1475-6773.13574 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Chung AE, Shoenbill K, Mitchell SA, et al. Patient free text reporting of symptomatic adverse events in cancer clinical research using the National Cancer Institute's Patient‐Reported Outcomes version of the Common Terminology Criteria for Adverse Events (PRO‐CTCAE). J Am Med Inform Assoc. 2019;26(4):276‐285. doi: 10.1093/jamia/ocy169 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. King‐Kallimanis BL, Bhatnagar V, Horodniceanu EG, Chen TY, Kluetz PG. Timing is everything: the importance of patient‐reported outcome assessment frequency when characterizing symptomatic adverse events. Clin Trials. 2022;19(3):267‐273. doi: 10.1177/17407745221093935 [DOI] [PubMed] [Google Scholar]
- 63. Paudel R, Enzinger AC, Uno H, et al. Effects of a change in recall period on reporting severe symptoms: an analysis of a pragmatic multisite trial. J Natl Cancer Inst. 2024;116(7):1137‐1144. doi: 10.1093/jnci/djae049 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. King‐Kallimanis BL, Calvert M, Cella D, et al. Perspectives on patient‐reported outcome data after treatment discontinuation in cancer clinical trials. Value Health. 2023;26(10):1543‐1548. doi: 10.1016/j.jval.2023.06.019 [DOI] [PubMed] [Google Scholar]
- 65. Cruz Rivera S, Aiyegbusi OL, Ives J, et al. Ethical considerations for the inclusion of patient‐reported outcomes in clinical research: the PRO ethics guidelines. JAMA. 2022;327(19):1910‐1919. doi: 10.1001/jama.2022.6421 [DOI] [PubMed] [Google Scholar]
- 66. Mc Laughlin AM, Schmulenson E, Teplytska O, et al. Developing a nationwide infrastructure for therapeutic drug monitoring of targeted oral anticancer drugs: the ON‐TARGET study protocol. Cancers. 2021;13(24):6281. doi: 10.3390/cancers13246281 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Kim J, Singh H, Ayalew K, et al. Use of PRO measures to inform tolerability in oncology trials: implications for clinical review, IND safety reporting, and clinical site inspections. Clin Cancer Res. 2018;24(8):1780‐1784. doi: 10.1158/1078-0432.Ccr-17-2555 [DOI] [PubMed] [Google Scholar]
- 68. Peddie N, Agnew S, Crawford M, Dixon D, MacPherson I, Fleming L. The impact of medication side effects on adherence and persistence to hormone therapy in breast cancer survivors: a qualitative systematic review and thematic synthesis. Breast. 2021;58:147‐159. doi: 10.1016/j.breast.2021.05.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Cocks K, Wells JR, Johnson C, et al. Content validity of the EORTC quality of life questionnaire QLQ‐C30 for use in cancer. Eur J Cancer. 2023;178:128‐138. doi: 10.1016/j.ejca.2022.10.026 [DOI] [PubMed] [Google Scholar]
- 70. Pandya C, Magnuson A, Flannery M, et al. Association between symptom burden and physical function in older patients with cancer. J Am Geriatr Soc. 2019;67(5):998‐1004. doi: 10.1111/jgs.15864 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Kluetz PG, Kanapuru B, Lemery S, et al. Informing the tolerability of cancer treatments using patient‐reported outcome measures: summary of an FDA and Critical Path Institute Workshop. Value Health. 2018;21(6):742‐747. doi: 10.1016/j.jval.2017.09.009 [DOI] [PubMed] [Google Scholar]
- 72. Schmidt K, Vogt L, Thiel C, Jager E, Banzer W. Validity of the six‐minute walk test in cancer patients. Int J Sports Med. 2013;34(7):631‐636. doi: 10.1055/s-0032-1323746 [DOI] [PubMed] [Google Scholar]
- 73. Pirl WF, Fujisawa D, Stagl J, et al. Actigraphy as an objective measure of performance status in patients with advanced cancer. J Clin Oncol. 2015;33(suppl 29):62. doi: 10.1200/jco.2015.33.29_suppl.62 [DOI] [Google Scholar]
- 74. Sikorskii A, Given CW, Chang S, Tam S, Movsas B, Given B. Patient reported outcomes and unscheduled health services use during oral anti‐cancer treatment. J Pain Symptom Manage. 2023;65(2):e115‐e121. doi: 10.1016/j.jpainsymman.2022.10.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Chang WH, Lai AG. Pan‐cancer analyses of the associations between 109 pre‐existing conditions and cancer treatment patterns across 19 adult cancers. Sci Rep. 2024;14(1):464. doi: 10.1038/s41598-024-51161-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Chen W, Altshuler RD, Daschner P, et al. Older adults with cancer and common comorbidities—challenges and opportunities in improving their cancer treatment outcomes. J Natl Cancer Inst. 2024;116(11):1730‐1738. doi: 10.1093/jnci/djae163 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. George M, Smith A, Sabesan S, Ranmuthugala G. Physical comorbidities and their relationship with cancer treatment and its outcomes in older adult populations: systematic review. JMIR Cancer. 