IMR Press / RCM / Volume 24 / Issue 6 / DOI: 10.31083/j.rcm2406160
Open Access Review
Current Evidence on the Benefit of Exercise in Cancer Patients: Effects on Cardiovascular Mortality, Cardiotoxicity, and Quality of Life
Show Less
1 Cardiology Department, Hospital Universitari Doctor Josep Trueta and Hospital Santa Caterina, 17007 Girona, Spain
2 Medical Science Department, School of Medicine, University of Girona, 17003 Girona, Spain
3 Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
4 Dirección Territorial de Radiologia y Medicina Nuclear de Girona, Insititut de Diagnòstic per la Imatge (IDI), 17007 Girona, Spain
5 Radiation Oncology Department, Institut Català d’Oncologia, 17007 Girona, Spain
6 Radiation Oncology Department, Institut Català d’Oncologia, L'Hospitalet de Llobregat, 08908 Barcelona, Spain
7 Center for Cardiovascular Genetics, Biomedical Research Institute of Girona, 17190 Girona, Spain
*Correspondence: moral.sergio@yahoo.es (Sergio Moral)
Rev. Cardiovasc. Med. 2023, 24(6), 160; https://doi.org/10.31083/j.rcm2406160
Submitted: 3 November 2022 | Revised: 15 February 2023 | Accepted: 17 February 2023 | Published: 6 June 2023
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Cancer and its treatments affect cardiovascular (CV) health, including an increased risk of CV death, decreased cardiorespiratory fitness (CRF), and cardiac dysfunction. Moreover, cancer-related fatigue and worse quality of life (QoL) are highly prevalent adverse effects experienced by patients during treatment and can persist years after therapy ends. Physical exercise has been proposed as a strategy to improve different aspects of life of cancer patients, and is recommended as a therapy in cardio-oncology guidelines. Exercise interventions reduce fatigue and improve QoL in patients with both solid tumors and hematological malignancies, although there is a lack of awareness of exercise recommendations, timing, and referral to such programs. New evidence indicates that physical activities improve CRF, which can lead to a reduction in CV mortality. Furthermore, cardiac dysfunction is a side effect of many oncological treatments, which may be mitigated by exercise interventions according to preclinical studies and recent publications. Nevertheless, specific physical exercise programs are not widely used in cancer patients. Thus, the goal of this review was to describe the current evidence on the benefits of exercise in cancer patients, the gaps that remain, and an approach to exercise prescription.

Keywords
cardiotoxicity
cardiorespiratory fitness
cardio-oncology
cardiovascular health
1. Introduction

The effects of cancer and its treatments including chemotherapy, radiation, hormonal, and/or biological therapies can affect cardiovascular (CV) health, causing worsening of the CV profile, prognosis, cardiac function, cachexia, fatigue, and quality of life (QoL). CV mortality is over 20% in patients with breast, endometrial, and thyroid cancers and is almost 30% in those with prostate cancer [1, 2]. Over half of cancer patients report moderate-to-severe fatigue during treatment, of which 25% have persistent fatigue more than 5 years after treatment completion, and 5–26% of cases have a decline in cardiorespiratory fitness (CRF) [3, 4]. Moreover, many current cancer therapies cause significant adverse CV events, such as decreased cardiac function or heart failure. Furthermore, CV disease (CVD) and cancer share risk factors such as obesity, hyperglycemia, hypertension, and hypertriglyceridemia-induced inflammation, promoting carcinogenesis and tumor progression [5, 6].

Physical exercise has been positioned as a strategy to improve different aspects of life of cancer patients (Table 1, Ref. [7, 8]), and is recommended as a therapy in cardio-oncology guidelines. Different studies have shown the benefits of exercise in decreasing CV mortality, and improving CRF and QoL. Accurate exercise prescription is mandatory to obtain the expected benefits of reducing CV risk and mortality [7, 8].

Table 1.Difficulties and benefits of exercise prescription in cancer patients.
Difficulties [7, 8] Benefits [7, 8]
Limited access to resources to support exercise Improves cardiorespiratory fitness
Limited financial coverage Ameliorates quality of life
Difficulty of adherence and motivation Reduces fatigue
Improves the cardiovascular profile
Reduces cardiovascular mortality

Here, we provide an overview of the current evidence on the benefits of exercise interventions in cancer patients in terms of mortality and CRF, primary prevention of cardiotoxicity, fatigue, and QoL as well as the optimal timing of physical exercise and prescription recommendations.

2. Cancer and CV Health

Cancer survivors have an increased risk of long-term CV mortality compared to the general population, either because of unhealthy lifestyles or the toxicities of their treatment. The risk of death from CVD differs depending on the type of cancer and is estimated to be between 3% and 5% for brain and liver cancers, but can increase to 30–40% for prostate and bladder malignancies [1, 9, 10, 11]. Furthermore, significant and marked impairment of CRF has been demonstrated along the entire disease continuum, which may not recover after the completion of treatment. An inversely proportional correlation between CRF and CV mortality has been described, leading to a significant increase in mortality risk with decreasing CRF levels. This relationship is independent of traditional CV risk factors [4, 12, 13, 14, 15].

