Skip to main content
Erschienen in: Clinical and Translational Oncology 10/2020

Open Access 13.02.2020 | Special Article

Exercise and cancer: a position statement from the Spanish Society of Medical Oncology

verfasst von: M. Pollán, S. Casla-Barrio, J. Alfaro, C. Esteban, M. A. Segui-Palmer, A. Lucia, M. Martín

Erschienen in: Clinical and Translational Oncology | Ausgabe 10/2020

Abstract

Due to improvements in the number of cancer survivors and survival time, there is a growing interest in healthy behaviors, such as physical activity (PA), and their potential impact on cancer- and non-cancer-related morbidity in individuals with cancer. Commissioned by the Spanish Society of Medical Oncology (SEOM), in this review, we sought to distill the most recent evidence on this topic, focusing on the mechanisms that underpin the effects of PA on cancer, the role of PA in cancer prevention and in the prognosis of cancer and practical recommendations for clinicians regarding PA counseling. Despite the available information, the introduction of exercise programs into the global management of cancer patients remains a challenge with several areas of uncertainty. Among others, the most effective behavioral interventions to achieve long-term changes in a patient’s lifestyle and the optimal intensity and duration of PA should be defined with more precision in future studies.
Hinweise
M. Pollán and S. Casla-Barrio have equally contributed to this work.

Publisher's Note

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

Introduction

Regular and adequate physical activity (PA) is associated with key benefits to human health, such as improvements in weight control, muscular and cardiorespiratory fitness, bone and functional health and a reduced risk of falls and several noncommunicable diseases, including diabetes, cardiovascular disease, depression and some cancers [1].
Due to improvements in the management of cancer, the number of cancer survivors and survival time are increasing. Consequently, interest in healthy behaviors, such as PA, and their potential impact on cancer- and non-cancer-related morbidity in these individuals has rapidly increased [2].
Commissioned by the Spanish Society of Medical Oncology (SEOM), in this review, we sought to distill the most recent evidence on this topic, focusing on the mechanisms that underpin the effects of PA on cancer, describing the role of PA in cancer prevention and prognosis, and providing practical recommendations to clinicians on managing PA counseling.

Biological mechanisms underpinning the potential anticancer effects of exercise

Of note, PA is any bodily movement produced by skeletal muscles that requires energy expenditure, whereas exercise is a subset of PA that is planned, structured and repetitive and that has a final or an intermediate objective of improving or maintaining physical fitness. The epidemiological evidence regarding the risk of some cancers mainly refers to regular PA (usually self-reported, i.e., through questionnaires). Thus, regular exercise or “exercise training” is a proxy, but not a perfect surrogate, for PA and is thought to induce more profound molecular adaptations than PA.
There is growing evidence from preclinical research that regular exercise can influence cancer development or the rate of tumor growth once malignancy has initiated. For instance, a recent meta-analysis of 28 preclinical studies in breast tumors (n = 2085 animals) found large favorable effects for exercise training on proliferation and apoptosis [3]. Exercise is also emerging as a potential coadjuvant treatment; when combined with cyclophosphamide, exercise delays murine breast tumor growth versus chemotherapy alone [4], and similar findings have been reported for exercise combined with anthracyclines [57]. There is, however, heterogeneity among studies in the tumor models used, ranging from tumor transplant (where there is also substantial variability, e.g., syngeneic versus xenograft models) to carcinogen-induced or genetically engineered mouse models [8]. The type of exercise training to which mice are typically subjected to before/upon tumor inoculation also varies between studies (forced treadmill, forced swimming, voluntary wheel running). The duration of exercise in the trials typically consists of several weeks (~ 4 to 10 weeks), which can be translated to human “years.”
Exercise can have an impact on tumor development, growth or dissemination through several mechanisms. First, exercise might help to prevent cancer by reducing the circulating levels of several mediators, such as insulin growth factor-1 (IGF-1) [914], a mitogen that triggers cell proliferation [15]. Exercise can also reduce the levels of hyperphosphorylated retinoblastoma protein (Rb) in a chemically induced rat model of mammary carcinogenesis [16, 17], increase ß-catenin phosphorylation in colon polyps [18, 19], and reduce the levels of micro-RNA 21 [20].
Exercise can upregulate tumor suppressors, such as the tumor suppressor programmed cell death protein 4 in a murine model of estrogen receptor-positive breast cancer (BC) [21]. In addition, exercise-induced catecholamines might reduce BC development through activation of the Hippo tumor suppressor pathway [22] and exercise-induced increased p53 activation, leading to tumor prevention, as shown in mouse models of skin [13, 23] and lung [24] cancer.
Exercise training can stimulate apoptosis, as shown in xenograft models of lung adenocarcinoma [24] and human pancreatic and prostate cancers [25, 26] and in murine models of skin tumorigenesis [12] and mammary carcinoma [4, 16, 27]. Exercise also exerts proapoptotic effects on cultured prostate cancer cells [28], estrogen receptor-positive BC cells [29, 30] and lymph node metastases of prostate tumor cells [25]. Exercise additionally reduces the levels of the antiapoptotic protein B cell lymphoma 2 [16, 31] and stimulates the proapoptotic proteins Bax and Bak [4, 16, 24] and the protein kinase AMPK [32, 33].
Hypoxia and poor blood supply promote an aggressive cancer phenotype and contribute to ineffective systemic therapy [34]. In this respect, exercise may promote a shift toward a more “normalized” tumor microenvironment by improving intratumoral perfusion/vascularization, at least in orthotopic murine models of human BC [4, 35] and prostate cancer [3638] and in xenografts of different tumors (melanoma, pancreas) [39].
Exercise might also attenuate the development of metastases. Mouse exercise training can decrease catenin while increasing E-cadherin inside tumors [19, 40]. Importantly, cadherins act as glue between epithelial cells, and their loss can favor malignancy by allowing the disaggregation of cells, which can then invade locally or metastasize [40]. Moderate-intensity mouse swimming can suppress liver cancer metastases via boosting the activity of dopamine receptor 2 [40]. Exercise may also modulate blood–brain barrier integrity by maintaining the expression levels of occludin or claudin-5 proteins [41], thereby preventing metastatic progression to the brain [42]. On the other hand, inflammatory cells within the tumor microenvironment supply bioactive molecules that sustain cancer hallmarks [4345]. In this context, mouse exercise training decreases macrophage infiltration in allogeneic lymphoid tumors [46], Ehrlich tumor cells [47] and colon polyps [18].
One major potential “anticancer” effect of exercise lies in an enhancement of immune function [43, 48]. At moderate intensities, exercise can stimulate the innate immune system, especially natural killer (NK) cells [49, 50]. A 6-week mouse wheel running program had preventive effects against the development of several types of tumors (melanoma, liver and lung cancers), and the effect was mediated by improved NK cell infiltration into the tumors, which in turn was mediated by the enhanced tumor expression of ligands for several NK cell-activating receptors [51]. A previous study showed that exercise training increased the cytolytic capacity of resident peritoneal macrophages against mastocytoma cells [52]. Mouse exercise training could also polarize the immunological response toward an efficient “antitumor” macrophage profile 1, which is linked to the production of T-helper 1 cytokines [5256]. Short-term (6-day) moderate exercise before the injection of melanoma cells into mice decreased their metastatic spread, which was partially mediated by increased antitumor macrophage cytotoxicity [57]. Preliminary data from mice [53, 5860] and cancer patients suggest that exercise training may help to reduce the immunosuppressive effects of T regulatory lymphocytes [61]. Finally, regular exercise can increase alveolar macrophage antitumor cytotoxicity in vitro, which would mediate a protective effect against mouse lung metastases [62, 63].
Importantly, skeletal muscle, especially during contractions, releases molecules collectively known as “myokines” into the bloodstream, which act systemically to induce a myriad of health-promoting effects, such as decreased inflammation and reduced insulin resistance [64]. Some myokines might also induce direct anticancer effects (via the stimulation of apoptosis in tumor cells), such as oncostatin M in hormone-sensitive BC [30] or secreted protein acidic and rich in cysteine (SPARC, also known as osteonectin) in colon cancer [65]. The aforementioned exercise-induced infiltration of NK cells into tumors seems to be mediated by the release of interleukin 6 by muscle into the bloodstream [49, 51, 66].

Physical activity and cancer prevention

According to the World Health Organization (WHO), up to 31% of the adult population worldwide and 35% in Europe are physically inactive [67]. PA is difficult to measure for the following reasons: (1) there are at least four domains: occupational, household, transportation and leisure time; (2) PA questionnaires on past and current activity are subject to recall bias; and (3) objective methods (pedometers or accelerometers) can only be used in prospective studies for short time periods, and they may not always represent overall PA. Fortunately, smartphones and other devices now allow PA monitoring and will hopefully provide more accurate measures in the future.
Body mass is related to PA and cancer risk, acting as a confounder. However, the prevention of adiposity may mediate the relationship between PA and cancer, and controlling for adiposity could lead to underestimating the real effect of PA [68].
Of note, 1 metabolic equivalent (MET) is the rate of energy expenditure while resting or 3.5 ml O2/kg body weight/min on average. Moderate PA (e.g., brisk walking) usually requires an energy expenditure of 3–6 MET, whereas vigorous PA (e.g., jogging) requires an energy expenditure above 6 MET. The WHO recommends that adults engage in ≥ 150 min/week of moderate PA or ≥ 75 min/week of vigorous PA or a combination thereof. If a person does an ~ 3 MET activity (e.g., brisk walking on a level surface) for 1 h, he or she has done 3 MET-hours of PA. If this person does this same PA on every day of the week, he/she has done 21 (= 3 × 7) MET-hours/week. If a person does an ~ 8 MET activity (e.g., jogging) for 1 h on each day of the week, he/she has done 56 (= 8 × 7) MET-hours/week.
The World Cancer Research Fund and the American Institute for Cancer Research periodically publish the conclusions of a panel reviewing evidence linking food, nutrition and PA with cancer risk [69]. The evidence is classified as follows: (1) convincing: available results support a causal relationship; (2) probable: evidence supports a probable causal relationship; and (3) limited: results are not considered sufficient to rate the relationship as convincing or probable. In the last category, a distinction is made between limited-suggestive evidence when an effect is reported but there were methodological problems and limited-not conclusive evidence when there were insufficient data and/or the results were too heterogeneous. The panel concluded that regular, sustained PA protects against several types of cancer independent of body fat [69]. This evidence comes from high-income countries and is mainly based on leisure-time PA. The three tumors with the most solid results are colon, postmenopausal BC and endometrial.

