Abstract
Importance:
Observational data linking physical activity and exercise exposure with either reduced risk or progression of cancer has fueled interest in the initiation of large-scale definitive trials to test the effects of exercise therapy on disease outcomes. However, several major knowledge gaps impede the rational and optimal design of such trials.
Observations:
Critical prerequisites underpinning the success of several recent contemporary anticancer agents have included adequate demonstration of antitumor activity (in phase 1/2 trials) as well as identification of essential prerequisites (e.g., biologically effective dose, predictors of response) permitting optimal design of definitive trials. The existing evidence base investigating exercise as a candidate anticancer preventive or treatment strategy is predominantly confined to observational data – data with several inherent limitations. Consequently, the antitumor activity of exercise remains unclear and perhaps more importantly, such data is not sufficient to accurately derive the dose, prescription regimen, or patients most likely to benefit from exercise. In adherence with translational frameworks for lifestyle therapy development, we highlight the urgent need for early phase 1/2-equivalent trials to fill current knowledge gaps to optimize the development and potential efficacy of exercise therapy.
Conclusions and Relevance:
Exercise therapy has significant promise to be a highly efficacious, low-toxicity, and cost-effective therapy to improve cancer outcomes. However, the majority of non-traditional therapies in cancer prevention and prognosis fail in definitive trials. We contend these failures provide critical lessons for the continued development of exercise as a candidate anticancer therapy.
Keywords: Exercise-oncology, physical activity, cancer drug development
Introduction
Despite a considerable body of observational evidence and guidelines from multiple international agencies,1–3 the prescription of exercise therapy is not considered standard of care for individuals at risk or with cancer. This scenario is in sharp contrast to the majority of other common chronic conditions wherein exercise therapy is considered ‘first-line’ primary or secondary preventive treatment. A reasonable explanation for the discrepancy in utilization of exercise therapy in oncology compared with other disease conditions likely relates to the fact that cancer is not a qualifying diagnosis for third party reimbursement of formalized exercise rehabilitation. In turn, the lack of reimbursement likely reflects the smaller body of evidence (in oncology), particularly data from definitive randomized controlled trials (RCTs) demonstrating improvement in “hard” outcomes or established surrogate markers of disease risk, progression or competing causes of mortality.4 To this end, at least two large, international phase 3 RCTs are underway investigating the efficacy of exercise therapy on cancer outcomes in stage II–III colon cancer5 and metastatic prostate cancer.6 Several additional phase 3 RCTs evaluating the efficacy of lifestyle approaches (combination of diet and physical activity promotion) on disease outcomes in various cancer populations.7,8
From the perspective of therapeutic development, the launch of phase 3 trials of exercise therapy raises the critically important question of whether the current evidence base is sufficient to support the initiation as well as rational design of such trials. The development of contemporary targeted anticancer agents closely adheres to a well-established framework involving compound screening and preclinical testing followed by ‘first in human’ phase 0 trials, phase 1 / 2 trials, and ultimately phase 3 RCTs. Among the many advantages, particular strengths of such a standardized approach include: (1) the development of only those agents exhibiting biological / antitumor activity with acceptable safety profile, and (2) elucidation of key prerequisites permitting optimal design of definitive trials (when appropriate), including drug mechanism of action, optimal dose (and schedule), and predictors of response. A comparable mandated approach is not required for the development and testing of non-regulated candidate strategies including lifestyle interventions. Consequently, over the past two decades, numerous non-regulated therapies (e.g., vitamin supplementation,9–11 metformin,12,13 aspirin,14 omega-3,15 dietary modification,16,17 psychosocial counseling18) have proceeded to definitive testing in cancer prevention or prognosis without having to achieve comparable developmental “go / no-go” milestones as that mandated in drug development.
It could be argued that a similar drug development-type framework is not required for non-regulated strategies for multiple reasons including (typical) stark differences in safety profile. However, the harsh reality is the vast majority of definitive trials of non-regulated therapies have either been negative or, in some instances, increased cancer incidence or other serious toxicities.10,11,15,19 Indeed, two recent multi-center phase 3 RCTs, both including exercise as an intervention component, did not confer a disease-free or overall survival benefit in early breast cancer20 or patients undergoing hematopoietic cell transplantation,21 respectively. We contend major contributing factors to the general failure of these trials may relate to the lack of adherence to a standardized rigorous development framework, resulting in therapies being advanced to definitive testing without: (1) adequate demonstration of antitumor activity (in smaller trials), and (2) identification of essential prerequisites (e.g., biologically effective dose, schedule) which permit rational or optimal trial design.
We recently proposed a translational framework specifically designed to facilitate investigation of exercise as a candidate anticancer strategy from initial epidemiology discovery to definitive RCTs.4 In this Opinion article, we leverage the tenets of this framework to critically evaluate the evidence base in exercise-oncology pertaining to (1) antitumor activity, (2) the most appropriate dose (and schedule), and (3) identification of patients most likely to benefit from exercise. In the final section, we consider next steps in the continued development of exercise as anticancer therapy.