Cachexia is a severe complication of cancer that negatively affects QoL, response to chemotherapy, and survival; it affects more than 50% of cancer patients and accounts for up to 20% of cancer-related deaths. Cachexia is considered a multi-organ disease that involves different tissues and organs, including the heart. Cardiac cachexia has been observed in some cancer types, such as lung, pancreatic, and gastrointestinal cancers, and occurs primarily as a consequence of cardiac protein loss [16, 17].

Cancer therapy can also adversely affect cardiac structure and/or function, resulting in ventricular dysfunction that may be asymptomatic or symptomatic, manifesting as heart failure. This condition has been termed “cancer therapy-related cardiac dysfunction” (CTRCD) and occurs with many cancer therapies. The incidences of CTRCD induced by the commonly administered therapies anthracyclines (ACs), human epidermal growth factor receptor 2 (HER2)-targeted agents (e.g., trastuzumab), and immune checkpoint inhibitors are 5–40% (depending on dose and duration of exposure), 18%, and 15% [18, 19, 20, 21], respectively. Moreover, cardiac diastolic function may be impaired even with a low dose of AC as well as with new cancer therapies, which some authors consider a precursor to cardiovascular events [22, 23, 24].

Both cancer survivors and patients undergoing active treatment report fatigue in 62–85% of cases, as well as impairment of health-related QoL (HRQoL) (Fig. 1). The mechanisms underlying cancer-related fatigue suggest the involvement of complex multifactorial processes linked to a range of molecular/physiological processes such as inflammation, as well as the cumulative effects of cancer treatment and psychological factors [25].

Fig. 1.

Effects of cancer in cardiovascular health. CRF, cardiorespiratory fitness; CTRCD, cancer therapy-related cardiac dysfunction; CV, cardiovascular.

3. Effects of Exercise on Cancer Patients
3.1 Exercise’s Effect on CV Mortality

In current guidelines, physical exercise has been proposed to reduce morbidity and mortality in cancer patients [7, 8]. Although randomized controlled trials (RCTs) are lacking, current published meta-analyses and retrospective studies provide reliable evidence to support physical exercise as a strategy to decrease CV mortality (Table 2, Ref. [26, 27, 28, 29, 30]). This benefit can be partly explained by the fact that a considerable proportion of patients are at significant risk of CVD at the time of cancer diagnosis or have a worsened CV profile during treatment. In addition, the risk of CV toxicity is higher in patients with hypertension, hyperlipidemia, smoking, diabetes, or obesity [31]. Exposure to chemotherapy leads to vascular endothelial dysfunction, which contributes to the development of CVD. Current evidence shows that exercise training improves vascular endothelial function and wall thickening, especially in breast and prostate cancer survivors [32, 33, 34]. Thus, a supervised and structured physical exercise program would improve the CV profile of cancer patients and, therefore, potentially reduce CV mortality in the medium and long term.

Table 2.Trials evaluating the effect of exercise on mortality and CRF.
Study Type Cancer N Results
Kuiper JG et al. 2012 [26] Prospective Colorectal 1339 PA of 18 MET-hours/week had significantly lower colorectal cancer-specific mortality and all-cause mortality
Beasley JM et al. 2016 [27] Prospective Breast 2265 Reduction in death from any cause and death from breast cancer for women who met the PA of 10 MET-hours/week 18 to 48 months post-diagnosis
Cormie et al. 2017 [28] Meta-analyses Breast 68,285 Trend to reduced risk of mortality in patients with higher exercise behaviours
15 cohort studies Colorectal
4 RCT Prostate
Scott et al. 2018 [30] Meta-analyses Breast 3632 VO2peak increased by +2.80 mL O2 × kg-1 × min-1 with exercise therapy compared with +0.02 mL O2 × kg-1 × min-1 in the control group
48 RCT Prostate 1990 (55%) Ex
Lung, Hematologic 1642 (45%) UC
Colorectal
Gastrointestinal
Kim et al. 2021 [29] Retrospective Breast 39,775 All exercise intensities had a lower risk of CVD

CVD, cardiovascular disease; Ex, exercise; PA, physical activity; RCT, randomized control trial; UC, usual care; CRF, cardiorespiratory fitness; VO2peak, oxigen consumption peak; MET, metabolic equivalents task units.

In addition to the positive impacts of physical exercise on the CV profile, the role of muscle-strengthening physical activity is essential for cancer patients and is associated with a lower risk of cancer mortality. This favorable effect could be explained by the reduction in systemic inflammation or insulin resistance [35, 36]. Recently, a J-shaped relationship has been observed between muscle-strengthening activities and cancer mortality, with the greatest reduction in risk occurring with 30–60 min of activity per week [37].

Cancer cachexia negatively affects survival. Exercise, as a therapeutic approach to decrease skeletal muscle degradation and body weight loss, could be adequate therapy. However, cachexia generally appears late in the disease, and there have been very few robust RCTs to determine the best therapeutic approach using physical activity [16].