Colorectal cancer

The evidence for colon cancer is judged as “convincing,” with an overall risk reduction of approximately 20% in the most physically active group compared with the less active group [70]. The effect is weaker or absent for rectal cancer. However, a pooled analysis of 12 prospective cohort studies with information on leisure-time PA at baseline compared the group at the 90th percentile of PA with the group under the 10th percentile and showed a reduced incidence of both colon (13% reduction) and rectal cancer (12%) after controlling for body mass index (BMI) [71]. Regarding the amount of PA required to obtain maximum benefit, a cohort of more than 40,000 men in the USA (The Health Professionals Follow-up Study) showed that aerobic PA seems to be more beneficial and that overall PA is more relevant than the intensity of PA [72]. Finally, while a benefit was observed in men meeting current guidelines (17% risk reduction), the maximum benefit (32% risk reduction) was observed for PA ≥ 30 metabolic equivalents (MET)-hours/week, which is equivalent to 10 h or more of walking/week [72]. A meta-analysis evaluating the dose–response shape of PA for different endpoints, including colon cancer, showed that major gains occurred at lower levels of activity (up to 50 MET-hours/week), while a decrease in risk was minimal at levels higher than 50–65 MET-hours/week [73].

Breast cancer

The evidence for postmenopausal BC is judged as “probable” [74]. Most studies show a protective effect with a 13% decreased risk in high versus low PA groups [74]. For recreational PA, a nonlinear dose–response was observed with a greater decrease in the risk for PA activity at > 20 MET-hours/week [74]. The pooling analysis with information on baseline leisure PA showed a reduction of 7% in the incidence of BC between the 90th and the 10th percentiles of PA [71]. Evidence for PA in premenopausal BC was rated as “limited-suggestive” for total PA and as “probable” for vigorous-intensity PA [74]. In Canada, a cohort study with 39,000 women reported a clear downward trend of BC incidence based on the number of MET-hours/week, which was mainly due to the risk reduction observed for premenopausal tumors [75]. Finally, a case–control study in Spain showed a reduced risk of 5% per 6 MET-hours/week [76]. The protection was particularly important for nulliparous women (12% risk reduction per 6 MET-hours/week) [76].

Endometrial cancer

The evidence for endometrial cancer was rated as “probable,” and the results showed a lower risk of endometrial cancer with higher levels of PA [77]. A meta-analysis reported a 20% risk reduction in high versus low PA groups [78]. This inverse association was only observed in overweight/obese women [78]. The pooled analysis of PA at baseline in 12 cohorts showed a risk reduction of 21% between the two extreme deciles of PA before taking BMI into account, while adjusting for BMI reduced the benefit to a nonsignificant risk reduction of 2% [71]. In the stratified analyses, PA was only associated with endometrial cancer in women with a BMI equal to or greater than 25 [71].

Lung cancer

A recent report classifies the evidence for lung cancer as “limited-suggestive” [69]. Leisure-time PA was considered in a systematic review, showing a clear inverse association with all histological lung cancer subtypes but only among former or current smokers [79]. The pooled analysis of cohort studies on leisure-time PA reported a 27% reduction in lung cancer incidence in the highest decile of PA compared with the lowest [71]. Again, the effect was only observed among smokers [71].

Liver cancer

The World Cancer Research Fund/American Institute for Cancer Research (WCRF/AICR) report classifies the evidence for liver cancer as “limited-suggestive” [80]. In the joint analysis of liver cancer incidence in 12 cohorts according to recreational PA at baseline, the highest decile had a hazard ratio (HR) of 0.73 before taking BMI into account and decreased to a nonsignificant HR of 0.81 when BMI was included as a confounder [71].

Esophageal cancer

There is limited but suggestive evidence of a protective effect of PA against esophageal adenocarcinoma and squamous cell carcinomas [81]. A meta-analysis found a risk ratio (RR) of 0.79 for esophageal adenocarcinoma and a nonsignificant RR of 0.94 for esophageal squamous cell carcinoma [82]. The pooled analyses of 1,44 million individuals from 12 cohorts showed an approximately 40% risk reduction in esophageal adenocarcinomas and a 24% risk reduction in esophageal squamous tumors for participants in the 90th percentile of leisure-time PA at baseline compared with the lowest PA group [71].

Stomach cancer

The recent WCRF/AICR update still considers limited-not conclusive evidence available for stomach cancer [83]. A previous meta-analysis on gastric cancer estimated an RR of 0.82 for high versus low PA [82]. The pooled analysis of leisure-time PA at baseline in the 12 cohorts showed a 22% risk reduction for gastric cardia tumors when BMI was not taken into account, but stratification by BMI showed that the protective effect was only observed among overweight/obese people [71].

Prostate cancer

There is a limited-not conclusive evidence of a link between PA and prostate cancer [84]. A systematic review showed substantial heterogeneity among 85 studies: 22 reported a statistically significant risk reduction, 25 reported a nonsignificant risk reduction, 31 did not find any association, and eight found an adverse effect of PA [85]. A higher incidence of prostate cancer (4% increase) was observed among the 10% more physically active participants in the pooled analysis of 12 cohort studies compared with those with a lower decile of activity. The authors hypothesized that this result could be due to a higher probability of prostate cancer screening in physically active men [71].

Ovarian cancer

The evidence for ovarian cancer was considered limited-not conclusive [86]. While most case–control studies found significant risk reductions among very active women, most cohort studies failed to show a clear effect [87]. The pooling analysis of 1.44 million participants in 12 prospective cohorts in the USA and Europe did not find a protective effect of high leisure-time PA for this tumor [71]. The Nurses’ Health Study, a prospective cohort with updated information on leisure-time PA, revealed an increased risk for both low and high levels of premenopausal PA, while no association was observed in postmenopausal women [88].

Pancreatic cancer

The WCRF/AICR report considers limited-not conclusive evidence for pancreatic cancer [89]. A meta-analysis yielded a statistically significant RR of 0.89 for high versus low PA [90]. Stronger effects were observed in case–control studies and for younger populations [90]. The pooling analysis of 12 cohort studies showed a non-statistically significant reduction of 5% in the most active group at baseline, but this effect was no longer observed when BMI was considered [71]. The EPIC-Norfolk cohort communicated a decreased risk in the highest category of total PA among participants younger than 60 years independent of BMI, while no effect was observed in older people [91].

Kidney cancer

The WCRF/AICR panel considers limited-not conclusive evidence for kidney cancer [92]; however, a meta-analysis in 2013 estimated a 12% risk reduction in the high PA group that was stronger when combining only high-quality studies [93]. The pooled analysis of the 12 cohorts showed a risk reduction of 16% independent of BMI among the most active group [71].

Bladder cancer

The evidence for bladder cancer is judged as limited-not conclusive [94]. A meta-analysis showed an RR of 0.85 for high versus low PA [95]. Moreover, the joint analysis of 12 prospective cohorts found a significantly reduced risk of bladder tumors in participants for the highest decile of leisure-time PA at baseline (HR = 0.88) [71].

Other tumors

A systematic review and meta-analysis on PA and hematologic cancers showed a reduced risk for non-Hodgkin lymphoma and nonsignificant results for multiple myeloma and leukemias [96]. The pooled analysis of 12 cohorts found a protective effect of PA against myeloid leukemia, myeloma and head–neck carcinomas [71]. Interestingly, malignant melanomas were more frequent in participants at the highest decile of leisure-time PA, a finding attributed to greater sun exposure due to outdoor activity and an increased risk of sunburn [71].

Summary and future directions

PA clearly reduces the risk of colon, BC and endometrial cancer. Furthermore, recent epidemiological studies suggest a protective effect for most cancer sites.
There is no conclusive evidence regarding the amount of PA needed to significantly reduce cancer risk, although it is likely tumor dependent.
New devices that routinely collect information on PA may help to increase the accuracy of PA measures and reduce information bias.