Antitumor Activity of Physical Activity and Exercise: Current Evidence
Physical activity is defined by the World Health Organization as any bodily movement produced by skeletal muscles that requires energy expenditure, whereas exercise is defined as a subcategory of physical activity consisting of planned, structured, repetitive, and purposeful. The vast majority of observational studies assess physical activity rather than exercise. On the basis of multiple meta-analyses22–24 and pooled analysis,25 the Physical Activity Guidelines Advisory Committee (PAGAC) determined strong evidence exists to support the conclusion that physical activity lowers the risk of colon, breast, kidney, endometrial, bladder, esophagus (adenocarcinoma), and stomach (cardia) cancers, with moderate evidence for lung cancer risk reduction.26 The committee determined there was no relationship between physical activity and risk of thyroid or rectal cancers.26 These conclusions are largely mirrored by others organizations.27,28 In the post-diagnosis setting, the PAGAC concluded that moderate evidence supports associations among higher amounts of physical activity and lower risks of breast-, colon-, and prostate-specific mortality, as well as all-cause mortality.26
Observational data, of course, cannot prove causality. Preclinical testing is a major facet of drug development and can also be utilized to confirm the biological plausibility that exercise has anticancer activity. Unfortunately, preclinical testing of the antitumor activity of exercise is limited. A recent systematic review only identified a total of 53 in vivo preclinical studies evaluating the activity of various exercise paradigms on tumor incidence, growth, or metastasis; 35 of 53 studies reported exercise inhibited cancer growth or progression.29 This evidence base, however, has considerable methodological weaknesses as well as heterogeneity in all aspects of study design, endpoints, and efficacy, precluding meaningful comparisons and conclusions regarding the preclinical activity of exercise in any cancer model. More recently, higher quality studies have demonstrated paradigms of physical activity30 and exercise31 have antitumor activity in several, but not all, mouse models of cancer. In drug development, positive preclinical data not only provides the basis to proceed with initial clinical testing but also informs starting dose as well as other study aspects. While data from definitive trials of exercise therapy on disease outcomes in either the prevention or post-diagnosis setting are not yet available at present, multiple RCTs have investigated whether exercise alters a wide-array of biomarkers postulated to underpin the exercise – cancer pathogenesis relationship (e.g., alterations in the circulating concentrations of metabolic, sex-steroid growth factors and hormones, inflammatory cytokines, and DNA damage response / repair factors, breast mammographic density). Overall, exercise therapy of various doses (e.g., 150 mins, 300 mins/wk) and exposure periods (e.g., 6 to 12 months) are associated with small to modest alterations in a select number of markers, however effects across trials are inconsistent.32,33 Moreover, whether the observed alterations have biological or clinical importance is uncertain.34 Only one trial has investigated the effects of exercise on a biomarker (i.e., breast tissue density35), demonstrated as a strong predictor of breast cancer risk, but effects were negative.36
Hence investigation of whether exercise directly alters the evolution or biology of tumor / normal cells or the tissue / tumor microenvironment (TME) would be of immense importance to further the contention that exercise has biological antitumor activity.37 A paucity of data is currently available however. In the prevention setting, moderate-to-vigorous intensity exercise therapy (planned dose ~360 mins/wk) for 12 months increased expression of the pro-apoptotic protein (Bax) in the bottom of the colon crypts among men, whereas it decreased among women in the middle of crypts.38 In a preoperative ‘window of opportunity’ phase 2 RCT, 4 weeks of home-based exercise was associated with alterations in intratumoral gene expression, predominantly involving immune pathways, with no changes in tumor cell proliferation (Ki67) in 49 patients with primary breast cancer.39 Finally, exploratory secondary analyses from at least two phase 2 RCTs suggest structured exercise therapy after diagnosis of primary breast cancer40 or lymphoma41 may favorably impact disease outcomes. However, caution is required when interpreting these data given neither trial was primarily designed nor powered to examine cancer outcomes.
In summary, the existing evidence base supporting the antitumor activity of exercise is confined predominantly to observational data – studies that use self-reported methods to measure physical activity or exercise exposure, with (very) limited confirmatory data from preclinical or clinical studies. The limitations of observational studies are well-established and include inaccurate assessment and quantification of physical activity, with high-risk of reverse causality.42 In the next sections, we evaluate the evidence informing the most appropriate dose (and schedule) as well as predictors of response – essential requisites for optimal design of definitive trials.
Exercise Treatment Regimen
Dose:
Arguably, the most critical consideration in the design of definitive RCTs testing any medical intervention is the treatment regimen (prescription). As described, the majority of evidence supporting antitumor activity of physical activity and exercise is from observational studies. Table 1 provides a brief overview of physical activity measurement and quantification methodology used in most observational studies. In theory, therapeutic ‘dose’ level could be extracted from physical activity or exercise exposure associated with disease benefit within the cancer histology of interest, to guide dose and schedule for subsequent clinical trial evaluation. This approach, however, has major caveats.
Table 1.