3.2 Exercise’s Effect on CRF

Cancer patients present with significant impairment of CRF, which can be improved by exercise therapy. Scott et al. [30] conducted a meta-analysis to evaluate the effects of exercise therapy on adult-onset cancer patients as measured by peak oxygen consumption (VO2peak), an integrative assessment of global CV function. A significant increase of VO2peak in the exercise group compared with usual care was reported, with no significant differences in terms of safety, which favored exercise therapy (Table 2). Furthermore, in breast and prostate cancers, an increase in CRF has been observed in patients involved in physical exercise programs, whereas a decrease has been observed in the control groups [38, 39].

3.3 Exercise as the Primary Prevention of CTRCD

Preclinical studies have shown that exercise interventions protect against AC-induced cardiotoxicity in rodents; less doxorubicin accumulation in cardiac tissue may be the key underlying mechanism [40, 41, 42]. Furthermore, an experimental study in rats demonstrated that exercise preserved cardiac function and attenuated the autophagic response in the heart and tumor tissues in cancer-induced cardiac cachexia [43]. In humans, there are only a few studies with a limited sample size that have evaluated the cardioprotective effects of exercise (Table 3, Ref. [44, 45, 46, 47, 48]).

Table 3.Trials evaluating cardioprotection strategy with exercise.
Study Type Cancer Cancer Therapy Trial intervention Exercise specification N Primary outcome measures
Haykowsky et al. 2009 [47] Single group study Breast Cancer HER2 positive Trastuzumab Exercise 3 days per week during 4 m of trastuzumab therapy N = 17 No statistically significant change in LV volumes, mass and ejection fraction
5 min warm-up
30–60 min cycle at a HR equal to 60% to 90% of peak oxygen consumption.
5 min cool down
Kirkham et al. 2017 [44] Two-arm proof-of-concept randomized controlled trial Breast cancer Anthracycline Single bout exercise training vs usual care Single bout of supervised treadmill exercise: N = 24 Change in biomarkers
Doxorubicin 10 min warm-up 11 = Ex No change in echocardiographyic paramteres
30 min at 70% of age-predicted HR 13 = UC
5 min cool down
Ma Z et al. 2018 [46] Randomized controlled trial Breast Cancer Anthracycline Exercise training vs usual care 16-week exercise supervised program N = 64 LVEF significantly increased after chemotheraphy in EX group and decrease in control group, statistically significant (p < 0.05)
33 = EX
31 = UC
Kirkham et al. 2020 [45] Prospective, non-randomized controlled trial Breast Cancer Anthracycline Exercise training vs usual care 3 sessions per week N = 37 No difference on resting cardiac function
20–30 min of treadmill, elliptical, or cycle ergometer 26 = EX
aerobic exercise at 50–75% of age-predicted HR 11 = UC
Hojan et al. 2020 [48] Randomized controlled trial Breast Cancer HER2 positive Trastuzumab Exercise training vs usual care Daily sessions 9 w N = 47 Statistically significant decrease of the LVEF (p < 0.05) in the control group compared to the intervention group
Endurance: 26 = EX
2 min warm-up 45 min aerobic activities 21 = UC
3 min relaxation period
Strength: 40–45 min

Min, minutes; EX, exercise; UC, usual care; HR, heart rate; LVEF, left ventricular ejection fraction; HER2, human epidermal growth factor receptor 2; LV, left ventricle.

In patients with breast cancer treated with AC, Kikhram et al. [44, 45] found no differences in resting cardiac function when comparing patients who performed aerobic exercise with the control group. However, an improvement in hemodynamic responses was detected by increasing cardiac output and decreasing systemic vascular resistance, which indicated a positive pathophysiological impact on the CV profile. In addition, Ma et al. [46] reported a positive result in the prevention of ventricular function decline in the physical exercise group.

Pertaining to preventing CTRCD secondary to trastuzumab therapy, discrepancies are found in the medical literature regarding the potential benefits of exercise in this clinical scenario. A prospective study by Haykowsky et al. [47] analyzed HER2-positive breast cancer patients who did aerobic training during the first 4 months of adjuvant trastuzumab, and underwent cardiac magnetic resonances before and after the exercise protocol. Left ventricular cavity dilation and worsening of ejection fraction were observed despite aerobic exercise training. Nonetheless, an RCT conducted by Hojan et al. [48] reported a statistically significant decrease of left ventricular ejection fraction (LVEF) in the control group compared to the intervention group.

Concerning diastolic function, a prospective cohort study of HER2-positive breast cancer women receiving AC and trastuzumab reported that physical activity of moderate-to-vigorous intensity was associated with better diastolic parameters (higher E/A and lower E/e’ ratio) [49]. Preclinical evidence and current clinical data available suggest that physical exercise may prevent the decline of LVEF secondary to cardiotoxic treatments, especially in selected patients; however, larger RCTs are needed to clarify the role of physical exercise in preventing cardiotoxicity in cancer patients.

3.4 Exercise’s Effects on Fatigue and QoL

There is strong evidence for the beneficial effect of exercise interventions on reducing fatigue and improving HRQoL in both solid tumors and hematological malignancies compared to usual care [50, 51, 52].