Effect of physical activity on the prognosis of cancer

Several reviews and meta-analyses of observational studies have suggested the benefit of PA on cancer outcomes. Most of the studies included breast cancer (Tables 1, 2) and colon cancer survivors (Table 3). A few studies have been conducted on patients with other types of neoplasms, such as prostate (Table 4), esophageal, lung and kidney cancer (Table 5). In these studies, PA is reported as lifetime PA in the latest years before or after diagnosis. The outcomes reported are usually overall survival, cancer-related survival, cancer recurrence and quality of life (QoL).
Table 1
Meta-analysis of observational and interventional studies on the impact of exercise on breast cancer outcome
References
Population
PA
Outcome
Results
Lahart et al. [97]
123,574 BC survivors
1994–2014
Most studies observational
Pre-diagnosis
All-cause mortality
HR 0.82 (95% CI 0.75–0.96)
BC mortality
HR 0.73 (95% CI 0.54–0.98)
BC events
HR 0.72 (95% CI 0.56–0.91)
After diagnosis
All-cause mortality
HR 0.52 (95% CI 0.43–0.64)
BC mortality
HR 0.59 (95% CI 0.45–0.78)
BC events
HR 0.79 (95% CI 0.63–0.98)
Lahart et al. [98]
5761 BC survivors from
63 randomized trials
PA intervention
After diagnosis
All-cause mortality
No data
BC recurrence
No data
HRQoL, emotional function, perceived physical function, anxiety, and cardiorespiratory fitness
Small to moderate improvement
BC, breast cancer; HRQoL, health-related quality of life; PA, physical activity
Table 2
Summary of prospective observational studies on physical activity and prognosis in breast cancer patients
Study/references
Population
LTPA
Outcome
Results
Holmes et al. [99]
Nurses’ Health Study
2987 Nurses with stage I-III BC, 1984–1998
After diagnosis
≥ 9 MET-h/week
BC-specific mortality
HR 0.50 (95% CI 0.34–0.74)
Irwin et al. [100]
HEAL Study
933 Women with BC 1995–1998
Pre-diagnosis
≥ 9 MET-h/week
Overall survival
HR 0.69 (95% CI 0.45–1.06)
After diagnosis
≥ 9 MET-h/week
Overall survival
HR 0.33 (95% CI 0.15–0.73)
Bao et al. [101]
Shanghai BCSS
518 Women with TNBC
After diagnosis
≥ 7.6 MET-h/week or ≥ 2.5 MET-h/week
BC-specific mortality
HR 0.58 (95% CI 0.39–0.86)
BC recurrence
HR 0.67 (95% CI 0.46–0.96)
Schmidt et al. [102]
Germany
3393 Women with early BC 50–74 year
Pre-diagnosis
≥ 42 MET-h/week
All-cause mortality
HR 0.66 (95% CI 0.47–0.92)
BC mortality
HR 0.80 (95% CI 0.53–1.21)
Cancer recurrence
HR 0.65 (95% CI 0.44–0.97)
Holick et al. [103]
Florida-Boston
4482 Invasive BC 1998–2001
After diagnosis
≥ 21 MET-h/week
BC mortality
HR 0.51 (95% CI 0.29–0.89)
All-cause mortality
HR 0.44 (95% CI 0.32–0.60)
Ammitzboll et al. [104]
Danish Diet, Cancer and Health Cohort
959 BC survivors
After diagnosis
≥ 8 MET-h/week
All-cause mortality
HR 0.67 (95% CI 0.47–0.99)
Friedenreich et al. [105]
Canadian
1233 BC survivors 1995–1997
Pre-diagnosis
46.9 MET-h/w
BC mortality
HR 0.56 (95% CI 0.38–0.82)
BC recurrence
HR 0.66 (95% CI 0.48–0.91)
Sternfeld et al. [106]
LACE Study
Multivariable
1970 BC survivors
PA 6 months prior to diagnosis
BC mortality
No association confirmed
BC recurrence
No association confirmed
All-cause mortality
HR 0.66 (95% CI 0.42–1.03)
Irwin et al. [107]
Women’s Health Initiative
4643 BC (in situ + invasive)
Prior to diagnosis
≥ 9 MET-h/week
All-cause mortality
HR 0.61 (95% CI 0.44–0.87)
After diagnosis
≥ 9 MET-h/week
BC mortality
HR 0.61 (95% CI 0.43–0.99)
All-cause mortality
HR 0.54 (95% CI 0.38–0.79)
Bertram et al. [108]
WHEL Study
2361 Women with stage I-III BC
Baseline active
All-cause mortality
HR 0.47 (95% CI 0.26–0.84)
 
BC events
No effect
Adherence to activity guidelines after 1 year post-diagnosis
All-cause mortality
HR 0.65 (95% CI 0.47–0.91)
BC events
No effect
Bradshaw et al. [109]
Long Island BC Study
1033 BC (in situ + invasive) 1995–1996
After diagnosis
≥ 9 MET-h/week
All-cause mortality
HR 0.33 (95% CI 0.22–0.48)
   
BC mortality
HR 0.27 (95% CI 0.15–0.46)
BC, breast cancer; LTPA, leisure-time physical activity; MET-h/week, metabolic equivalent task hours per week; TNBC: triple-negative breast cancer
BC events: BC progression, new primary BC, recurrence of BC
Table 3
Summary of observational studies on physical activity and prognosis in colorectal cancer patients
Study
Population
LTPA
Outcome
Results
Walter et al. [110]
3121 CRC patients
Latest LTPA
≥ 56 MET-h/week
Overall mortality
CRC mortality
HR 0.75 (95% CI 0.61–0.91)
HR 0.81 (95% CI 0.64–1.02)
Arem et al. [111]
AARP Diet and Health Study
3797 CRC patients
1759 CRC patients
Pre-diagnosis LTPA
> 7 MET-h/week
Post-diagnosis LTPA
> 7 MET-h/week
Overall mortality
Overall mortality
HR 0.80 (95% CI 0.68–0.95)
HR 0.69 (95% CI 0.49–0.98)
Meyerhardt et al. [112]
CALGB 89803
832 Patients with stage III CRC
Post-diagnosis LTPA
> 18 MET-h/week
Disease-free survival
HR 0.51 (95% CI 0.26–0.97)
van Blarigan et al. [113]
CALGB 89803
992 Patients with stage III colon cancer
Post-diagnosis LTPA
≥ 8.75 MET-h/week
Overall survival
HR 0.64 (95% CI 0.45–0.92)
Meyerhardt et al. [112]
Nurses’ Health Study
57 Women with stage I-III CRC
Post-diagnosis LTPA
> 18 MET-h/week
CRC mortality
Overall mortality
HR 0.39 (95% CI 0.18–0.82)
HR 0.43 (95% CI 0.25–0.74)
Campbell et al. [114]
2293 Patients with stage I-III CRC
Pre-diagnosis LTPA
≥ 8.75 MET-h/week
Post-diagnosis LTPA
≥ 8.75 MET-h/week
All-cause mortality
All-cause mortality
RR 0.72 (95% CI 0.58–0.89)
RR 0.58 (95% CI 0.47–0.71)
CRC, colorectal cancer; LTPA, leisure-time physical activity; MET-h/week, metabolic equivalent task hours per week
Table 4
Summary of observational studies on physical activity and prognosis in prostate cancer patients
Study
Population
LTPA
Outcome
Results
Richman et al. [115]
N = 1455
Non-metastatic PC
Walk briskly
≥ 3 h/week
Rate of progression
HR 0.43 (95% CI 0.21–0.91)
Friedenreich et al. [116]
N = 830
Stage II–IV PC
1997–2000
Post-diagnosis total activity > 119 MET-hours/week
All-cause mortality
HR 0.58 (95% CI 0.42–0.79)
Pre- and post-diagnosis activity
PC mortality
HR 0.56 (95% CI 0.35–0.90)
> 18 MET-hours/week
All-cause mortality
HR 0.66 (95% CI 0.49–0.88)
Kenfield et al. [117]
Health Professional Follow-up Study
N = 2705
Non-metastatic PC
1990–2008
Post-diagnosis walking ≥ 90 min per week
All-cause mortality
HR 0.54 (95% CI 0.41–0.71)
Post-diagnosis walking ≥ 3 h per week or vigorous activity
All-cause mortality
HR 0.51 (95% CI 0.36–0.72)
LTPA, leisure-time physical activity; MET-h/week, metabolic equivalent task hours per week; PC, prostate cancer
Table 5
Prospective observational studies on physical activity and prognosis in other cancers
Study
Population
LTPA
Outcome
Results
Liss et al. [118]
Texas and San Diego
222,163 Kidney cancer survivors 1998–2004
Any PA
Kidney cancer-specific mortality
HR 0.50 (95% CI 0.27–0.93)
Sloan et al. [119]
Rochester, US
1466 Lung cancer survivors 1997–2009
Physically active
Recurrence rate
Overall survival
81% versus 82% (P = 0.62)
8.4 year versus 4.4 year (P < 0.0001)
Wang et al. [120]
Chinese
303 Early esophageal cancer survivors
After surgery
> 9 MET-h/week
All-cause mortality
Risk of recurrence
HR 0.67 (95% CI 0.48–0.92)
HR 0.31 (95% CI 0.22–0.43)
HR, hazard ratio; LTPA, leisure-time physical activity; MET, metabolic equivalent; PA, physical activity
Epidemiologic and observational studies show a decrease in the risk of cancer recurrence and all-cause mortality in patients who practice regular PA [121123]. A systematic review of studies published through June 2013 concluded that PA performed before or after cancer diagnosis is associated with a reduced mortality risk among BC and colorectal cancer survivors [124]. Mortality in adult survivors of childhood cancer was inferior in those patients who practiced vigorous exercise after diagnosis in a large multicentric observational study [125]. In 2015, Lahart et al. [97] published a meta-analysis of 22 studies analyzing the impact of PA on BC outcomes. A literature search was performed using PubMed, EMBASE and CENTRAL databases from 1995 to October 2014. In 40% of the observational studies, the risk of relapse and death in BC survivors decreased in most physically active women. Most studies included an analysis of leisure-time PA and only a few of interventional PA programs. The majority of the studies did not perform a multivariable analysis to exclude the effect of known confounding factors, and less than half included clinical prognostic factors, such as stage, nodal status, age or type of treatment. These studies found a positive impact on all-cause and BC mortality in patients who practiced moderate or intense lifetime PA before the diagnosis of BC and in recent years before diagnosis. However, the authors recommend interpreting these results with caution due to the large heterogeneity of the studies. A post-diagnosis activity of at least 10 MET-hours/week was associated with a decrease in all-cause, and BC mortality and was not influenced by the heterogeneity; however, not all the studies could corroborate a decrease in recurrence risk. The “After Breast Cancer Pooling Project” included more than 13,000 women from four prospective cohorts of BC survivors in the USA and Shanghai and analyzed the association between PA at 18–48 months after diagnosis and risk of all-cause and BC-specific mortality and BC recurrence [126]. BC mortality was reduced in patients who achieved 18.7 or more MET-hours/week, and no association was found between PA and BC recurrence [126]. A comprehensive review of sixty-three interventional studies on women after BC adjuvant therapy concluded that PA interventions might have certain beneficial effects on QoL, cardiorespiratory fitness and psychological and social functions, but conclusions about BC recurrence, BC mortality and all-cause mortality could not be made [98].
Several prospective observational studies and meta-analyses in patients with colorectal cancer have suggested the benefit of PA before and after diagnosis in terms of improvements on all-cause and cancer-specific mortality after controlling for other confounding factors, such as BMI, sex, number of positive lymph nodes, age, baseline performance status (PS), adjuvant chemotherapy regimen or recurrence-free survival period [110, 111, 127, 128]. Similarly, three observational prospective studies in prostate cancer found a strong inverse relationship between exercise and the risk of cancer progression regardless of other known prognostic factors [115117]. A Chinese study in patients who underwent esophagectomy for esophageal cancer supported the benefit of PA (> 9 MET-hours/week) on recurrence risk and all-cause mortality [120]. Data from a prospective observational study in kidney cancer survivors investigating PA and diet changes suggested a decrease in the recurrence rate in patients who did any PA compared with those that were totally inactive [118]. The only study in lung cancer survivors showed better overall survival in patients who met > 9 MET-hours/week, but no difference in the recurrence rate was observed [119].
In conclusion, the real impact of PA on the risk of relapse and cancer mortality is not well-defined. PA may contribute to reduced cancer-related mortality and all-cause mortality in cancer survivors by modifying fat accumulation and improving cardiovascular and skeletal muscle function [129]. Numerous prospective observational studies consistently showed the benefit of PA on cancer outcomes; however, most of these studies were based on measures from self-reported questionnaires, including heterogeneous populations, and only a few performed a multivariable analysis to exclude the contribution of other confounding factors. Interventional studies with reliable and objective measures of PA in homogeneous populations are needed to confirm the data from observational studies and to evaluate the real effect of exercise on cancer prognosis.