Method | Example |
---|---|
Self-reported validated surveys used to measure physical activity and exercise | Participant recalls the average frequency (times/week) and duration (minutes/session) of different physical activity types, as appropriate, participated in over a distinct period of time (e.g., past 7 days or past month) |
Intensity of each reported activity is weighted using the standardized classification of energy expenditure [METs]) | One MET is the energy expenditure for sitting quietly for an hour (~3.5 mL.kg.−1min−1) |
MET values for specific activities are defined as the ratio of the metabolic rate of the activity divided by resting metabolic rate | Average pace walking has a MET value of 3 while jogging and running have MET values of 7 and 12, respectively |
MET-hours per week for each activity are calculated from the reported hours per week for that activity weighted by the assigned MET value. | Data are summed to calculate total MET-hrs.wk−1 |
Categorization of MET-hrs.wk−1 varies across studies and no standardized classification system is currently in use | Physical activity levels categorized by quartile or quintile |
Abbreviations: MET, metabolic equivalent task
First, the assessment and quantification of physical activity or exercise using self-report methods is imprecise.42 For example, the energy expenditure (or metabolic cost) associated with the same activity (of the same duration) varies considerably due to inter-individual differences in both resting and maximal metabolic rate. Relatedly, the survey instruments vary across individual studies and are often inconsistent with absolute measurements (e.g., accelerometer) – thus, the definition as well as calculation of dose is neither uniform nor acurate.43,44 Second, the amount of physical activity or exercise associated with reductions in either the primary risk or post-diagnosis disease outcomes varies considerably across individual studies. For instance, in studies examining breast cancer risk, the amount of physical activity or exercise associated with significant reductions range from 7.6 MET-hrs.wk−1 to ≥22.8 MET-hrs.wk−1; such doses are approximately the equivalent to ~120 to ~360 mins/wk of moderate-intensity exercise or 75 to 230 mins/wk of vigorous-intensity exercise.45 Similarly, studies examining the relationship between post-diagnosis physical activity / exercise and breast cancer mortality reported ‘beneficial’ amounts range from 7.6 MET-hrs.wk−1 to ≥14.9 MET-hrs.wk−1 (~120 to ~250 mins/wk of moderate-intensity exercise or ~75 to ~150 mins/wk of vigorous-intensity exercise).22 The minimum physical activity or exercise exposure associated with benefit is often not reported or emphasized, with focus primarily on the amount associated with the largest magnitude of risk reduction. Finally, clinical trials will be designed to test whether treating patients with a certain exercise dose (and schedule) impacts disease outcomes in inactive patients (selection of inactive patients is of obvious importance). Hence, the critical information required to adequately inform trial design is the change in exercise associated with disease outcomes; this data is currently not available since the vast majority of studies only assess exercise exposure at one timepoint. Interestingly, in an unplanned exploratory analysis of the Women’s Healthy Eating and Living (WHEL) Study,17 patients inactive at baseline (n=1,186) that subsequently met national guidelines at 1 year (i.e., at least 150 or 75 mins/wk of moderate or vigorous activity, respectively) via self-report was not associated with an improvement in breast cancer outcomes.46
In oncology drug development, the starting dose of new agents in first-in-human phase 0 studies is usually one-tenth of the maximum-tolerated dose (MTD) or dose-limiting toxicity in rodents; interspecies (animal to human) scaling factors are then applied to normalize to body-surface area and dosage in milligrams per kilogram.The recommended phase 2 dose (RP2D) of a particular agent is then further empirically derived from dose-finding phase 1 studies. The application of a comparable preclinical approach is not appropriate at present since similar interspecies scaling factors have not been identified for exercise. It could be argued that the exercise dose for definitive trial testing could be guided by the prescriptions tested in the numerous prevention and post-diagnosis RCTs investigating the effects of structured exercise training on alterations in various biomarkers or symptom control outcomes. Although these trials establish initial feasibility, tolerability and safety of exercise therapy in a given indication and setting (of particular importance in patients with cancer), this data is not sufficient or appropriate to guide the RP2D for therapeutic-intent trials. Indeed, the peril of this approach is observed in RCTs of established pharmacotherapeutics shown to exhibit minimal antitumor efficacy when repurposed on the basis of the originally labeled indication.47 The elucidation of standardized metrics to translate preclinical dosing to human exercise-oncology trials would represent a major advance in the field.
Regimen (Prescription) Characteristics:
Similar to systemic anticancer agents, the treatment regimen used to deliver the selected exercise dose will undoubtedly influence activity, feasibility, and safety. The major exercise regimen parameters are described in Table 2. A detailed discussion of how all the different elements of an exercise prescription may impact antitumor activity is beyond the scope of this article but has been reviewed elsewhere.48,49 On the basis of limitations described in the prior section, information derived from observational studies is arguably of minimal value in guiding the dose and schedule of a particular exercise regimen to be evaluated in a specific definitive trial. Exercise-oncology RCTs have generally tested the efficacy of standard exercise prescriptions on changes in symptom control end points such as exercise capacity and patient-reported outcomes (e.g., 3 days/wk of aerobic exercise for 30 to 45 mins/sessions over 12 to 24 weeks).48 However, using a prescription demonstrated to be efficacious for improving a non-tumor outcome symptom in a therapeutic-intent trial is problematic since it assumes the exercise load required to modulate both outcomes is the same.49 In the setting of cardiovascular medicine, the efficacy of exercise (on different outcomes) differs as a function of the exercise dose, suggesting that in order to optimize benefit, a prescription should be targeted to the primary endpoint, system(s) or pathway(s) of interest.Rational dosing loads and schedules that modulate growth factors involved in specific oncogenic pathways could be tested in clinically relevant animal models prior to guide selection of exercise prescriptions for clinical investigation in pre-specified tumor subtypes.49
Table 2.