In solid tumors, the benefits of exercise have been reported in several cancer types, such as breast, colorectal cancer (CRC), lung, and prostate cancers. A meta-analysis by Juvet et al. [53] investigated the effects of exercise interventions on women with breast cancer during and after treatment (chemotherapy and/or radiation) and demonstrated that regular exercise decreases fatigue; this benefit persisted at the 6-month follow-up compared to usual care. Exercise programs in patients with CRC have been shown to improve several patient-reported HRQoL outcomes, including physical function, cancer-specific QoL, sleep quality, and fatigue [54]. This benefit is described in different stages of CRC, either pre-surgery, during chemotherapy, or after treatment. Therefore, exercise after CRC diagnosis is feasible and has beneficial effects on HRQoL irrespective of the timing of chemotherapy or surgery [55]. The role of physical activity in anxiety, depression, sleep quality, and fatigue in lung cancer is also favorable. Moreover, structured exercise with or without nutrition therapy has been shown to reduce cancer-related fatigue and improve QoL in patients with prostate cancer [56, 57].

For hematological malignancies, hematopoietic stem cell transplantation (HSCT) is a standard and curative treatment for some of these illnesses, and more than 50,000 HSCTs are performed annually worldwide. A meta-analysis of 10 RCTs of HSCT has demonstrated that exercise interventions have positive effects on reducing fatigue and improving HRQoL and muscle strength, even if initiated before transplant hospitalization (the concept of “pre-hab”) [58]. Based on that data, Mohananey et al. [59] proposed a model of cardiac rehabilitation and exercise in patients undergoing HSCT that included aerobic and strength training.

4. Optimal Timing of Physical Exercise and Prescription Approach
4.1 Optimal Timing of Physical Exercise

The role of cardiac rehabilitation in cancer patients is well established in current cardio-oncology clinical practice guidelines for pre-, active-, and post-specific treatments [60]. The optimal timing of physical exercise interventions to obtain the expected benefits of physical activity and mitigate chemotherapy-induced side effects is not well defined. However, the current evidence supports the benefit of physical exercise throughout the disease. The concept of “pre-habilitation” has been considered before HSCT and breast cancer treatment, with a significant positive impact observed [57, 61]. Physical exercise to improve HRQoL and diastolic and systolic left ventricular function parameters, reduce fatigue, and increase VO2peak is also favorable during chemotherapy for breast and colon cancers [49, 62]. Van der Schoot et al. [63] investigated the impact of exercise intervention during or after chemotherapy on VO2peak in the breast, testis, and CRC. Although immediately post-chemotherapy, the decline of VO2peak was less in the exercise group than during chemotherapy, no differences were found at 1 year post-intervention. Recommendations for physical activity for survivors of breast, CRC, and prostate cancers are unequivocal; however, evidence for long-term gynecologic cancers is limited [64].

4.2 Approach to Physical Exercise Prescription

Accurate exercise prescription is mandatory to obtain the expected benefits of reducing CV risk and mortality, increasing capacity functional respiratory (CFR), and improving psychosocial well-being. Initially, cancer survivors should receive a comprehensive assessment of all components of health-related physical fitness (e.g., CRF, muscle strength and endurance, body composition, and flexibility), with cancer-specific considerations such as the disease stage and toxicities, in order to individualize the exercise prescription. Moreover, during active cancer therapy, an individual’s ability to tolerate exercise may fluctuate from day to day [65].

For the adequate evaluation of these patients, ergospirometry (cardiopulmonary exercise testing) provides an assessment of integrative exercise responses involving pulmonary, CV, neuropsychological, and skeletal muscle systems [66]. It also involves measurements of VO2peak, carbon dioxide production, and ventilation during a symptom-limited exercise test [67].

Global recommendations in cancer patients include aerobic and resistance exercises, warm-up and cool-down activities, and flexibility stretching exercises [8]. An effective exercise prescription includes moderate-intensity aerobic training at least three times per week for at least 30 min (90–150 min per week), for a minimum of 8–12 weeks. Moderate-intensity aerobic training reduces anxiety, depressive symptoms, improves HRQoL, bone health, and sleep in cancer patients. Moreover, it improves the lipid profile, lowers hypertension, and provides CV benefits [68, 69]. Resistance training added to aerobic exercise should be done two times per week, using no less than two sets of 8–15 repetitions with at least 60% of one repetition maximum [7]. However, apart from physical exercise, it is necessary to provide the patient with a comprehensive long-term service including medical evaluation, prescriptive exercise, and modification of cardiac risk factors and education [70].

The goals of cardio-oncology rehabilitation include increasing functional capacity and reducing morbidity/mortality, attenuating the drop in LVEF, decreasing cancer-related fatigue, and improving QoL and psychosocial well-being (Fig. 2). Multidisciplinary teams supported by cardio-oncology units working with different healthcare specialists and aided by advanced cardiac imaging techniques and ergospirometry are required.

Fig. 2.

Benefits of physical exercise in cancer patients. Physical exercise increases functional capacity, reduces cardiovascular mortality, decreases cancer-related fatigue, and improves quality of life. Current evidence suggests that exercise prevents cancer therapy-related cardiac dysfunction, although more evidence is needed. VO2peak, oxygen consumption peak; CV, cardiovascular.