Exercise-oncology: a pragmatic point of view for clinicians

Exercise-oncology is a new field of cancer care with the goal of the appropriate and rationale introduction of exercise programs into the overall management of cancer patients to take advantage of the numerous benefits associated with PA. Several major comprehensive cancer centers have created exercise-oncology units to implement these programs in a timely and organized manner. A collaborative work among rehab specialists, physiotherapists and exercise physiologists, as well as oncologists and radio-oncology specialists, is developed in these units.
Exercise has demonstrated numerous benefits on the QoL of patients with cancer throughout the history of the disease, ameliorating the negative impact of cancer on physical and psychological health and having a positive impact on patient survival [130133].
Despite these benefits, many questions about PA/exercise in cancer patients remain, as it is particularly challenging to elucidate how much exercise is needed to achieve patient improvements and how the exercise should be recommended and monitored by clinicians.

General PA/exercise recommendations for cancer patients

In 2010, the first exercise guidelines were published by a roundtable of the American College of Sports Medicine (ACSM) based on general WHO PA guidelines to the general population. These guidelines consist of a minimum exercise recommendation: 150 min of moderate-intensity exercise in 3–5 days combining 2 days of resistance exercise and 3 days of aerobic exercise or 70 min of high-intensity exercise combining 1 day of resistance exercise and 2 days of aerobic exercise [133].

Exercise in the cancer treatment continuum

First, it is important to highlight that exercise is feasible, effective and safe in patients with cancer throughout the course of the disease. However, there are specific recommendations for the different moments of the disease and its therapies.

Presurgical exercise

It has been shown that presurgical high-intensity interval training in cancer patients is feasible and effective in improving cardiorespiratory fitness, which is typically measured as peak oxygen uptake (VO2peak); this training makes sense when patients need to achieve a specific VO2peak to undergo surgery, as noted for patients with lung cancer [134]. The intervention was based on high-intensity aerobic exercise (cycling) from 50 to 100% of VO2peak for 30 min, 5 days per week.
In another study in patients with BC, a presurgical intervention consisting of 180 min of moderate aerobic exercise and 40 min of strength training per week was associated with physiological changes and alterations in gene expression in tumor tissue (notably, downregulation of pathways related to cell cycle, RNA transport and DNA replication) [135].

Exercise during chemotherapy

Several studies using PA concomitantly with neoadjuvant and adjuvant chemotherapy have been performed with different approaches, demonstrating safety, effectiveness and fitness improvements [136].
Exercise programs concomitant with neoadjuvant chemotherapy are usually focused on improving the VO2peak level or maintaining it at the baseline range after cancer treatment. Interventions are based on at least 3 days per week with different durations (from 4 to 12 weeks) in 30- to 60-min session with variable intensities, which range from 55 to 60% of VO2peak at the start to 70–100% of VO2peak at the end [137].
Exercise interventions concomitant with adjuvant therapy must take into account a safe starting time to be sure that surgical wounds are completely scarred. Different reviews have shown that exercise improves fitness capacity [138] and might reduce some cancer-related side effects, such as fatigue [136]. However, interventions in these studies were heterogeneous and did not often describe the intensity or type of exercise used. A recent meta-analysis suggested that a workload of 600 MET (intensity-minutes) was associated with a clinically significant improvement in fitness capacity, suggesting that a 10-week program of 90 min/week of supervised training at 70% of VO2peak may be sufficient [139141]. Another meta-analysis found that cancer survivors who completed 15 MET-h/week presented a 27% lower risk of cancer mortality with respect to controls, and this effect was greater in patients who were sedentary at pre-diagnosis (35% lower risk) [142].
Despite the aggressiveness of cancer therapies, medium to high-intensity exercise and different types of exercise interventions are well-tolerated by most patients. Both previous reviews mentioned above focused on exercise intervention during neoadjuvant and adjuvant treatments, including high-intensity intervention [137, 143].
In addition, resistance training has been shown to be safe and effective in preventing lean body mass loss and reducing body fat mass during neoadjuvant and adjuvant treatments [144].

Exercise in cancer survivors

It is well-known that cancer survivors obtain an improvement in QoL, body composition and physical fitness with exercise [130, 131, 144146]. Again, the challenge in this population is to determine how much exercise is needed to achieve the maximum benefits. Related to exercise intensity, Gil-Rey et al. [147] showed that cancer survivors have an important reduction in their fitness capacity after cancer therapy and therefore suggest a reduction in exercise intensity at the beginning of training (i.e., 41–64% of VO2max). However, high-intensity training is feasible, safe and effective for cancer patients, and a shorter time of training is likely sufficient to obtain benefits, which should be taken into account in the implementation of exercise strategies [148].
More research is needed to determine the dose–response relationship between exercise and physical improvements given that some data from past clinical studies have suggested that an exercise intensity higher than that included in the general WHO recommendations might be needed to improve patients’ health status [147].

Exercise in patients with advanced and metastatic disease

Previous studies and reviews have shown that exercise is a safe and effective tool to improve fitness and functional capacity, strength, QoL and fatigue. Fitness and functional capacity were assessed by the VO2peak and 6-min walking tests, showing significantly better results compared with the control group. In these studies, the aerobic exercise intensity ranged from 55 to 75% of VO2peak [15, 16, 19, 29]. Muscle strength was assessed with the one-repetition maximum (1RM) or estimated 1RM test (lower and upper limbs), and exercise intensity in these studies ranged from 40 to 80% of 1RM. Program durations ranged from 5 to 12 weeks [22, 25]. With respect to body composition, significant changes were observed in lean mass, but no changes in fat mass, body mass or BMI were observed in previous studies. The low intensity of the exercise intervention (from 55 to 70% of VO2peak) might be a reason for these inconsistent results [15, 16, 33].

Exercise supervision

Although the benefits of exercise are well-established, the exercise dose–response and the best type of exercise in terms of duration and intensity remain unclear, making it difficult to establish how to provide specific recommendations to each individual patient and how to supervise the patient’s exercise by clinicians. With these caveats in mind, it might be wise to differentiate between patients who clearly need specialist counseling (as those under active treatment or metastatic patients, and all patients with side effects who limit them physically) and patients who do not (survival with limited side effects) (Fig. 1). An exercise-oncology specialist is an exercise professional with a previous background that includes a general qualification in exercise and health with specific knowledge in oncology items. Related to the oncology items, general knowledge about cancer biology, biomarkers and treatments and their side effects should be used to adapt to and individualize exercise to patients’ needs.

Challenges for patients: general supervision

For those patients who do not present the need for specialist counseling, the control of patients by informed clinicians could be sufficient to achieve reasonable results. In this respect, there are some specific guidelines that could be followed by patients and supervised by a nonspecialist with the help of different tests, devices or scales. For example, following WHO/ACSM guidelines or achieving more than 10,000 steps per day [150] are reasonable goals for cancer survivors (Table 6).
Table 6
General challenges for patients without specialist counseling needs based on existing guidelines
Recommendation
Challenge
Intensity
WHO/ACSM guidelines
150 min per week
 30 min/3 times “aerobic” exercise
 30 min/2 times strength exercises
Moderate
75 min per week
 25 min/2 times aerobic exercise
 25 min/1 time strength exercises
High intensity
Survival recommendations [99, 107, 147, 151]
9 MET corresponding to 180 min of walking
5 km/h
Review psychological benefits [152]
12 MET; 90–120 min
Moderate intensity
Minimum step recommendations [150]
< 5000 steps/day
“Sedentary lifestyle index”
5000–7499 steps/day
It is typical of daily activity excluding sports/exercise and might be considered “low active”
7500–9999
“Somewhat active”
≥ 10,000 steps/day
“Active”
Individuals who take > 12,500 steps/day
“Highly active”
MET, metabolic equivalent
New technologies are improving methods to supervise the quality and quantity of exercise [153]. While behavioral interventions using text messages (with or without educational material and internet support) have produced limited effects on exercise adherence, mobile applications have been shown to be an effective and useful tool for both patients and providers to establish a healthy lifestyle. To achieve significant changes, it has been observed that these apps should include self-monitoring merged with other motivational techniques (goal setting, feedback on performance, review of goals, prompts, planning or barrier identifications, among others), allowing better supervision and control for patients and trainers [153].

Challenges for clinicians: learning exercise techniques

At present, exercise provides empowerment among health care providers, presenting a new challenge for them. One of the most important issues to address is who might prescribe and control exercise. It is possible that multidisciplinary committees, including oncologists, rehab departments and exercise physiologists, should be created to provide patients with the best counseling and training physicians to help individuals not requiring special help (Fig. 2).
However, while this scenario seems far away, other achievable proposals are feasible. The education of clinicians taking care of cancer patients and survivors about exercise techniques and control is a crucial point. Education and training should ideally start during university studies, although few institutions worldwide provide exercise theory and training to their future professionals. This education would have an impact not only in cancer patients but also on many other common pathologies such as cardiovascular, metabolic, joint and other diseases [154]. In accordance, patients’ associations playing a crucial role at present in providing exercise training and assistance until exercise will be included in the usual care.