Ongoing Phase 3 RCTs | |||
---|---|---|---|
Component | Description | INTERVAL6 | CHALLENGE5 |
Modality | Type of exercise | Structured resistance exercise and aerobic exercise | Recreational physical activity |
Frequency | Treatment sessions per week | 3 sessions per week | Increase by at least 10 MET-hrs.wk−1 to maximum of 27 MET-hrs.wk−1 |
Intensity | Dose intensity of each treatment (i.e. mild to vigorous) | High and moderate intensity sessions | Varies |
Duration | Length of each treatment session | Varies | Varies |
Length | Total period of treatment exposure | 24 months | 36 months |
Schedule | Linear (each session performed at the same intensity) or non-linear (intensity is continually altered and progressed) | Non-linear | Linear |
Setting | Supervised versus unsupervised, facility- versus home based | Supervised, facility-based x12 months; unsupervised, home-based for subsequent 12 months | Supervised, facility-based sessions for at least 6 months plus unsupervised sessions |
Delivery | One-on-one, group-based, or remote via telemedicine | One-on-one x12 months, self-managed for subsequent 12 months | One-one-on or group-based |
Abbreviations: RCT, randomized control trial; INTERVAL, Intense Exercise for Survival among Men with Metastatic Castrate-Resistant Prostate Cancer; CHALLENGE, Colon Health and Life-Long Exercise Change; MET, metabolic equivalent task.
In summary, the outlined limitations reveal that neither the dose nor prescription regimen of exercise for testing in definitive trials can be empirically or accurately derived. Thus, selection of the RP2D on the basis of the current evidence will likely be imprecise.43
Predictors of Response
Closer inspection of exercise – cancer outcomes from meta- and pooled analyses of observational data reveal that results of individual studies are not consistent, with many reporting overlapping and wide 95% confidence intervals.22,25 These data indicate considerable inter-patient heterogeneity in tumor response to exercise. Factors that may alter such a response can be broadly characterized into tumor-related and host-related factors.
Tumor-related factors.
Evaluation of the exercise – cancer pathogenesis relationship has broadly assumed that cancer is a genetic and physiologically homogeneous disease.4 However, the impact of exercise may differ as a function of clinicopathologic features (e.g., tumor size, estrogen receptor status) in early-stage breast cancer,50–52 whereas in colorectal cancer, tumorPTGS2positivity, CTNNB1 negativity, expression of CDKN1B (p27), and CD3+ cell density (infiltration) predict sensitivity to exercise.53–56 In preclinical work, exercise inhibited, had no effect, or accelerated tumor growth compared with control in three distinct, claudin-low, triple-negative breast cancer mouse models.31 These data are contrary to the observational evidence indicating that exercise benefit may be confined to ER positive disease,50 rather indicating exercise sensitivity may be subtype or molecular subtype independent. Clearly, such findings are hypothesis generating, requiring validation in independent cohorts and biologic confirmation in additional preclinical studies.
Host-related factors.
Host factors such as germline SNPs, circulating concentrations and function of immune surveillance phenotypes, inflammatory or metabolic effectors, and gut microbiota contribute to and/or modify the antitumor activity of conventional and novel agents.57–61 A paucity of data is available on whether such factors predict exercise response. Nkondjock et al62 found no association between physical activity and risk of breast cancer among BRCA mutation carriers, whereas King et al63 found physical activity delayed/reduced the lifetime risks of ovarian cancer by 54% inBRCA1mutation carriers and 23% forBRCA2mutation carriers. To our knowledge, the predictive value of circulating host factors has not been investigated in any setting. Nevertheless, related work showed the antiproliferative effect of metformin differed as a function of pre-treatment circulating metabolic factors.64,65
At present, it is not yet possible to select which patients are the most likely to derive antitumor exercise benefit. Elucidation of such factors is important to inform patient selection and insights into personalization of exercise therapy.48 Secondary analysis based on individual stratification variables (e.g., age, treatment, receptor status) will undoubtedly reveal subgroups predictive of exercise response.66 However, this approach has a number of well-recognized limitations including treatment response / heterogeneity as optimally identified by stratification based on multiple rather than single variables, and are typically underpowered, and, is at best, hypothesis-generating as opposed to hypothesis-testing.67
Where to Go From Here?
As reviewed here, essential knowledge gaps related to (1) biological activity, (2) optimal dose and schedule, and (3) predictors of response currently impede the rational as well as optimal development of exercise as cancer treatment. Clearly, discoveries from observational and preclinical studies as well as anticipated results from ongoing definitive trials will provide insights into these gaps, however, the aforementioned inherent nature and associated limitations suggest that such data will likely be of limited value to optimally guide rational trial design. Hence, in addition to ongoing and planned work in early steps outlined in our development framework (i.e., (molecular) epidemiological, preclinical testing, and safety / feasibility clinical studies), the evidence base is sufficient to proceed to the next step -conduct of early phase 1/2-equivalent trials to identify the RP2D and predictors of response.
Consistent with phase 1 studies of contemporary oncology therapeutics,68 the RP2D of exercise could be selected on the basis of feasibility / tolerability together with consideration of biological activity.4 Phase 1a dose-finding / escalation studies offer a mechanism to identify the maximal feasible dose (MFD) of exercise using metrics adapted from oncology drug development. Specifically, feasibility can be evaluated using a combination of study attrition, adherence, and frequency of dose modifications and interruptions,69,70 with safety assessed using Common Terminology Criteria for Adverse Events (CTCAE). The threshold of acceptable feasibility and safety should be defined a priori. The types of correlative science markers used to evaluate biological activity (of identified feasible doses) are dependent on setting. For example, pre-surgical ‘window of opportunity’ studies provide a unique opportunity to test the biological activity of exercise in treatment-naïve patients using surrogates of tumor response (e.g., tumor cell proliferation, pathway modulation). Indeed, standard clinical practice paradigms in several oncologic settings offer the opportunity to test the biological activity of short-term (e.g., 4 to 6 week) exercise therapy during preoperative staging workup, or in longer-term (e.g., 4 to 6 month) during neoadjuvant therapy. If supported by preclinical studies, testing the additive effects of exercise to indicated conventional therapy in the neoadjuvant or metastatic setting provide alternative settings to assess biological activity as well as explore effects on established disease surrogates (pathological complete response, progression-free survival). In the setting of minimal residual disease, measurement of circulating tumor DNA can evaluate therapeutic response via quantification of mutational load (number of mutant DNA fragments in plasma). Acquisition of pre-treatment tissue specimens will be critical for discovery of novel genomic and clinical predictors of response. As in drug trials, dosing cohorts achieving the minimal feasibility criteria but exhibiting biological activity could be selected for further evaluation in safety / dose-expansion (phase 1b) testing. Other considerations in clinical testing of exercise are summarized in Table 3. A summary of the current evidence base, limitations, and recommended future directions are presented in Table 4.