2021;7(4):e26425. doi: 10.2196/26425 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Xu H, Mohamed M, Flannery M, et al. An unsupervised machine learning approach to evaluating the association of symptom clusters with adverse outcomes among older adults with advanced cancer: a secondary analysis of a randomized clinical trial. JAMA Netw Open. 2023;6(3):e234198. doi: 10.1001/jamanetworkopen.2023.4198 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Wang Y, Zhang Y, Liang X, Liu J, Zhao Y, Su Q. The impact of frailty on chemotherapy intolerance in patients with cervical cancer: a longitudinal study. Eur J Oncol Nurs. 2025;74:102725. doi: 10.1016/j.ejon.2024.102725 [DOI] [PubMed] [Google Scholar]
- 80. Krueger E, Secinti E, Wu W, et al. Measurement of patients' acceptable symptom levels and priorities for symptom improvement in advanced lung cancer. Support Care Cancer. 2021;29(10):5895‐5904. doi: 10.1007/s00520-021-06159-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Peipert JD, Shaunfield S, Kaiser K, et al. How do patients interpret and respond to a single‐item global indicator of cancer treatment tolerability? Support Care Cancer. 2022;31(1):37. doi: 10.1007/s00520-022-07484-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Griffiths P, Peipert JD, Leith A, et al. Validity of a single‐item indicator of treatment side effect bother in a diverse sample of cancer patients. Support Care Cancer. 2022;30(4):3613‐3623. doi: 10.1007/s00520-022-06802-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Roydhouse JK, King‐Kallimanis BL, Roy P, et al. Exploration of baseline patient‐reported side effect bother from cancer therapy. Clin Trials. 2020;17(3):332‐337. doi: 10.1177/1740774520910389 [DOI] [PubMed] [Google Scholar]
- 84. Regnault A, Bunod L, Loubert A, et al. Assessing tolerability with the Functional Assessment of Cancer Therapy item GP5: psychometric evidence from LIBRETTO‐531, a phase 3 trial of selpercatinib in medullary thyroid cancer. J Patient Rep Outcomes. 2024;8(1):149. doi: 10.1186/s41687-024-00823-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Arizmendi C, Zhu Y, Khan M, et al. The FACT‐GP5 as a global tolerability measure: responsiveness and robustness to missing assessments. Qual Life Res. 2024;33(10):2869‐2880. doi: 10.1007/s11136-024-03740-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Wildiers H, de Glas NA. Anticancer drugs are not well tolerated in all older patients with cancer. Lancet Healthy Longev. 2020;1(1):e43‐e47. doi: 10.1016/s2666-7568(20)30001-5 [DOI] [PubMed] [Google Scholar]
- 87. Rosenzweig MQ, Mazanec SR. Racial differences in breast cancer therapeutic toxicity: implications for practice. Cancer Epidemiol Biomarkers Prev. 2023;32(2):157‐158. doi: 10.1158/1055-9965.Epi-22-1111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Shahrokni A, Sun CL, Tew WP, et al. The association between social support and chemotherapy‐related toxicity in older patients with cancer. J Geriatr Oncol. 2020;11(2):274‐279. doi: 10.1016/j.jgo.2019.08.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Li S, He Y, Liu J, et al. An umbrella review of socioeconomic status and cancer. Nat Commun. 2024;15(1):9993. doi: 10.1038/s41467-024-54444-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Munhoz R, Sabesan S, Thota R, Merrill J, Hensold JO. Revolutionizing rural oncology: innovative models and global perspectives. Am Soc Clin Oncol Educ Book. 2024;44(3):e432078. doi: 10.1200/edbk_432078 [DOI] [PubMed] [Google Scholar]
- 91. Hutchings H, Behinaein P, Enofe N, et al. Association of social determinants with patient‐reported outcomes in patients with cancer. Cancers. 2024;16(5):1015. doi: 10.3390/cancers16051015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Bulls HW, Chang PH, Brownstein NC, et al. Patient‐reported symptom burden in routine oncology care: examining racial and ethnic disparities. Cancer Rep (Hoboken). 