Future lines of research

Despite the increasing number of studies addressing the benefits of exercise for cancer patients and survivors in the last 5 years, further research remains essential to clarify many unanswered questions. The establishment of new, more concrete guidelines for exercise in cancer patients is necessary not only for the exercise-oncology specialist but also for oncologists and other clinicians who take care of cancer patients.
There is an urgent need to further clarify the biological mechanism that makes exercise an effective method of intervention to decrease cancer incidence and mortality and to improve overall health in cancer patients. Studies of the modification of biological biomarkers before and after exercise are crucial for understanding the underlying mechanisms through which exercise can exert its influence on cancer biology. Several preclinical studies (discussed above) and clinical studies of small sample sizes have provided preliminary evidence on the relevance of the immune system, cytokines and insulin-related pathways [155]; however, because the evidence is preliminary, larger and statistically powerful studies are required.
In addition, new studies aimed at identifying the optimal intensity and duration of PA are needed. The characteristics of cancer survivors differ from those of the healthy population to whom the recommendations of the different health organizations are directed.
The best method of introducing exercise into the lifestyle of patients is also a matter to be addressed. The most effective behavioral interventions to achieve long-term changes in a patient’s lifestyle must be defined, bearing in mind that cancer diagnosis and treatment are “learning moments” in which patients are willing to change their daily activities to improve their health. The feasibility of using new technologies, such as mobile health applications and wrist and watch bands, as well as interventions based on social networks should be investigated to favor adherence and motivation to these programs of adapted PA.
In addition, future research on intervention in metastatic cancer stages should be performed due to the lack of knowledge in this area and the potential interest in improving the tolerance and effectiveness of treatments and the QoL of patients, many of whom can live today for many years after relapse due to the effective and sustained disease palliation that can be achieved with modern systemic treatments.

Conclusions

Regular PA is associated with major benefits to human health, including a reduced risk of some cancers.
The mechanisms through which exercise exerts its antitumor activity are still poorly understood but might be related to a direct effect on tumor cells (inhibition of tumor cell proliferation, induction of apoptosis, upregulation of tumor suppressor genes, anti-inflammatory effects) or to an enhancement of immune function.
There is convincing evidence that regular PA reduces the risk of colorectal cancer, while the reduction in postmenopausal BC and endometrial cancer risk is judged as probable. The effect of PA on the risk of other tumors is less evident but still possible.
Several epidemiological studies have suggested an association of regular PA with reduced cancer-related and all-cause mortality in some tumor types, particularly BC and colorectal cancer. The minimum amount of PA needed to achieve such a benefit is still unknown, although the US recommendations suggest that a minimum 10 MET-hours/week (equivalent to ≥ 150 min of moderate-intensity PA) is needed.
Exercise-oncology is a field of cancer care in which the goal is the introduction of exercise programs into the overall management of cancer patients. The first exercise guidelines for cancer patients were published in 2010 by the ACSM. These guidelines, which are mainly based on general WHO guidelines to the general population, consider that regular PA in cancer patients is safe and exerts positive effects in patients at multiple levels, particularly QoL. Exercise programs in cancer patients are feasible along the course of the disease, including the presurgical period, during adjuvant antitumor medical treatment (including chemotherapy) and in cancer survivors; a summary of these recommendations is shown in Table 7. However, the experience with regular exercise in metastatic cancer patients is limited.
Table 7
Exercise duration and intensity recommendation to cancer patients [147, 156]
Cancer treatment moment
Type of exercise
Description
Intensity
Duration
(start with…)
Examples
Pre-surgery without other treatment
Endurance
High-intensity interval training
60–90% of VO2peak [157, 158]
20–35 min
Walking
Running
Spinning
Strength
Global strength exercises
Exercises with patient’s own body weight
0–40% of 1RM
2 sets of 10 repetitions
Global strength circuits
Stretching
All body
Passive or active stretches
30 s/exercise
Yoga
Stretching classes
Surgery/mammary reconstructions
Endurance
Light intensity, avoid pain
Depending on patient’ mobility [143]
20–40 min
Walking
biking
Avoid swimming
Strength
Rehabilitation recommendations
Rehabilitation recommendations
Rehabilitation recommendations
Upper-limb surgery: light arm mobility and postural exercises
Abdominal surgery: hypopressive abdominal exercises, and postural exercises—including isometric exercises
Stretching
Rehabilitation recommendations
Rehabilitation recommendations
Rehabilitation recommendations
Passive stretching without pain, focusing on affected area, when specialist allows it
Under chemo/radiotherapy
Endurance
Adapted intervention to patient needs
From 41 to 64% (moderate) to 80–90% of VO2peak (high intensity, if the patient was previously active)
3 days per week/20–35 min
Walking
Dance
Bike
Spinning
Running
Avoid swimming
Strength
Global strength to prevent sarcopenia or cachexia
From light movements to 40–60% of 1RM at first [159]. Depending on the patient’s comorbidities [143]
2 days per week, 2 sets of 10 repetitions
Yoga
Pilates
Elastic bands
Global strength circuits
Machines
Stretching
Stretch gently all body. Special care with radiation areas. Avoid pain
Passive stretches
30 s/exercise
Yoga
Stretching classes
Under hormone therapy
Endurance
Moderate to high intensity depending on the patient’s previous situation. Rest for 48 h after high-intensity training
From 41–64% (moderate) to 80–90% of VO2peak (high intensity) if the patient was previously active
3 days per week/30–40 min
Walking
Dance
Bike
Spinning
Running
Swimming
Strength
Moderate to high intensity depending on the patient’s situation. Rest for 48 h after high-intensity training
From light movements to 40–60% of 1RM [159] at first. Depending on the patient’s comorbidities [143]
2 days per week, 2 sets of 10 repetitions
Yoga
Pilates
Elastic bands
Global strength circuits
Machines
Stretch
All body
Passive or active stretches
30 s/exercise
Yoga
Stretching classes
Survivors
Endurance
Moderate to high intensity depending on previous patients’ situation
From 41 to 64% (moderate) to 80–100% of VO2peak (intense) if the patient was initially active
3 days per week/30–40 min
Walking
Dance
Bike
Spinning
Running
Swimming
Strength
Moderate to high intensity depending on the patient’s previous situation. Special care for patients with functional limitations. Avoid pain.
From light movements to 40–60% of 1RM [159] at first. Depending on the patient comorbidities [143]
2 days per week, 2 sets of 10 repetitions
Yoga
Pilates
Elastic bands
Global strength circuits
Machines
Stretch
All body
Passive or active stretches
30 s/exercise
Yoga
Stretching classes
RM, repetition maximum; VO2peak, peak oxygen uptake
The introduction of exercise programs into the global management of cancer patients remains a challenge due to conceptual and logistic issues. The most effective behavioral interventions to achieve long-term changes in a patient’s lifestyle must be defined. New technologies, such as mobile health applications and wrist and watch bands (the so-called “mHealth”), can be of great help to monitor the compliance to these programs. The optimal intensity and duration of PA should be defined with more precision in future studies. Regarding logistics, the intervention of both exercise-oncology specialists and trained clinicians is probably necessary at different time points to provide the best care. Several major comprehensive cancer centers have created exercise-oncology units to implement these programs in a timely and organized manner, and these models could serve as a reference for other institutions.

Acknowledgements

We would like to thank to Fernando Rico-Villademoros, MD (COCIENTE S.L., Madrid, Spain) for his editorial assistance; his participation was funded by the Spanish Society of Medical Oncology (SEOM).

Compliance with ethical standards

Conflict of interest

MP declares that she have no conflict of interest; SC declares that she have no conflict of interest; JA received institutional research grants from Merck Serono and Novartis; CE received speaker’s honoraria from AstraZeneca, Roche, Novartis and Pfizer; MAS-P received consulting/advisory fees from AstraZeneca, Pfizer and Amgen, and speakers’ honoraria from Amgen, Roche and Pfizer; AL declares that he have no conflict of interest; MM has received research grants from Roche and Novartis, consulting/advisory fees from AstraZeneca, Amgen, Taiho Oncology, Roche/Genentech, Novartis, PharmaMar, Eli Lilly, PUMA Taiho Oncology and Pfizer, and speakers’ honoraria from AstraZeneca, Amgen, Roche/Genentech, Novartis and Pfizer.

Ethical approval

The manuscript does not contain any studies with human participants or animals performed by any of the authors.
Informed consent/ethical approval is not required for this type of project.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