5. Conclusions

Physical exercise has been demonstrated to provide clinical and emotional benefits to cancer patients. The evidence is reliable for the positive effect of exercise programs in reducing cancer-related fatigue and improving HRQoL, anxiety, depression, and well-being in all cancer populations. The exercise is established as a safe and effective strategy to increase CRF and potentially reduce CV mortality in cancer patients in the medium and long term. Furthermore, preclinical evidence and available clinical data suggest that exercise prevents CTRCD, although more rigorous and extensive RCTs are required, leading to an emerging field of investigation. To obtain the maximum benefits from exercise, this should be individualized according to the patient’s functional capacity, type and stage of cancer, and treatment approach. Moreover, multidisciplinary teams as well as reassessment and continued monitoring of patients during follow-up are critical.

Author Contributions

NC and SM designed the research study. NC, MV and EP performed the research. AE and RB provided help and advice on the data analysis. EB assisted in the analysis and interpretation of the obtained data. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This research received no external funding.

Conflict of Interest

The author declares no conflict of interest. Sergio Moral is serving as Guest Editor of this journal. We declare that Sergio Moral had no involvement in the peer review of this article and has no access to information regarding its peer review. Full responsibility for the editorial process for this article was delegated to Peter Kokkinos.

References
[1]
Nonaka M, Hosoda H, Uezono Y. Cancer treatment-related cardiovascular disease: Current status and future research priorities. Biochemical Pharmacology. 2021; 190: 114599.
[2]
Kobo O, Khattak S, Lopez-Mattei J, Van Spall HGC, Graham M, Cheng RK, et al. Trends in cardiovascular mortality of cancer patients in the US over two decades 1999-2019. International Journal of Clinical Practice. 2021; 75: e14841.
[3]
Haykowsky MJ, Beaudry R, Brothers RM, Nelson MD, Sarma S, La Gerche A. Pathophysiology of exercise intolerance in breast cancer survivors with preserved left ventricular ejection fraction. Clinical Science. 2016; 130: 2239–2244.
[4]
Cramer L, Hildebrandt B, Kung T, Wichmann K, Springer J, Doehner W, et al. Cardiovascular function and predictors of exercise capacity in patients with colorectal cancer. Journal of the American College of Cardiology. 2014; 64: 1310–1319.
[5]
Zamorano JL, Lancellotti P, Muñoz DR, Aboyans V, Asteggiano R, Galderisi M, et al. 2016 ESC Position Paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines. Kardiologia Polska. 2016; 74: 1193–1233. (In Polish)
[6]
Koene RJ, Prizment AE, Blaes A, Konety SH. Shared Risk Factors in Cardiovascular Disease and Cancer. Circulation. 2016; 133: 1104–1114.
[7]
Campbell KL, Winters-Stone KM, Wiskemann J, May AM, Schwartz AL, Courneya KS, et al. Exercise Guidelines for Cancer Survivors: Consensus Statement from International Multidisciplinary Roundtable. Medicine and Science in Sports and Exercise. 2019; 51: 2375–2390.
[8]
Gilchrist SC, Barac A, Ades PA, Alfano CM, Franklin BA, Jones LW, et al. Cardio-Oncology Rehabilitation to Manage Cardiovascular Outcomes in Cancer Patients and Survivors: A Scientific Statement From the American Heart Association. Circulation. 2019; 139: e997–e1012.
[9]
Ramin C, Schaeffer ML, Zheng Z, Connor AE, Hoffman-Bolton J, Lau B, et al. All-Cause and Cardiovascular Disease Mortality Among Breast Cancer Survivors in CLUE II, a Long-Standing Community-Based Cohort. Journal of the National Cancer Institute. 2021; 113: 137–145.
[10]
Zhao J, Su L, Zhong J. Risk Factors for Cancer-specific Mortality and Cardiovascular Mortality in Patients With Diffuse Large B-cell Lymphoma. Clinical Lymphoma, Myeloma & Leukemia. 2020; 20: e858–e863.
[11]
Sturgeon KM, Deng L, Bluethmann SM, Zhou S, Trifiletti DM, Jiang C, et al. A population-based study of cardiovascular disease mortality risk in US cancer patients. European Heart Journal. 2019; 40: 3889–3897.
[12]
Jones LW, Courneya KS, Mackey JR, Muss HB, Pituskin EN, Scott JM, et al. Cardiopulmonary function and age-related decline across the breast cancer survivorship continuum. Journal of Clinical Oncology. 2012; 30: 2530–2537.
[13]
Gong J, Payne D, Caron J, Bay CP, McGregor BA, Hainer J, et al. Reduced Cardiorespiratory Fitness and Increased Cardiovascular Mortality After Prolonged Androgen Deprivation Therapy for Prostate Cancer. JACC: CardioOncology. 2020; 2: 553–563.