Table 3.
Consideration | Description |
---|---|
Dose quantification |
|
Novel trial designs |
|
Biological activity |
|
Quality reporting |
|
Implementation |
|
Table 4.
Current evidence |
|
Limitations |
|
Future directions |
|
Conclusion
Exercise therapy has significant promise to be a highly efficacious, low-toxicity, and cost-effective therapy to improve cancer risk and outcomes. Patients are also interested in the therapeutic potential of exercise – a strategy within patients’ control that can potentially alter disease pathogenesis and symptomology without the adverse effects of conventional pharmacological agents. However, the majority of non-traditional therapies fail in definitive trials. We contend these failures provide critical lessons for the continued development of exercise as a candidate antitumor strategy. Indeed, persisting with the current approach used to develop and test non-traditional strategies may continue to yield null results, thereby potentiating the sentiment that such interventions (including exercise therapy or lifestyle interventions) are not effective means to reduce primary cancer incidence and progression. In this paper, we attempted to highlight some of the important knowledge gaps currently precluding optimal development of exercise as anticancer therapy as well as outline potential next steps. We hope these concepts provide one potential platform and approach to optimize the therapeutic promise of exercise therapy.
ACKNOWLEDGEMENTS
NMI is supported by research grants from the National Cancer Institute, the Breast Cancer Research Foundation, and the Conquer Cancer Foundation of the American Society of Clinical Oncology. LWJ is supported by research grants from the National Cancer Institute, the KavliTrust and AKTIV Against Cancer. NMI and LWJ are supported by the Memorial Sloan Kettering Cancer Center Support Grant/Core Grant (P30 CA008748).
Footnotes
Conflict of interests: NMI – consulting fees from Novartis and Puma Biotechnologies; LWJ – stock ownership in Pacylex, Inc.
REFERENCES
- 1.Rock CL, Doyle C, Demark-Wahnefried W, et al. Nutrition and physical activity guidelines for cancer survivors. CA: a cancer journal for clinicians. Jul-Aug 2012;62(4):243–274. [DOI] [PubMed] [Google Scholar]
- 2.Schmitz KH, Courneya KS, Matthews C, et al. American College of Sports Medicine roundtable on exercise guidelines for cancer survivors. Med Sci Sports Exerc. July 2010;42(7):1409–1426. [DOI] [PubMed] [Google Scholar]
- 3.Cormie P, Atkinson M, Bucci L, et al. Clinical Oncology Society of Australia position statement on exercise in cancer care. Med J Aust. August 20 2018;209(4):184–187. [DOI] [PubMed] [Google Scholar]
- 4.Jones LW. Precision Oncology Framework for Investigation of Exercise As Treatment for Cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. December 10 2015;33(35):4134–4137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Courneya KS, Booth CM, Gill S, et al. The Colon Health and Life-Long Exercise Change trial: a randomized trial of the National Cancer Institute of Canada Clinical Trials Group. Curr Oncol. December 2008;15(6):279–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Newton RU, Kenfield SA, Hart NH, et al. Intense Exercise for Survival among Men with Metastatic Castrate-Resistant Prostate Cancer (INTERVAL-GAP4): a multicentre, randomised, controlled phase III study protocol. BMJ Open. May 14 2018;8(5):e022899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Chlebowski RT, Reeves MM. Weight Loss Randomized Intervention Trials in Female Cancer Survivors. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. December 10 2016;34(35):4238–4248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Rack B, Andergassen U, Neugebauer J, et al. The German SUCCESS C Study - The First European Lifestyle Study on Breast Cancer. Breast Care (Basel). 2010;5(6):395–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Goodman GE, Schaffer S, Omenn GS, Chen C, King I. The association between lung and prostate cancer risk, and serum micronutrients: results and lessons learned from beta-carotene and retinol efficacy trial. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. June 2003;12(6):518–526. [PubMed] [Google Scholar]