2022;5(3):e1478. doi: 10.1002/cnr2.1478 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Hu X, Walker MS, Stepanski E, et al. Racial differences in patient‐reported symptoms and adherence to adjuvant endocrine therapy among women with early‐stage, hormone receptor‐positive breast cancer. JAMA Netw Open. 2022;5(8):e2225485. doi: 10.1001/jamanetworkopen.2022.25485 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Obeng‐Gyasi S, Graham N, Kumar S, et al. Examining allostatic load, neighborhood socioeconomic status, symptom burden and mortality in multiple myeloma patients. Blood Cancer J. 2022;12(4):53. doi: 10.1038/s41408-022-00648-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Henry NL, Kim S, Hays RD, et al. Toxicity index, patient‐reported outcomes, and early discontinuation of endocrine therapy for breast cancer risk reduction in NRG Oncology/NSABP B‐35. J Clin Oncol. 2021;39(34):3800‐3812. doi: 10.1200/JCO.21.00910 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. You KL, Sereika SM, Bender CM, et al. Health‐related quality of life over chemotherapy course among individuals with early‐stage breast cancer: the association of social determinants of health and neighborhood socioeconomic disadvantage. Support Care Cancer. 2024;32(4):224. doi: 10.1007/s00520-024-08429-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Kwok W, Bhuvanakrishna T. The relationship between ethnicity and the pain experience of cancer patients: a systematic review. Indian J Palliat Care. 2014;20(3):194. doi: 10.4103/0973-1075.138391 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Cheung CK, Lee H, Levin NJ, et al. Disparities in cancer care among sexual and gender minority adolescent and young adult patients: a scoping review. Cancer Med. 2023;12(13):14674‐14693. doi: 10.1002/cam4.6090 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Rincon MA, Smith AW, Yu M, Kent EE. Trends in racial/ethnic disparity of health‐related quality of life in older adults with and without cancer (1998–2012). Cancer Epidemiol Biomarkers Prev. 2020;29(6):1188‐1195. doi: 10.1158/1055-9965.EPI-19-0819 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Boehmer U, Gereige J, Winter M, Ozonoff A. Cancer survivors’ access to care and quality of life: do sexual minorities fare worse than heterosexuals? Cancer. 2019;125(17):3079‐3085. doi: 10.1002/cncr.32151 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Dean LT, George M, Lee KT, Ashing K. Why individual‐level interventions are not enough: systems‐level determinants of oral anticancer medication adherence. Cancer. 2020;126(16):3606‐3612. doi: 10.1002/cncr.32946 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Wieder R, Adam N. Racial disparities in breast cancer treatments and adverse events in the SEER‐Medicare data. Cancers. 2023;15(17):4333. doi: 10.3390/cancers15174333 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Check DK, Chawla N, Kwan ML, et al. Understanding racial/ethnic differences in breast cancer‐related physical well‐being: the role of patient‐provider interactions. Breast Cancer Res Treat. 2018;170(3):593‐603. doi: 10.1007/s10549-018-4776-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Jacobs JM, Walsh EA, Park ER, et al. The patient's voice: adherence, symptoms, and distress related to adjuvant endocrine therapy after breast cancer. Int J Behav Med. 2020;27(6):687‐697. doi: 10.1007/s12529-020-09908-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Crowley F, Sheppard R, Lehrman S, et al. Optimizing care in early phase cancer trials: the role of palliative care. Cancer Treat Rev. 2024;128:102767. doi: 10.1016/j.ctrv.2024.102767 [DOI] [PubMed] [Google Scholar]
- 106. Sallis JF, Owen N. Ecological models of health behavior. In: Glanz K, Rimer BK, Viswanath KV, eds. Health Behavior: Theory, Research and Practice. 5th ed. Jossey‐Bass/Wiley; 2015:43‐64. [Google Scholar]
- 107. Neuman HB. Patient experience, adverse event reporting, and clinical trial design. J Clin Oncol. 2024;42(3):247‐249. doi: 10.1200/jco.23.01976 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Spreafico A, Hansen AR, Abdul Razak AR, Bedard PL, Siu LL. The future of clinical trial design in oncology. Cancer Discov. 2021;11(4):822‐837. doi: 10.1158/2159-8290.Cd-20-1301 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Araujo D, Greystoke A, Bates S, et al. Oncology phase I trial design and conduct: time for a change—MDICT guidelines 2022. Ann Oncol. 2023;34(1):48‐60. doi: 10.1016/j.annonc.2022.09.158 [DOI] [PubMed] [Google Scholar]