[14]
Fardman A, Banschick GD, Rabia R, Percik R, Fourey D, Segev S, et al. Cardiorespiratory fitness and survival following cancer diagnosis. European Journal of Preventive Cardiology. 2021; 28: 1242–1249.
[15]
Imboden MT, Harber MP, Whaley MH, Finch WH, Bishop DL, Kaminsky LA. Cardiorespiratory Fitness and Mortality in Healthy Men and Women. Journal of the American College of Cardiology. 2018; 72: 2283–2292.
[16]
Sadeghi M, Keshavarz-Fathi M, Baracos V, Arends J, Mahmoudi M, Rezaei N. Cancer cachexia: Diagnosis, assessment, and treatment. Critical Reviews in Oncology/hematology. 2018; 127: 91–104.
[17]
Rausch V, Sala V, Penna F, Porporato PE, Ghigo A. Understanding the common mechanisms of heart and skeletal muscle wasting in cancer cachexia. Oncogenesis. 2021; 10: 1.
[18]
Slamon D, Eiermann W, Robert N, Pienkowski T, Martin M, Press M, et al. Adjuvant trastuzumab in HER2-positive breast cancer. The New England Journal of Medicine. 2011; 365: 1273–1283.
[19]
Suter TM, Procter M, van Veldhuisen DJ, Muscholl M, Bergh J, Carlomagno C, et al. Trastuzumab-associated cardiac adverse effects in the herceptin adjuvant trial. Journal of Clinical Oncology. 2007; 25: 3859–3865.
[20]
Farmakis D, Mantzourani M, Filippatos G. Anthracycline-induced cardiomyopathy: secrets and lies. European Journal of Heart Failure. 2018; 20: 907–909.
[21]
Herrmann J, Lenihan D, Armenian S, Barac A, Blaes A, Cardinale D, et al. Defining cardiovascular toxicities of cancer therapies: an International Cardio-Oncology Society (IC-OS) consensus statement. European Heart Journal. 2022; 43: 280–299.
[22]
Minotti G, Menna P, Camilli M, Salvatorelli E, Levi R. Beyond hypertension: Diastolic dysfunction associated with cancer treatment in the era of cardio-oncology. Advances in Pharmacology. 2022; 94: 365–409.
[23]
Minotti G, Reggiardo G, Camilli M, Salvatorelli E, Menna P. From Cardiac Anthracycline Accumulation to Real-Life Risk for Early Diastolic Dysfunction: A Translational Approach. JACC: CardioOncology. 2022; 4: 139–140.
[24]
Upshaw JN, Finkelman B, Hubbard RA, Smith AM, Narayan HK, Arndt L, et al. Comprehensive Assessment of Changes in Left Ventricular Diastolic Function With Contemporary Breast Cancer Therapy. JACC: Cardiovascular Imaging. 2020; 13: 198–210.
[25]
Thong MSY, van Noorden CJF, Steindorf K, Arndt V. Correction to: Cancer-Related Fatigue: Causes and Current Treatment Options. Current Treatment Options in Oncology. 2022; 23: 450–451.
[26]
Kuiper JG, Phipps AI, Neuhouser ML, Chlebowski RT, Thomson CA, Irwin ML, et al. Recreational physical activity, body mass index, and survival in women with colorectal cancer. Cancer Causes & Control. 2012; 23: 1939–1948.
[27]
Beasley JM, Kwan ML, Chen WY, Weltzien EK, Kroenke CH, Lu W, et al. Meeting the physical activity guidelines and survival after breast cancer: findings from the after breast cancer pooling project. Breast Cancer Research and Treatment. 2012; 131: 637–643.
[28]
Cormie P, Zopf EM, Zhang X, Schmitz KH. The Impact of Exercise on Cancer Mortality, Recurrence, and Treatment-Related Adverse Effects. Epidemiologic Reviews. 2017; 39: 71–92.
[29]
Kim KH, Choi S, Kim K, Chang J, Kim SM, Kim SR, et al. Association between physical activity and subsequent cardiovascular disease among 5-year breast cancer survivors. Breast Cancer Research and Treatment. 2021; 188: 203–214.
[30]
Scott JM, Zabor EC, Schwitzer E, Koelwyn GJ, Adams SC, Nilsen TS, et al. Efficacy of Exercise Therapy on Cardiorespiratory Fitness in Patients With Cancer: A Systematic Review and Meta-Analysis. Journal of Clinical Oncology. 2018; 36: 2297–2305.
[31]
Lyon AR, Dent S, Stanway S, Earl H, Brezden-Masley C, Cohen-Solal A, et al. Baseline cardiovascular risk assessment in cancer patients scheduled to receive cardiotoxic cancer therapies: a position statement and new risk assessment tools from the Cardio-Oncology Study Group of the Heart Failure Association of the European Society of Cardiology in collaboration with the International Cardio-Oncology Society. European Journal of Heart Failure. 2020; 22: 1945–1960.
[32]
Lee K, Kang I, Mack WJ, Mortimer J, Sattler F, Salem G, et al. Effects of high-intensity interval training on vascular endothelial function and vascular wall thickness in breast cancer patients receiving anthracycline-based chemotherapy: a randomized pilot study. Breast Cancer Research and Treatment. 2019; 177: 477–485.
[33]