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

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat World Health Organization. Physical activity. Geneva: WHO; 2018. World Health Organization. Physical activity. Geneva: WHO; 2018.
3.
Zurück zum Zitat Figueira A, Cortinhas A, Soares J, Leitão J, Ferreira R, Duarte J. Efficacy of exercise on breast cancer outcomes: a systematic review and meta-analysis of preclinical data. Int J Sports Med. 2018;39:327–42.PubMed Figueira A, Cortinhas A, Soares J, Leitão J, Ferreira R, Duarte J. Efficacy of exercise on breast cancer outcomes: a systematic review and meta-analysis of preclinical data. Int J Sports Med. 2018;39:327–42.PubMed
4.
Zurück zum Zitat Betof AS, Lascola CD, Weitzel D, Landon C, Scarbrough PM, Devi GR, Palmer G, Jones LW, Dewhirst MW. Modulation of murine breast tumor vascularity, hypoxia, and chemotherapeutic response by exercise. J Natl Cancer Inst. 2015;107:djv040.PubMedPubMedCentral Betof AS, Lascola CD, Weitzel D, Landon C, Scarbrough PM, Devi GR, Palmer G, Jones LW, Dewhirst MW. Modulation of murine breast tumor vascularity, hypoxia, and chemotherapeutic response by exercise. J Natl Cancer Inst. 2015;107:djv040.PubMedPubMedCentral
5.
Zurück zum Zitat Jones LW, Eves N, Courneya K, Chiu B, Baracos V, Hanson J, Johnson L, Mackey J. Effects of exercise training on antitumor efficacy of doxorubicin in MDA-MB-231 breast cancer xenografts. Clin Cancer Res. 2005;11:6695–8.PubMed Jones LW, Eves N, Courneya K, Chiu B, Baracos V, Hanson J, Johnson L, Mackey J. Effects of exercise training on antitumor efficacy of doxorubicin in MDA-MB-231 breast cancer xenografts. Clin Cancer Res. 2005;11:6695–8.PubMed
6.
Zurück zum Zitat Jones LW, Fels DR, West M, Allen JD, Broadwater G, Barry WT, Wilke LG, Masko E, Douglas PS, Dash RC, Povsic TJ, Peppercorn J, Marcom PK, Blackwell KL, Kimmick G, Turkington TG, Dewhirst MW. Modulation of circulating angiogenic factors and tumor biology by aerobic training in breast cancer patients receiving neoadjuvant chemotherapy. Cancer Prev Res. 2013;6:925–37. Jones LW, Fels DR, West M, Allen JD, Broadwater G, Barry WT, Wilke LG, Masko E, Douglas PS, Dash RC, Povsic TJ, Peppercorn J, Marcom PK, Blackwell KL, Kimmick G, Turkington TG, Dewhirst MW. Modulation of circulating angiogenic factors and tumor biology by aerobic training in breast cancer patients receiving neoadjuvant chemotherapy. Cancer Prev Res. 2013;6:925–37.
7.
Zurück zum Zitat Sturgeon K, Schadler K, Muthukumaran G, Ding D, Bajulaiye A, Thomas NJ, Ferrari V, Ryeom S, Libonati JR. Concomitant low-dose doxorubicin treatment and exercise. Am J Physiol Regul Integr Comp Physiol. 2014;307:R685–92.PubMedPubMedCentral Sturgeon K, Schadler K, Muthukumaran G, Ding D, Bajulaiye A, Thomas NJ, Ferrari V, Ryeom S, Libonati JR. Concomitant low-dose doxorubicin treatment and exercise. Am J Physiol Regul Integr Comp Physiol. 2014;307:R685–92.PubMedPubMedCentral
8.
Zurück zum Zitat Ruiz-Casado A, Martín-Ruiz A, Pérez LM, Provencio M, Fiuza-Luces C, Lucia A. Exercise and the hallmarks of cancer. Trends Cancer. 2017;3:423–41.PubMed Ruiz-Casado A, Martín-Ruiz A, Pérez LM, Provencio M, Fiuza-Luces C, Lucia A. Exercise and the hallmarks of cancer. Trends Cancer. 2017;3:423–41.PubMed
9.
Zurück zum Zitat Colbert LH, Mai V, Tooze J, Perkins S, Berrigan D, Hursting S. Negative energy balance induced by voluntary wheel running inhibits polyp development in APCMin mice. Carcinogenesis. 2006;27:2103–7.PubMed Colbert LH, Mai V, Tooze J, Perkins S, Berrigan D, Hursting S. Negative energy balance induced by voluntary wheel running inhibits polyp development in APCMin mice. Carcinogenesis. 2006;27:2103–7.PubMed
10.
Zurück zum Zitat Leung P-S, Aronson WJ, Ngo TH, Golding LA, Barnard RJ. Exercise alters the IGF axis in vivo and increases p53 protein in prostate tumor cells in vitro. J Appl Physiol. 2004;96:450–4.PubMed Leung P-S, Aronson WJ, Ngo TH, Golding LA, Barnard RJ. Exercise alters the IGF axis in vivo and increases p53 protein in prostate tumor cells in vitro. J Appl Physiol. 2004;96:450–4.PubMed
11.
Zurück zum Zitat Ouyang P, Jiang Y, Doan HM, Xie L, Vasquez D, Welti R, Su X, Lu N, Herndon B, Yang SS, Jeannotte R, Wang W. Weight loss via exercise with controlled dietary intake may affect phospholipid profile for cancer prevention in murine skin tissues. Cancer Prev Res. 2010;3:466–77. Ouyang P, Jiang Y, Doan HM, Xie L, Vasquez D, Welti R, Su X, Lu N, Herndon B, Yang SS, Jeannotte R, Wang W. Weight loss via exercise with controlled dietary intake may affect phospholipid profile for cancer prevention in murine skin tissues. Cancer Prev Res. 2010;3:466–77.
12.
Zurück zum Zitat Xie L, Jiang Y, Ouyang P, Chen J, Doan H, Herndon B, Sylvester JE, Zhang K, Molteni A, Reichle M, Zhang R, Haub MD, Baybutt RC, Wang W. Effects of dietary calorie restriction or exercise on the PI3K and ras signaling pathways in the skin of mice. J Biol Chem. 2007;282:28025–35.PubMed Xie L, Jiang Y, Ouyang P, Chen J, Doan H, Herndon B, Sylvester JE, Zhang K, Molteni A, Reichle M, Zhang R, Haub MD, Baybutt RC, Wang W. Effects of dietary calorie restriction or exercise on the PI3K and ras signaling pathways in the skin of mice. J Biol Chem. 2007;282:28025–35.PubMed
13.
Zurück zum Zitat Yu M, King B, Ewert E, Su X, Mardiyati N, Zhao Z, Wang W. Exercise activates p53 and negatively regulates IGF-1 pathway in epidermis within a skin cancer model. PLoS ONE. 2016;11:e0160939.PubMedPubMedCentral Yu M, King B, Ewert E, Su X, Mardiyati N, Zhao Z, Wang W. Exercise activates p53 and negatively regulates IGF-1 pathway in epidermis within a skin cancer model. PLoS ONE. 2016;11:e0160939.PubMedPubMedCentral
14.
Zurück zum Zitat Zhu Z, Jiang W, Zacher JH, Neil ES, McGinley JN, Thompson HJ. Effects of energy restriction and wheel running on mammary carcinogenesis and host systemic factors in a rat model. Cancer Prev Res. 2012;5:414–22. Zhu Z, Jiang W, Zacher JH, Neil ES, McGinley JN, Thompson HJ. Effects of energy restriction and wheel running on mammary carcinogenesis and host systemic factors in a rat model. Cancer Prev Res. 2012;5:414–22.
15.
Zurück zum Zitat Kalaany NY, Sabatini DM. Tumours with PI3K activation are resistant to dietary restriction. Nature. 2009;458:725–31.PubMedPubMedCentral Kalaany NY, Sabatini DM. Tumours with PI3K activation are resistant to dietary restriction. Nature. 2009;458:725–31.PubMedPubMedCentral
16.
Zurück zum Zitat Jiang W, Zhu Z, Thompson HJ. Effects of physical activity and restricted energy intake on chemically induced mammary carcinogenesis. Cancer Prev Res. 2009;2:338–44. Jiang W, Zhu Z, Thompson HJ. Effects of physical activity and restricted energy intake on chemically induced mammary carcinogenesis. Cancer Prev Res. 2009;2:338–44.
17.
Zurück zum Zitat Zhu Z, Jiang W, Sells JL, Neil ES, McGinley JN, Thompson HJ. Effect of nonmotorized wheel running on mammary carcinogenesis: circulating biomarkers, cellular processes, and molecular mechanisms in rats. Cancer Epidemiol Biomark Prev. 2008;17:1920–9. Zhu Z, Jiang W, Sells JL, Neil ES, McGinley JN, Thompson HJ. Effect of nonmotorized wheel running on mammary carcinogenesis: circulating biomarkers, cellular processes, and molecular mechanisms in rats. Cancer Epidemiol Biomark Prev. 2008;17:1920–9.
18.
Zurück zum Zitat Baltgalvis KA, Berger FG, Peña MMO, Davis JM, Carson JA. Effect of exercise on biological pathways in ApcMin/+ mouse intestinal polyps. J Appl Physiol. 2008;104:1137–43.PubMed Baltgalvis KA, Berger FG, Peña MMO, Davis JM, Carson JA. Effect of exercise on biological pathways in ApcMin/+ mouse intestinal polyps. J Appl Physiol. 2008;104:1137–43.PubMed
19.
Zurück zum Zitat Ju J, Nolan B, Cheh M, Bose M, Lin Y, Wagner GC, Yang CS. Voluntary exercise inhibits intestinal tumorigenesis in Apc Min/+ mice and azoxymethane/dextran sulfate sodium-treated mice. BMC Cancer. 2008;8:316.PubMedPubMedCentral Ju J, Nolan B, Cheh M, Bose M, Lin Y, Wagner GC, Yang CS. Voluntary exercise inhibits intestinal tumorigenesis in Apc Min/+ mice and azoxymethane/dextran sulfate sodium-treated mice. BMC Cancer. 2008;8:316.PubMedPubMedCentral
20.
Zurück zum Zitat Horak M, Zlamal F, Iliev R, Kucera J, Cacek J, Svobodova L, Hlavonova Z, Kalina T, Slaby O, Bienertova-Vasku J. Exercise-induced circulating microRNA changes in athletes in various training scenarios. PLoS ONE. 2018;13:e0191060.PubMedPubMedCentral Horak M, Zlamal F, Iliev R, Kucera J, Cacek J, Svobodova L, Hlavonova Z, Kalina T, Slaby O, Bienertova-Vasku J. Exercise-induced circulating microRNA changes in athletes in various training scenarios. PLoS ONE. 2018;13:e0191060.PubMedPubMedCentral
21.