- 10.Byers T Anticancer vitamins du Jour--The ABCED’s so far. American journal of epidemiology. July 1 2010;172(1):1–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Manson JE, Cook NR, Lee IM, et al. Vitamin D Supplements and Prevention of Cancer and Cardiovascular Disease. The New England journal of medicine. January 3 2019;380(1):33–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Parulekar W, Chen BE, Elliott C, et al. A phase III randomized trial of metformin versus placebo on recurrence and survival in early-stage breast cancer (BC) (NCIC Clinical Trials Group MA.32) 2011;29(15_suppl):TPS103–TPS103. [Google Scholar]
- 13.Kitson S, Maskell Z, Sivalingam VN, et al. PRE-surgical Metformin In Uterine Malignancy (PREMIUM): a multi-center, randomized double-blind, placebo-controlled phase 3 trial. Clinical cancer research : an official journal of the American Association for Cancer Research. December 18 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Chen WY, Winer EP, Barry WT, et al. ABC trial (A011502): Randomized phase III double blinded placebo controlled trial of aspirin as adjuvant therapy for breast cancer. 2018;36(15_suppl):TPS597–TPS597. [Google Scholar]
- 15.Manson JE, Cook NR, Lee IM, et al. Marine n-3 Fatty Acids and Prevention of Cardiovascular Disease and Cancer. The New England journal of medicine. January 3 2019;380(1):23–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Chlebowski RT, Blackburn GL, Thomson CA, et al. Dietary fat reduction and breast cancer outcome: interim efficacy results from the Women’s Intervention Nutrition Study. Journal of the National Cancer Institute. December 20 2006;98(24):1767–1776. [DOI] [PubMed] [Google Scholar]
- 17.Pierce JP, Natarajan L, Caan BJ, et al. Influence of a diet very high in vegetables, fruit, and fiber and low in fat on prognosis following treatment for breast cancer: the Women’s Healthy Eating and Living (WHEL) randomized trial. JAMA : the journal of the American Medical Association. July 18 2007;298(3):289–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Spiegel D, Bloom JR, Kraemer HC, Gottheil E. Effect of psychosocial treatment on survival of patients with metastatic breast cancer. Lancet. October 14 1989;2(8668):888–891. [DOI] [PubMed] [Google Scholar]
- 19.Goodwin PJ, Leszcz M, Ennis M, et al. The effect of group psychosocial support on survival in metastatic breast cancer. The New England journal of medicine. December 13 2001;345(24):1719–1726. [DOI] [PubMed] [Google Scholar]
- 20.Janni W, Rack B, Friedl T, et al. Abstract GS5–03: Lifestyle Intervention and Effect on Disease-free Survival in Early Breast Cancer Pts: Interim Analysis from the Randomized SUCCESS C Study. 2019;79(4 Supplement):GS5–03-GS05–03. [Google Scholar]
- 21.Jacobsen PB, Le-Rademacher J, Jim H, et al. Exercise and stress management training prior to hematopoietic cell transplantation: Blood and Marrow Transplant Clinical Trials Network (BMT CTN) 0902. Biol Blood Marrow Transplant. October 2014;20(10):1530–1536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Friedenreich CM, Neilson HK, Farris MS, Courneya KS. Physical Activity and Cancer Outcomes: A Precision Medicine Approach. Clinical cancer research : an official journal of the American Association for Cancer Research. October 1 2016;22(19):4766–4775. [DOI] [PubMed] [Google Scholar]
- 23.Lahart IM, Metsios GS, Nevill AM, Carmichael AR. Physical activity, risk of death and recurrence in breast cancer survivors: A systematic review and meta-analysis of epidemiological studies. Acta Oncol. May 2015;54(5):635–654. [DOI] [PubMed] [Google Scholar]
- 24.Schmid D, Leitzmann MF. Association between physical activity and mortality among breast cancer and colorectal cancer survivors: a systematic review and meta-analysis. Annals of oncology : official journal of the European Society for Medical Oncology / ESMO. July 2014;25(7):1293–1311. [DOI] [PubMed] [Google Scholar]
- 25.Moore SC, Lee IM, Weiderpass E, et al. Association of Leisure-Time Physical Activity With Risk of 26 Types of Cancer in 1.44 Million Adults. JAMA Intern Med. June 1 2016;176(6):816–825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Physical Activity Guidelines Advisory Committee Scientific Report. 2018; https://health.gov/news/blog-bayw/2018/03/2018-physical-activity-guidelines-advisory-committee-submits-scientific-report/. Accessed March 8, 2019.
- 27.Diet, Nutrition, Physical Activity and Cancer: a Global Perspective. World Cancer Research Fund / American Institute for Cancer Research 2018; https://www.wcrf.org/dietandcancer. Accessed March 8, 2019.