Beaudry RI, Liang Y, Boyton ST, Tucker WJ, Brothers RM, Daniel KM, et al. Meta-analysis of Exercise Training on Vascular Endothelial Function in Cancer Survivors. Integrative Cancer Therapies. 2018; 17: 192–199.
[34]
Mclaughlin M, Florida-James G, Ross M. Breast cancer chemotherapy vascular toxicity: a review of mediating mechanisms and exercise as a potential therapeutic. Vascular Biology. 2021; 3: R106–R120.
[35]
Siahpush M, Farazi PA, Wang H, Robbins RE, Singh GK, Su D. Muscle-strengthening physical activity is associated with cancer mortality: results from the 1998-2011 National Health Interview Surveys, National Death Index record linkage. Cancer Causes & Control. 2019; 30: 663–670.
[36]
Gorzelitz J, Trabert B, Katki HA, Moore SC, Watts EL, Matthews CE. Independent and joint associations of weightlifting and aerobic activity with all-cause, cardiovascular disease and cancer mortality in the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial. British Journal of Sports Medicine. 2022; 56: 1277–1283.
[37]
Momma H, Kawakami R, Honda T, Sawada SS. Muscle-strengthening activities are associated with lower risk and mortality in major non-communicable diseases: a systematic review and meta-analysis of cohort studies. British Journal of Sports Medicine. 2022; 56: 755–763.
[38]
Toohey K, Pumpa K, McKune A, Cooke J, Welvaert M, Northey J, et al. The impact of high-intensity interval training exercise on breast cancer survivors: a pilot study to explore fitness, cardiac regulation and biomarkers of the stress systems. BMC Cancer. 2020; 20: 787.
[39]
Kang DW, Fairey AS, Boulé NG, Field CJ, Wharton SA, Courneya KS. Effects of Exercise on Cardiorespiratory Fitness and Biochemical Progression in Men With Localized Prostate Cancer Under Active Surveillance: The ERASE Randomized Clinical Trial. JAMA Oncology. 2021; 7: 1487–1495.
[40]
Lien CY, Jensen BT, Hydock DS, Hayward R. Short-term exercise training attenuates acute doxorubicin cardiotoxicity. Journal of Physiology and Biochemistry. 2015; 71: 669–678.
[41]
Marques-Aleixo I, Santos-Alves E, Mariani D, Rizo-Roca D, Padrão AI, Rocha-Rodrigues S, et al. Physical exercise prior and during treatment reduces sub-chronic doxorubicin-induced mitochondrial toxicity and oxidative stress. Mitochondrion. 2015; 20: 22–33.
[42]
Farzanegi P, Asadi M, Abdi A, Etemadian M, Amani M, Amrollah V, et al. Swimming exercise in combination with garlic extract administration as a therapy against doxorubicin-induced hepatic, heart and renal toxicity to rats. Toxin Reviews. 2020; 39: 434–443.
[43]
Parry TL, Hayward R. Exercise Protects against Cancer-induced Cardiac Cachexia. Medicine and Science in Sports and Exercise. 2018; 50: 1169–1176.
[44]
Kirkham AA, Shave RE, Bland KA, Bovard JM, Eves ND, Gelmon KA, et al. Protective effects of acute exercise prior to doxorubicin on cardiac function of breast cancer patients: A proof-of-concept RCT. International Journal of Cardiology. 2017; 245: 263–270.
[45]
Kirkham AA, Virani SA, Bland KA, McKenzie DC, Gelmon KA, Warburton DER, et al. Exercise training affects hemodynamics not cardiac function during anthracycline-based chemotherapy. Breast Cancer Research and Treatment. 2020; 184: 75–85.
[46]
Ma Z. Effect of anthracycline combined with aerobic exercise on the treatment of breast cancer. Pakistan Journal of Pharmaceutical Sciences. 2018; 31: 1125–1129.
[47]
Haykowsky MJ, Mackey JR, Thompson RB, Jones LW, Paterson DI. Adjuvant trastuzumab induces ventricular remodeling despite aerobic exercise training. Clinical Cancer Research. 2009; 15: 4963–4967.
[48]
Hojan K, Procyk D, Horyńska-Kęstowicz D, Leporowska E, Litwiniuk M. The Preventive role of Regular Physical Training in Ventricular Remodeling, Serum Cardiac Markers, and Exercise Performance Changes in Breast Cancer in Women Undergoing Trastuzumab Therapy-An REH-HER Study. Journal of Clinical Medicine. 2020; 9: 1379.
[49]
Peck SS, Esmaeilzadeh M, Rankin K, Shalmon T, Fan CPS, Somerset E, et al. Self-Reported Physical Activity, QoL, Cardiac Function, and Cardiorespiratory Fitness in Women With HER2+ Breast Cancer. JACC: CardioOncology. 2022; 4: 387–400.
[50]
Mustian KM, Alfano CM, Heckler C, Kleckner AS, Kleckner IR, Leach CR, et al. Comparison of Pharmaceutical, Psychological, and Exercise Treatments for Cancer-Related Fatigue: A Meta-analysis. JAMA Oncology. 2017; 3: 961–968.
[51]
Sweegers MG, Altenburg TM, Chinapaw MJ, Kalter J, Verdonck-de Leeuw IM, Courneya KS, et al. Which exercise prescriptions improve quality of life and physical function in patients with cancer during and following treatment? A systematic review and meta-analysis of randomised controlled trials. British Journal of Sports Medicine. 2018; 52: 505–513.