Zurück zum Zitat Khori V, Amani Shalamzari S, Isanejad A, Alizadeh Ali M, Alizadeh S, Khodayari S, Khodayari H, Shahbazi S, Zahedi A, Sohanaki H, Khaniki M, Mahdian R, Saffari M, Fayad R. Effects of exercise training together with tamoxifen in reducing mammary tumor burden in mice: possible underlying pathway of miR-21. Eur J Pharmacol. 2015;765:179–87.PubMed Khori V, Amani Shalamzari S, Isanejad A, Alizadeh Ali M, Alizadeh S, Khodayari S, Khodayari H, Shahbazi S, Zahedi A, Sohanaki H, Khaniki M, Mahdian R, Saffari M, Fayad R. Effects of exercise training together with tamoxifen in reducing mammary tumor burden in mice: possible underlying pathway of miR-21. Eur J Pharmacol. 2015;765:179–87.PubMed
22.
Zurück zum Zitat Dethlefsen C, Hansen LS, Lillelund C, Andersen C, Gehl J, Christensen JF, Pedersen BK, Hojman P. Exercise-induced catecholamines activate the hippo tumor suppressor pathway to reduce risks of breast cancer development. Cancer Res. 2017;77:4894–904.PubMed Dethlefsen C, Hansen LS, Lillelund C, Andersen C, Gehl J, Christensen JF, Pedersen BK, Hojman P. Exercise-induced catecholamines activate the hippo tumor suppressor pathway to reduce risks of breast cancer development. Cancer Res. 2017;77:4894–904.PubMed
23.
Zurück zum Zitat Lu YP, Lou YR, Nolan B, Peng QY, Xie JG, Wagner GC, Conney AH. Stimulatory effect of voluntary exercise or fat removal (partial lipectomy) on apoptosis in the skin of UVB light-irradiated mice. Proc Natl Acad Sci USA. 2006;103:16301–6.PubMedPubMedCentral Lu YP, Lou YR, Nolan B, Peng QY, Xie JG, Wagner GC, Conney AH. Stimulatory effect of voluntary exercise or fat removal (partial lipectomy) on apoptosis in the skin of UVB light-irradiated mice. Proc Natl Acad Sci USA. 2006;103:16301–6.PubMedPubMedCentral
24.
Zurück zum Zitat Higgins KA, Park D, Lee GY, Curran WJ, Deng X. Exercise-induced lung cancer regression: mechanistic findings from a mouse model. Cancer. 2014;120:3302–10.PubMed Higgins KA, Park D, Lee GY, Curran WJ, Deng X. Exercise-induced lung cancer regression: mechanistic findings from a mouse model. Cancer. 2014;120:3302–10.PubMed
25.
Zurück zum Zitat Zheng X, Cui XX, Huang MT, Liu Y, Shih WJ, Lin Y, Lu YP, Wagner GC, Conney AH. Inhibitory effect of voluntary running wheel exercise on the growth of human pancreatic Panc-1 and prostate PC-3 xenograft tumors in immunodeficient mice. Oncol Rep. 2008;19:1583–8.PubMed Zheng X, Cui XX, Huang MT, Liu Y, Shih WJ, Lin Y, Lu YP, Wagner GC, Conney AH. Inhibitory effect of voluntary running wheel exercise on the growth of human pancreatic Panc-1 and prostate PC-3 xenograft tumors in immunodeficient mice. Oncol Rep. 2008;19:1583–8.PubMed
26.
Zurück zum Zitat Zheng X, Cui X-X, Huang M-T, Liu Y, Wagner GC, Lin Y, Shih WJ, Lee M-J, Yang CS, Conney AH. Inhibition of progression of androgen-dependent prostate LNCaP tumors to androgen independence in SCID mice by oral caffeine and voluntary exercise. Nutr Cancer. 2012;64:1029–37.PubMed Zheng X, Cui X-X, Huang M-T, Liu Y, Wagner GC, Lin Y, Shih WJ, Lee M-J, Yang CS, Conney AH. Inhibition of progression of androgen-dependent prostate LNCaP tumors to androgen independence in SCID mice by oral caffeine and voluntary exercise. Nutr Cancer. 2012;64:1029–37.PubMed
27.
Zurück zum Zitat Zhu Z, Jiang W, McGinley JN, Thompson HJ. Energetics and mammary carcinogenesis: effects of moderate-intensity running and energy intake on cellular processes and molecular mechanisms in rats. J Appl Physiol. 2009;106:911–8.PubMed Zhu Z, Jiang W, McGinley JN, Thompson HJ. Energetics and mammary carcinogenesis: effects of moderate-intensity running and energy intake on cellular processes and molecular mechanisms in rats. J Appl Physiol. 2009;106:911–8.PubMed
28.
Zurück zum Zitat Soliman S, Aronson WJ, Barnard RJ. Analyzing serum-stimulated prostate cancer cell lines after low-fat, high-fiber diet and exercise intervention. Evid Based Complement Altern Med. 2011;2011:529053. Soliman S, Aronson WJ, Barnard RJ. Analyzing serum-stimulated prostate cancer cell lines after low-fat, high-fiber diet and exercise intervention. Evid Based Complement Altern Med. 2011;2011:529053.
29.
Zurück zum Zitat Barnard RJ, Hong Gonzalez J, Liva ME, Ngo TH. Effects of a low-fat, high-fiber diet and exercise program on breast cancer risk factors in vivo and tumor cell growth and apoptosis in vitro. Nutr Cancer. 2006;55:28–34.PubMed Barnard RJ, Hong Gonzalez J, Liva ME, Ngo TH. Effects of a low-fat, high-fiber diet and exercise program on breast cancer risk factors in vivo and tumor cell growth and apoptosis in vitro. Nutr Cancer. 2006;55:28–34.PubMed
30.
Zurück zum Zitat Hojman P, Dethlefsen C, Brandt C, Hansen J, Pedersen L, Pedersen BK. Exercise-induced muscle-derived cytokines inhibit mammary cancer cell growth. Am J Physiol Endocrinol Metabol. 2011;301:E504–10. Hojman P, Dethlefsen C, Brandt C, Hansen J, Pedersen L, Pedersen BK. Exercise-induced muscle-derived cytokines inhibit mammary cancer cell growth. Am J Physiol Endocrinol Metabol. 2011;301:E504–10.
31.
Zurück zum Zitat He C, Bassik MC, Moresi V, Sun K, Wei Y, Zou Z, An Z, Loh J, Fisher J, Sun Q, Korsmeyer S, Packer M, May HI, Hill JA, Virgin HW, Gilpin C, Xiao G, Bassel-Duby R, Scherer PE, Levine B. Exercise-induced BCL2-regulated autophagy is required for muscle glucose homeostasis. Nature. 2012;481:511–5.PubMedPubMedCentral He C, Bassik MC, Moresi V, Sun K, Wei Y, Zou Z, An Z, Loh J, Fisher J, Sun Q, Korsmeyer S, Packer M, May HI, Hill JA, Virgin HW, Gilpin C, Xiao G, Bassel-Duby R, Scherer PE, Levine B. Exercise-induced BCL2-regulated autophagy is required for muscle glucose homeostasis. Nature. 2012;481:511–5.PubMedPubMedCentral
32.
Zurück zum Zitat Piguet A-C, Saran U, Simillion C, Keller I, Terracciano L, Reeves HL, Dufour J-F. Regular exercise decreases liver tumors development in hepatocyte-specific PTEN-deficient mice independently of steatosis. J Hepatol. 2015;62:1296–303.PubMed Piguet A-C, Saran U, Simillion C, Keller I, Terracciano L, Reeves HL, Dufour J-F. Regular exercise decreases liver tumors development in hepatocyte-specific PTEN-deficient mice independently of steatosis. J Hepatol. 2015;62:1296–303.PubMed
33.
Zurück zum Zitat Sanchis-Gomar F. Sestrins: novel antioxidant and AMPK-modulating functions regulated by exercise? J Cell Physiol. 2013;228:1647–50.PubMed Sanchis-Gomar F. Sestrins: novel antioxidant and AMPK-modulating functions regulated by exercise? J Cell Physiol. 2013;228:1647–50.PubMed
34.
Zurück zum Zitat Shannon AM, Bouchier-Hayes DJ, Condron CM, Toomey D. Tumour hypoxia, chemotherapeutic resistance and hypoxia-related therapies. Cancer Treat Rev. 2003;29:297–307.PubMed Shannon AM, Bouchier-Hayes DJ, Condron CM, Toomey D. Tumour hypoxia, chemotherapeutic resistance and hypoxia-related therapies. Cancer Treat Rev. 2003;29:297–307.PubMed
35.
Zurück zum Zitat Jones LW, Viglianti BL, Tashjian JA, Kothadia SM, Keir ST, Freedland SJ, Potter MQ, Jung Moon E, Schroeder T, Herndon JE, Dewhirst MW. Effect of aerobic exercise on tumor physiology in an animal model of human breast cancer. J Appl Physiol. 2010;108:343–8.PubMed Jones LW, Viglianti BL, Tashjian JA, Kothadia SM, Keir ST, Freedland SJ, Potter MQ, Jung Moon E, Schroeder T, Herndon JE, Dewhirst MW. Effect of aerobic exercise on tumor physiology in an animal model of human breast cancer. J Appl Physiol. 2010;108:343–8.PubMed
36.
Zurück zum Zitat Jones LW, Antonelli J, Masko EM, Broadwater G, Lascola CD, Fels D, Dewhirst MW, Dyck JRB, Nagendran J, Flores CT, Betof AS, Nelson ER, Pollak M, Dash RC, Young ME, Freedland SJ. Exercise modulation of the host-tumor interaction in an orthotopic model of murine prostate cancer. J Appl Physiol. 2012;113:263–72.PubMedPubMedCentral Jones LW, Antonelli J, Masko EM, Broadwater G, Lascola CD, Fels D, Dewhirst MW, Dyck JRB, Nagendran J, Flores CT, Betof AS, Nelson ER, Pollak M, Dash RC, Young ME, Freedland SJ. Exercise modulation of the host-tumor interaction in an orthotopic model of murine prostate cancer. J Appl Physiol. 2012;113:263–72.PubMedPubMedCentral
37.
Zurück zum Zitat McCullough DJ, Nguyen LMD, Siemann DW, Behnke BJ. Effects of exercise training on tumor hypoxia and vascular function in the rodent preclinical orthotopic prostate cancer model. J Appl Physiol. 2013;115:1846–54.PubMedPubMedCentral McCullough DJ, Nguyen LMD, Siemann DW, Behnke BJ. Effects of exercise training on tumor hypoxia and vascular function in the rodent preclinical orthotopic prostate cancer model. J Appl Physiol. 2013;115:1846–54.PubMedPubMedCentral
38.
Zurück zum Zitat McCullough DJ, Stabley JN, Siemann DW, Behnke BJ. Modulation of blood flow, hypoxia, and vascular function in orthotopic prostate tumors during exercise. J Natl Cancer Inst. 2014;106:dju036.PubMedPubMedCentral McCullough DJ, Stabley JN, Siemann DW, Behnke BJ. Modulation of blood flow, hypoxia, and vascular function in orthotopic prostate tumors during exercise. J Natl Cancer Inst. 2014;106:dju036.PubMedPubMedCentral
39.