- 28.Kushi LH, Doyle C, McCullough M, et al. American Cancer Society Guidelines on nutrition and physical activity for cancer prevention: reducing the risk of cancer with healthy food choices and physical activity. CA: a cancer journal for clinicians. Jan-Feb 2012;62(1):30–67. [DOI] [PubMed] [Google Scholar]
- 29.Ashcraft KA, Peace RM, Betof AS, Dewhirst MW, Jones LW. Efficacy and Mechanisms of Aerobic Exercise on Cancer Initiation, Progression, and Metastasis: A Critical Systematic Review of In Vivo Preclinical Data. Cancer research. July 15 2016;76(14):4032–4050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Pedersen L, Idorn M, Olofsson GH, et al. Voluntary Running Suppresses Tumor Growth through Epinephrine- and IL-6-Dependent NK Cell Mobilization and Redistribution. Cell metabolism. March 8 2016;23(3):554–562. [DOI] [PubMed] [Google Scholar]
- 31.Glass OK, Bowie M, Fuller J, et al. Differential response to exercise in claudin-low breast cancer. Oncotarget. November 24 2017;8(60):100989–101004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Betof AS, Dewhirst MW, Jones LW. Effects and potential mechanisms of exercise training on cancer progression: a translational perspective. Brain Behav Immun. March 2013;30 Suppl:S75–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kang DW, Lee J, Suh SH, Ligibel J, Courneya KS, Jeon JY. Effects of Exercise on Insulin, IGF Axis, Adipocytokines, and Inflammatory Markers in Breast Cancer Survivors: A Systematic Review and Meta-analysis. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. March 2017;26(3):355–365. [DOI] [PubMed] [Google Scholar]
- 34.Chlebowski RT. Lifestyle and breast cancer risk: the way forward? Journal of clinical oncology : official journal of the American Society of Clinical Oncology. March 20 2010;28(9):1445–1447. [DOI] [PubMed] [Google Scholar]
- 35.Boyd NF, Guo H, Martin LJ, et al. Mammographic density and the risk and detection of breast cancer. The New England journal of medicine. January 18 2007;356(3):227–236. [DOI] [PubMed] [Google Scholar]
- 36.Woolcott CG, Courneya KS, Boyd NF, et al. Mammographic density change with 1 year of aerobic exercise among postmenopausal women: a randomized controlled trial. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. April 2010;19(4):1112–1121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Koelwyn GJ, Quail DF, Zhang X, White RM, Jones LW. Exercise-dependent regulation of the tumour microenvironment. Nature reviews. Cancer. September 25 2017;17(10):620–632. [DOI] [PubMed] [Google Scholar]
- 38.Campbell KL, McTiernan A, Li SS, et al. Effect of a 12-month exercise intervention on the apoptotic regulating proteins Bax and Bcl-2 in colon crypts: a randomized controlled trial. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. September 2007;16(9):1767–1774. [DOI] [PubMed] [Google Scholar]
- 39.Ligibel J, Dillon DA, Giobbie-Hurder A, et al. Impact of a pre-operative exercise intervention on breast cancer proliferation and gene expression: Results from the Pre-Operative Health and Body (PreHAB) Study. Clinical cancer research : an official journal of the American Association for Cancer Research. April 24 2019. [DOI] [PubMed] [Google Scholar]
- 40.Courneya KS, Segal RJ, McKenzie DC, et al. Effects of exercise during adjuvant chemotherapy on breast cancer outcomes. Med Sci Sports Exerc. September 2014;46(9):1744–1751. [DOI] [PubMed] [Google Scholar]
- 41.Courneya KS, Friedenreich CM, Franco-Villalobos C, et al. Effects of supervised exercise on progression-free survival in lymphoma patients: an exploratory follow-up of the HELP Trial. Cancer causes & control : CCC. February 2015;26(2):269–276. [DOI] [PubMed] [Google Scholar]
- 42.van Poppel MN, Chinapaw MJ, Mokkink LB, van Mechelen W, Terwee CB. Physical activity questionnaires for adults: a systematic review of measurement properties. Sports Med. July 1 2010;40(7):565–600. [DOI] [PubMed] [Google Scholar]
- 43.Jones LW. Precision Oncology Framework for Investigation of Exercise as Treatment for Cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. October 12 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Dyrstad SM, Hansen BH, Holme IM, Anderssen SA. Comparison of self-reported versus accelerometer-measured physical activity. Med Sci Sports Exerc. January 2014;46(1):99–106. [DOI] [PubMed] [Google Scholar]
- 45.Friedenreich CM. Physical activity and breast cancer: review of the epidemiologic evidence and biologic mechanisms. Recent Results Cancer Res. 2011;188:125–139. [DOI] [PubMed] [Google Scholar]
- 46.Bertram LA, Stefanick ML, Saquib N, et al. Physical activity, additional breast cancer events, and mortality among early-stage breast cancer survivors: findings from the WHEL Study. Cancer causes & control : CCC. March 2011;22(3):427–435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Abdullah MI, de Wolf E, Jawad MJ, Richardson A. The poor design of clinical trials of statins in oncology may explain their failure - Lessons for drug repurposing. Cancer treatment reviews. September 2018;69:84–89. [DOI] [PubMed] [Google Scholar]
- 48.Scott JM, Nilsen TS, Gupta D, Jones LW. Exercise Therapy and Cardiovascular Toxicity in Cancer. Circulation. March 13 2018;137(11):1176–1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Jones LW, Eves ND, Scott JM. Bench-to-Bedside Approaches for Personalized Exercise Therapy in Cancer. American Society of Clinical Oncology educational book / ASCO. American Society of Clinical Oncology. Meeting 2017;37:684–694. [DOI] [PubMed] [Google Scholar]
- 50.Jones LW, Kwan ML, Weltzien E, et al. Exercise and Prognosis on the Basis of Clinicopathologic and Molecular Features in Early-Stage Breast Cancer: The LACE and Pathways Studies. Cancer research. September 15 2016;76(18):5415–5422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Holmes MD, Chen WY, Feskanich D, Kroenke CH, Colditz GA. Physical activity and survival after breast cancer diagnosis. JAMA : the journal of the American Medical Association. May 25 2005;293(20):2479–2486. [DOI] [PubMed] [Google Scholar]