- 110. Xia H, Booth BP, Wang Y, et al. Use of patient‐reported outcomes (PRO) data to complement exposure‐response analysis in early clinical cancer drug development. J Patient Rep Outcomes. 2023;7(1):116. doi: 10.1186/s41687-023-00651-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Basch E, Dueck AC, Mitchell SA, et al. Patient‐reported outcomes during and after treatment for locally advanced rectal cancer in the PROSPECT trial (Alliance N1048). J Clin Oncol. 2023;41(21):3724‐3734. doi: 10.1200/JCO.23.00903 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Zhu J, Schroeder A, Frank S, et al. Oncology dose optimization: tailored approaches to different molecular classes. Clin Pharmacol Ther. 2025;118(1):74‐79. doi: 10.1002/cpt.3658 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Shin S, Moon J, Oum C, et al. Discontinuation risk from adverse events: immunotherapy alone vs. combined with chemotherapy: a systematic review and network meta‐analysis. BMC Cancer. 2024;24(1):152. doi: 10.1186/s12885-024-11897-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Kluetz PG, King‐Kallimanis BL, Suzman D, et al. Advancing assessment, analysis, and reporting of safety and tolerability in cancer trials. J Natl Cancer Inst. 2021;113(5):507‐508. doi: 10.1093/jnci/djaa135 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. George GC, Barata PC, Campbell A, et al. Improving attribution of adverse events in oncology clinical trials. Cancer Treat Rev. 2019;76:33‐40. doi: 10.1016/j.ctrv.2019.04.004 [DOI] [PubMed] [Google Scholar]
- 116. Othus M, Patel SP, Chae YK, et al. First cycle toxicity and survival in patients with rare cancers treated with checkpoint inhibitors. J Natl Cancer Inst. 2025;117(4):692‐700. doi: 10.1093/jnci/djae297 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Ip EH, Saldana S, Miller KD, et al. Tolerability of bevacizumab and chemotherapy in a phase 3 clinical trial with human epidermal growth factor receptor 2‐negative breast cancer: a trajectory analysis of adverse events. Cancer. 2021;127(24):4546‐4556. doi: 10.1002/cncr.33992 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Sartor O. Adverse event reporting in clinical trials: time to include duration as well as severity. Oncologist. 2018;23(1):1. doi: 10.1634/theoncologist.2017-0437 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Biard L, Andrillon A, Silva RB, Lee SM. Dose optimization for cancer treatments with considerations for late‐onset toxicities. Clin Trials. 2024;21(3):322‐330. doi: 10.1177/17407745231221152 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Pettit SD, Kirch R. Do current approaches to assessing therapy related adverse events align with the needs of long‐term cancer patients and survivors? Cardiooncology. 2018;4(1):5. doi: 10.1186/s40959-018-0031-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Mokbel K, Weedon M, Moye V, Jackson L. Pharmacogenetics of toxicities related to endocrine treatment in breast cancer: a systematic review and meta‐analysis. Cancer Genomics Proteomics. 2024;21(5):421‐438. doi: 10.21873/cgp.20461 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Drago JZ, Gönen M, Thanarajasingam G, et al. Inferences about drug safety in phase III trials in oncology: examples from advanced prostate cancer. J Natl Cancer Inst. 2021;113(5):553‐561. doi: 10.1093/jnci/djaa134 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Wang Y, Chen C, Du W, et al. Adverse event reporting quality in cancer clinical trials evaluating immune checkpoint inhibitor therapy: a systematic review. Front Immunol. 2022;13:874829. doi: 10.3389/fimmu.2022.874829 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Snyder C, Crossnohere N, King M, et al. The PROTEUS‐Trials Consortium: optimizing the use of patient‐reported outcomes in clinical trials. Clin Trials. 2022;19(3):277‐284. doi: 10.1177/17407745221077691 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Calvert M, Kyte D, Mercieca‐Bebber R, et al. Guidelines for inclusion of patient‐reported outcomes in clinical trial protocols: the SPIRIT‐PRO extension. JAMA. 2018;319(5):483‐494. doi: 10.1001/jama.2017.21903 [DOI] [PubMed] [Google Scholar]
- 126. Kaneyasu T, Hoshino E, Naito M, et al. How to select and understand guidelines for patient‐reported outcomes: a scoping review of existing guidance. BMC Health Serv Res. 2024;24(1):334. doi: 10.1186/s12913-024-10707-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Major A, Dueck AC, Thanarajasingam G. SOHO state of the art updates and next questions | Measuring patient‐reported outcomes (PROs) and treatment tolerability in patients with hematologic malignancies. Clin Lymphoma Myeloma Leuk. 2025;25(3):142‐155. doi: 10.1016/j.clml.2024.07.018 [DOI] [PubMed] [Google Scholar]