[52]
Belloni S, Arrigoni C, Caruso R. Effects from physical exercise on reduced cancer-related fatigue: a systematic review of systematic reviews and meta-analysis. Acta Oncologica. 2021; 60: 1678–1687.
[53]
Juvet LK, Thune I, Elvsaas IKØ, Fors EA, Lundgren S, Bertheussen G, et al. The effect of exercise on fatigue and physical functioning in breast cancer patients during and after treatment and at 6 months follow-up: A meta-analysis. Breast. 2017; 33: 166–177.
[54]
Brown JC, Damjanov N, Courneya KS, Troxel AB, Zemel BS, Rickels MR, et al. A randomized dose-response trial of aerobic exercise and health-related quality of life in colon cancer survivors. Psycho-oncology. 2018; 27: 1221–1228.
[55]
Singh B, Hayes SC, Spence RR, Steele ML, Millet GY, Gergele L. Exercise and colorectal cancer: a systematic review and meta-analysis of exercise safety, feasibility and effectiveness. The International Journal of Behavioral Nutrition and Physical Activity. 2020; 17: 122.
[56]
Avancini A, Sartori G, Gkountakos A, Casali M, Trestini I, Tregnago D, et al. Physical Activity and Exercise in Lung Cancer Care: Will Promises Be Fulfilled? The Oncologist. 2020; 25: e555–e569.
[57]
Baguley BJ, Bolam KA, Wright ORL, Skinner TL. The Effect of Nutrition Therapy and Exercise on Cancer-Related Fatigue and Quality of Life in Men with Prostate Cancer: A Systematic Review. Nutrients. 2017; 9: 1003.
[58]
Liang Y, Zhou M, Wang F, Wu Z. Exercise for physical fitness, fatigue and quality of life of patients undergoing hematopoietic stem cell transplantation: a meta-analysis of randomized controlled trials. Japanese Journal of Clinical Oncology. 2018; 48: 1046–1057.
[59]
Mohananey D, Sarau A, Kumar R, Lewandowski D, Abreu-Sosa SM, Nathan S, et al. Role of Physical Activity and Cardiac Rehabilitation in Patients Undergoing Hematopoietic Stem Cell Transplantation. JACC: CardioOncology. 2021; 3: 17–34.
[60]
Lyon AR, López-Fernández T, Couch LS, Asteggiano R, Aznar MC, Bergler-Klein J, et al. 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). European Heart Journal. 2022; 43: 4229–4361.
[61]
Sturgeon KM, Ky B, Libonati JR, Schmitz KH. The effects of exercise on cardiovascular outcomes before, during, and after treatment for breast cancer. Breast Cancer Research and Treatment. 2014; 143: 219–226.
[62]
Van Vulpen JK, Velthuis MJ, Steins Bisschop CN, Travier N, Van Den Buijs BJW, Backx FJG, et al. Effects of an Exercise Program in Colon Cancer Patients undergoing Chemotherapy. Medicine and Science in Sports and Exercise. 2016; 48: 767–775.
[63]
van der Schoot GGF, Ormel HL, Westerink NDL, May AM, Elias SG, Hummel YM, et al. Optimal Timing of a Physical Exercise Intervention to Improve Cardiorespiratory Fitness: During or After Chemotherapy. JACC: CardioOncology. 2022; 4: 491–503.
[64]
Rock CL, Thomson CA, Sullivan KR, Howe CL, Kushi LH, Caan BJ, et al. American Cancer Society nutrition and physical activity guideline for cancer survivors. CA: A Cancer Journal for Clinicians. 2022; 72: 230–262.
[65]
Fakhraei R, Peck BKin SS, Abdel-Qadir H, Thavendiranathan P, Sabiston CM, Rivera-Theurel F, et al. Research Quality and Impact of Cardiac Rehabilitation in Cancer Survivors: A Systematic Review and Meta-Analysis. JACC: CardioOncology. 2022; 4: 195–206.
[66]
Albouaini K, Egred M, Alahmar A, Wright DJ. Cardiopulmonary exercise testing and its application. Postgraduate Medical Journal. 2007; 83: 675–682.
[67]
Wasserman K, Hansen JE, Sue DY, Stringer WW, Whipp BJ. Principles of Exercise Testingand Interpretation: Including Pathophysiology and Clinical Applications (pp. 585). 4th edn. Lippincott Williams & Wilkins: Philadelphia. 2004.
[68]
Wang Y, Xu D. Effects of aerobic exercise on lipids and lipoproteins. Lipids in Health and Disease. 2017; 16: 132.
[69]
Saco-Ledo G, Valenzuela PL, Ruiz-Hurtado G, Ruilope LM, Lucia A. Exercise Reduces Ambulatory Blood Pressure in Patients With Hypertension: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Journal of the American Heart Association. 2020; 9: e018487.
[70]
Wenger NK, Froelicher ES, Smith LK, Ades PA, Berra K, Blumenthal JA, et al. Cardiac rehabilitation as secondary prevention. Agency for Health Care Policy and Research and National Heart, Lung, and Blood Institute. Clinical Practice Guideline: Quick Reference Guide for Clinicians. 1995; 1–23.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top