- 52.Irwin ML, Duggan C, Wang CY, et al. Fasting C-peptide levels and death resulting from all causes and breast cancer: the health, eating, activity, and lifestyle study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. January 1 2011;29(1):47–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Morikawa T, Kuchiba A, Yamauchi M, et al. Association of CTNNB1 (beta-catenin) alterations, body mass index, and physical activity with survival in patients with colorectal cancer. JAMA : the journal of the American Medical Association. April 27 2011;305(16):1685–1694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Meyerhardt JA, Ogino S, Kirkner GJ, et al. Interaction of molecular markers and physical activity on mortality in patients with colon cancer. Clinical cancer research : an official journal of the American Association for Cancer Research. September 15 2009;15(18):5931–5936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Yamauchi M, Lochhead P, Imamura Y, et al. Physical activity, tumor PTGS2 expression, and survival in patients with colorectal cancer. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. June 2013;22(6):1142–1152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Koh H, Hamada T, Song M, et al. Physical Activity and Colorectal Cancer Prognosis According to Tumor-Infiltrating T Cells. JNCI Cancer Spectrum. 2019;2(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Helmink BA, Khan MAW, Hermann A, Gopalakrishnan V, Wargo JA. The microbiome, cancer, and cancer therapy. Nature medicine. March 6 2019. [DOI] [PubMed] [Google Scholar]
- 58.Sauna ZE, Kimchi-Sarfaty C, Ambudkar SV, Gottesman MM. Silent polymorphisms speak: how they affect pharmacogenomics and the treatment of cancer. Cancer research. October 15 2007;67(20):9609–9612. [DOI] [PubMed] [Google Scholar]
- 59.Dranoff G Cytokines in cancer pathogenesis and cancer therapy. Nature reviews. Cancer. January 2004;4(1):11–22. [DOI] [PubMed] [Google Scholar]
- 60.Goodwin PJ, Ennis M, Pritchard KI, et al. Fasting insulin and outcome in early-stage breast cancer: results of a prospective cohort study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. January 1 2002;20(1):42–51. [DOI] [PubMed] [Google Scholar]
- 61.Hegde PS, Karanikas V, Evers S. The Where, the When, and the How of Immune Monitoring for Cancer Immunotherapies in the Era of Checkpoint Inhibition. Clinical cancer research : an official journal of the American Association for Cancer Research. April 15 2016;22(8):1865–1874. [DOI] [PubMed] [Google Scholar]
- 62.Nkondjock A, Robidoux A, Paredes Y, Narod SA, Ghadirian P. Diet, lifestyle and BRCA-related breast cancer risk among French-Canadians. Breast cancer research and treatment. August 2006;98(3):285–294. [DOI] [PubMed] [Google Scholar]
- 63.King MC, Marks JH, Mandell JB, New York Breast Cancer Study G. Breast and ovarian cancer risks due to inherited mutations in BRCA1 and BRCA2. Science. October 24 2003;302(5645):643–646. [DOI] [PubMed] [Google Scholar]
- 64.Bonanni B, Puntoni M, Cazzaniga M, et al. Dual effect of metformin on breast cancer proliferation in a randomized presurgical trial. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. July 20 2012;30(21):2593–2600. [DOI] [PubMed] [Google Scholar]
- 65.DeCensi A, Puntoni M, Gandini S, et al. Differential effects of metformin on breast cancer proliferation according to markers of insulin resistance and tumor subtype in a randomized presurgical trial. Breast cancer research and treatment. November 2014;148(1):81–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Yeh RW, Kramer DB. Decision Tools to Improve Personalized Care in Cardiovascular Disease: Moving the Art of Medicine Toward Science. Circulation. March 21 2017;135(12):1097–1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Barraclough H, Govindan R. Biostatistics primer: what a clinician ought to know: subgroup analyses. J Thorac Oncol. May 2010;5(5):741–746. [DOI] [PubMed] [Google Scholar]
- 68.Hansen AR, Cook N, Amir E, Siu LL, Abdul Razak AR. Determinants of the recommended phase 2 dose of molecular targeted agents. Cancer. April 15 2017;123(8):1409–1415. [DOI] [PubMed] [Google Scholar]
- 69.Scott JM, Iyengar NM, Nilsen TS, et al. Feasibility, safety, and efficacy of aerobic training in pretreated patients with metastatic breast cancer: A randomized controlled trial. Cancer. June 15 2018;124(12):2552–2560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Nilsen TS, Scott JM, Michalski M, et al. Novel Methods for Reporting of Exercise Dose and Adherence: An Exploratory Analysis. Med Sci Sports Exerc. June 2018;50(6):1134–1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Sasso JP, Eves ND, Christensen JF, Koelwyn GJ, Scott J, Jones LW. A framework for prescription in exercise-oncology research. J Cachexia Sarcopenia Muscle. June 2015;6(2):115–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Mao X, Cheung YK. Sequential designs for individualized dosing in phase I cancer clinical trials. Contemporary clinical trials. August 31 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Cheung YK, Chappell R. Sequential designs for phase I clinical trials with late-onset toxicities. Biometrics. December 2000;56(4):1177–1182. [DOI] [PubMed] [Google Scholar]
- 74.Normolle D, Lawrence T. Designing dose-escalation trials with late-onset toxicities using the time-to-event continual reassessment method. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. September 20 2006;24(27):4426–4433. [DOI] [PubMed] [Google Scholar]
- 75.Hopewell S, Dutton S, Yu LM, Chan AW, Altman DG. The quality of reports of randomised trials in 2000 and 2006: comparative study of articles indexed in PubMed. BMJ. March 23 2010;340:c723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Plint AC, Moher D, Morrison A, et al. Does the CONSORT checklist improve the quality of reports of randomised controlled trials? A systematic review. Med J Aust. 2006;185(5):263. [DOI] [PubMed] [Google Scholar]
- 77.Adams SC, Iyengar NM, Scott JM, Jones LW. Exercise Implementation in Oncology: One Size Does Not Fit All. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. March 20 2018;36(9):925–926. [DOI] [PubMed] [Google Scholar]