- 128. Coens C, Pe M, Dueck AC, et al. International standards for the analysis of quality‐of‐life and patient‐reported outcome endpoints in cancer randomised controlled trials: recommendations of the SISAQOL Consortium. Lancet Oncol. 2020;21(2):e83‐e96. doi: 10.1016/s1470-2045(19)30790-9 [DOI] [PubMed] [Google Scholar]
- 129. Polubriaginof FCG, Lipitz‐Snyderman A, Chimonas S, Kuperman GJ, Stetson PD. Patient‐reported outcomes program at scale at a cancer center. JCO Clin Cancer Inform. 2025;9:e2400253. doi: 10.1200/cci-24-00253 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Aanes SG, Wiig S, Nieder C, Haukland EC. Implementing digital patient‐reported outcomes in routine cancer care: barriers and facilitators. ESMO Real World Data Digit Oncol. 2024;6:100088. doi: 10.1016/j.esmorw.2024.100088 [DOI] [Google Scholar]
- 131. Brady KJS, Peipert JD, Atkinson TM, et al. International Society for Quality of Life Research commentary on the US Food and Drug Administration draft guidance for industry on core patient‐reported outcomes in cancer clinical trials. Qual Life Res. 2023;32(8):2155‐2163. doi: 10.1007/s11136-023-03396-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Brown R, Wages NA, Liu L, Sutton AL, Poklepovic AS. Understanding the role of patient‐reported outcomes for decision‐making in early‐phase dose‐finding clinical trials. Curr Oncol. 2025;32(3):176. doi: 10.3390/curroncol32030176 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Yap C, Lee Aiyegbusi O, Alger E, et al. Advancing patient‐centric care: integrating patient reported outcomes for tolerability assessment in early phase clinical trials—insights from an expert virtual roundtable. EClinicalMedicine. 2024;76:102838. doi: 10.1016/j.eclinm.2024.102838 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Elgarten CW, Thompson JC, Angiolillo A, et al. Improving infectious adverse event reporting for children and adolescents enrolled in clinical trials for acute lymphoblastic leukemia: a report from the Children's Oncology Group. Pediatr Blood Cancer. 2022;69(11):e29937. doi: 10.1002/pbc.29937 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Miller TP, Getz KD, Li Y, et al. Rates of laboratory adverse events by course in paediatric leukaemia ascertained with automated electronic health record extraction: a retrospective cohort study from the Children's Oncology Group. Lancet Haematol. 2022;9(9):e678‐e688. doi: 10.1016/s2352-3026(22)00168-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Bhatnagar V, Dueck AC, Efficace F, et al. Beyond maximum grade: using patient‐generated data to inform tolerability of treatments for haematological malignancies. Lancet Haematol. 2025;12(6):e463‐e469. doi: 10.1016/s2352-3026(25)00036-5 [DOI] [PubMed] [Google Scholar]
- 137. Langlais B, Mazza GL, Thanarajasingam G, et al. Evaluating treatment tolerability using the toxicity index with patient‐reported outcomes data. J Pain Symptom Manage. 2022;63(2):311‐320. doi: 10.1016/j.jpainsymman.2021.07.031 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Thanarajasingam G, Atherton PJ, Novotny PJ, Loprinzi CL, Sloan JA, Grothey A. Longitudinal adverse event assessment in oncology clinical trials: the toxicity over time (ToxT) analysis of Alliance trials NCCTG N9741 and 979254. Lancet Oncol. 2016;17(5):663‐670. doi: 10.1016/s1470-2045(16)00038-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Shalviri G, Mohebbi N, Mirbaha F, et al. Improving adverse drug event reporting by healthcare professionals. Cochrane Database Syst Rev. 2024;10(10):CD012594. doi: 10.1002/14651858.CD012594.pub2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Cancer Treatment Tolerability Consortium. National Cancer Institute. Accessed December 5, 2024. https://prevention.cancer.gov/major‐programs/cancer‐treatment‐tolerability‐consortium
- 141. Analyzing and Interpreting Clinician and Patient Adverse Event Data to Better Understand Tolerability. National Institutes of Health. Accessed July 30, 2024. https://grants.nih.gov/grants/guide/rfa‐files/rfa‐ca‐17‐052.html
- 142. Toolkit for Patient‐Focused Therapy Development. National Center for Advancing Translational Sciences. Accessed June 4, 2025. https://toolkit.ncats.nih.gov/glossary/serious‐adverse‐event
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The original contributions are included in the article; further inquiries can be directed to the corresponding author.