Zurück zum Zitat Schadler KL, Thomas NJ, Galie PA, Bhang DH, Roby KC, Addai P, Till JE, Sturgeon K, Zaslavsky A, Chen CS, Ryeom S. Tumor vessel normalization after aerobic exercise enhances chemotherapeutic efficacy. Oncotarget. 2016;7:65429–40.PubMedPubMedCentral Schadler KL, Thomas NJ, Galie PA, Bhang DH, Roby KC, Addai P, Till JE, Sturgeon K, Zaslavsky A, Chen CS, Ryeom S. Tumor vessel normalization after aerobic exercise enhances chemotherapeutic efficacy. Oncotarget. 2016;7:65429–40.PubMedPubMedCentral
40.
Zurück zum Zitat Zhang QB, Zhang BH, Zhang KZ, Meng XT, Jia QA, Zhang QB, Bu Y, Zhu XD, Ma DN, Ye BG, Zhang N, Ren ZG, Sun HC, Tang ZY. Moderate swimming suppressed the growth and metastasis of the transplanted liver cancer in mice model: with reference to nervous system. Oncogene. 2016;35:4122–31.PubMed Zhang QB, Zhang BH, Zhang KZ, Meng XT, Jia QA, Zhang QB, Bu Y, Zhu XD, Ma DN, Ye BG, Zhang N, Ren ZG, Sun HC, Tang ZY. Moderate swimming suppressed the growth and metastasis of the transplanted liver cancer in mice model: with reference to nervous system. Oncogene. 2016;35:4122–31.PubMed
41.
Zurück zum Zitat Wolff G, Davidson SJ, Wrobel JK, Toborek M. Exercise maintains blood–brain barrier integrity during early stages of brain metastasis formation. Biochem Biophys Res Commun. 2015;463:811–7.PubMedPubMedCentral Wolff G, Davidson SJ, Wrobel JK, Toborek M. Exercise maintains blood–brain barrier integrity during early stages of brain metastasis formation. Biochem Biophys Res Commun. 2015;463:811–7.PubMedPubMedCentral
42.
Zurück zum Zitat Jia W, Lu R, Martin TA, Jiang WG. The role of claudin-5 in blood-brain barrier (BBB) and brain metastases (review). Mol Med Rep. 2014;9:779–85.PubMed Jia W, Lu R, Martin TA, Jiang WG. The role of claudin-5 in blood-brain barrier (BBB) and brain metastases (review). Mol Med Rep. 2014;9:779–85.PubMed
43.
Zurück zum Zitat Hanahan D, Weinberg Robert A. Hallmarks of cancer: the next generation. Cell. 2011;144:646–74.PubMed Hanahan D, Weinberg Robert A. Hallmarks of cancer: the next generation. Cell. 2011;144:646–74.PubMed
44.
Zurück zum Zitat Koelwyn GJ, Quail DF, Zhang X, White RM, Jones LW. Exercise-dependent regulation of the tumour microenvironment. Nat Rev Cancer. 2017;17:620–32.PubMed Koelwyn GJ, Quail DF, Zhang X, White RM, Jones LW. Exercise-dependent regulation of the tumour microenvironment. Nat Rev Cancer. 2017;17:620–32.PubMed
45.
Zurück zum Zitat Wiggins JM, Opoku-Acheampong AB, Baumfalk DR, Siemann DW, Behnke BJ. Exercise and the tumor microenvironment. Exerc Sport Sci Rev. 2018;46:56–64.PubMed Wiggins JM, Opoku-Acheampong AB, Baumfalk DR, Siemann DW, Behnke BJ. Exercise and the tumor microenvironment. Exerc Sport Sci Rev. 2018;46:56–64.PubMed
46.
Zurück zum Zitat Zielinski MR, Muenchow M, Wallig MA, Horn PL, Woods JA. Exercise delays allogeneic tumor growth and reduces intratumoral inflammation and vascularization. J Appl Physiol. 2004;96:2249–56.PubMed Zielinski MR, Muenchow M, Wallig MA, Horn PL, Woods JA. Exercise delays allogeneic tumor growth and reduces intratumoral inflammation and vascularization. J Appl Physiol. 2004;96:2249–56.PubMed
47.
Zurück zum Zitat Almeida PWM, Gomes-Filho A, Ferreira AJ, Rodrigues CEM, Dias-Peixoto MF, Russo RC, Teixeira MM, Cassali GD, Ferreira E, Santos IC, Garcia AMC, Silami-Garcia E, Wisløff U, Pussieldi GA. Swim training suppresses tumor growth in mice. J Appl Physiol. 2009;107:261–5.PubMed Almeida PWM, Gomes-Filho A, Ferreira AJ, Rodrigues CEM, Dias-Peixoto MF, Russo RC, Teixeira MM, Cassali GD, Ferreira E, Santos IC, Garcia AMC, Silami-Garcia E, Wisløff U, Pussieldi GA. Swim training suppresses tumor growth in mice. J Appl Physiol. 2009;107:261–5.PubMed
48.
Zurück zum Zitat Walsh NP, Gleeson M, Shephard RJ, Gleeson M, Woods JA, Bishop NC, Fleshner M, Green C, Pedersen BK, Hoffman-Goetz L, Rogers CJ, Northoff H, Abbasi A, Simon P. Position statement. Part one: immune function and exercise. Exerc Immunol Rev. 2011;17:6–63.PubMed Walsh NP, Gleeson M, Shephard RJ, Gleeson M, Woods JA, Bishop NC, Fleshner M, Green C, Pedersen BK, Hoffman-Goetz L, Rogers CJ, Northoff H, Abbasi A, Simon P. Position statement. Part one: immune function and exercise. Exerc Immunol Rev. 2011;17:6–63.PubMed
49.
Zurück zum Zitat Idorn M, Hojman P. Exercise-dependent regulation of NK cells in cancer protection. Trends Mol Med. 2016;22:565–77.PubMed Idorn M, Hojman P. Exercise-dependent regulation of NK cells in cancer protection. Trends Mol Med. 2016;22:565–77.PubMed
50.
Zurück zum Zitat Kruijsen-Jaarsma M, Revesz D, Bierings MB, Buffart LM, Takken T. Effects of exercise on immune function in patients with cancer: a systematic review. Exerc Immunol Rev. 2013;19:120–43.PubMed Kruijsen-Jaarsma M, Revesz D, Bierings MB, Buffart LM, Takken T. Effects of exercise on immune function in patients with cancer: a systematic review. Exerc Immunol Rev. 2013;19:120–43.PubMed
51.
Zurück zum Zitat Pedersen L, Idorn M, Olofsson Gitte H, Lauenborg B, Nookaew I, Hansen Rasmus H, Johannesen Helle H, Becker Jürgen C, Pedersen Katrine S, Dethlefsen C, Nielsen J, Gehl J, Pedersen Bente K, Thor Straten P, Hojman P. Voluntary running suppresses tumor growth through epinephrine- and IL-6-dependent NK cell mobilization and redistribution. Cell Metabol. 2016;23:554–62. Pedersen L, Idorn M, Olofsson Gitte H, Lauenborg B, Nookaew I, Hansen Rasmus H, Johannesen Helle H, Becker Jürgen C, Pedersen Katrine S, Dethlefsen C, Nielsen J, Gehl J, Pedersen Bente K, Thor Straten P, Hojman P. Voluntary running suppresses tumor growth through epinephrine- and IL-6-dependent NK cell mobilization and redistribution. Cell Metabol. 2016;23:554–62.
52.
Zurück zum Zitat Lu Q, Ceddia MA, Price EA, Ye SM, Woods JA. Chronic exercise increases macrophage-mediated tumor cytolysis in young and old mice. Am J Physiol Regul Integr Comp Physiol. 1999;276:R482–9. Lu Q, Ceddia MA, Price EA, Ye SM, Woods JA. Chronic exercise increases macrophage-mediated tumor cytolysis in young and old mice. Am J Physiol Regul Integr Comp Physiol. 1999;276:R482–9.
53.
Zurück zum Zitat Abdalla DR, Murta EFC, Michelin MA. The influence of physical activity on the profile of immune response cells and cytokine synthesis in mice with experimental breast tumors induced by 7,12-dimethylbenzanthracene. Eur J Cancer Prev. 2013;22:251–8.PubMed Abdalla DR, Murta EFC, Michelin MA. The influence of physical activity on the profile of immune response cells and cytokine synthesis in mice with experimental breast tumors induced by 7,12-dimethylbenzanthracene. Eur J Cancer Prev. 2013;22:251–8.PubMed
54.
Zurück zum Zitat Abdalla DR, Aleixo AAR, Murta EFC, Michelin MA. Innate immune response adaptation in mice subjected to administration of DMBA and physical activity. Oncol Lett. 2014;7:886–90.PubMed Abdalla DR, Aleixo AAR, Murta EFC, Michelin MA. Innate immune response adaptation in mice subjected to administration of DMBA and physical activity. Oncol Lett. 2014;7:886–90.PubMed
55.
Zurück zum Zitat Kizaki T, Takemasa T, Sakurai T, Izawa T, Hanawa T, Kamiya S, Haga S, Imaizumi K, Ohno H. Adaptation of macrophages to exercise training improves innate immunity. Biochem Biophys Res Commun. 2008;372:152–6.PubMed Kizaki T, Takemasa T, Sakurai T, Izawa T, Hanawa T, Kamiya S, Haga S, Imaizumi K, Ohno H. Adaptation of macrophages to exercise training improves innate immunity. Biochem Biophys Res Commun. 2008;372:152–6.PubMed
56.
Zurück zum Zitat Koelwyn GJ, Wennerberg E, Demaria S, Jones LW. Exercise in regulation of inflammation-immune axis function in cancer initiation and progression. Oncology (Williston Park). 2015;29(908–20):22. Koelwyn GJ, Wennerberg E, Demaria S, Jones LW. Exercise in regulation of inflammation-immune axis function in cancer initiation and progression. Oncology (Williston Park). 2015;29(908–20):22.
57.
Zurück zum Zitat Murphy EA, Davis JM, Brown AS, Carmichael MD, Mayer EP, Ghaffar A. Effects of moderate exercise and oat β-glucan on lung tumor metastases and macrophage antitumor cytotoxicity. J Appl Physiol. 2004;97:955–9.PubMed Murphy EA, Davis JM, Brown AS, Carmichael MD, Mayer EP, Ghaffar A. Effects of moderate exercise and oat β-glucan on lung tumor metastases and macrophage antitumor cytotoxicity. J Appl Physiol. 2004;97:955–9.PubMed
58.
Zurück zum Zitat Goh J, Tsai J, Bammler TK, Farin FM, Endicott E, Ladiges WC. Exercise training in transgenic mice is associated with attenuation of early breast cancer growth in a dose-dependent manner. PLoS ONE. 2013;8:e80123.PubMedPubMedCentral Goh J, Tsai J, Bammler TK, Farin FM, Endicott E, Ladiges WC. Exercise training in transgenic mice is associated with attenuation of early breast cancer growth in a dose-dependent manner. PLoS ONE. 2013;8:e80123.PubMedPubMedCentral
59.
Zurück zum Zitat McClellan JL, Steiner JL, Day SD, Enos RT, Davis MJ, Singh UP, Murphy EA. Exercise effects on polyp burden and immune markers in the ApcMin/+ mouse model of intestinal tumorigenesis. Int J Oncol. 2014;45:861–8.PubMedPubMedCentral McClellan JL, Steiner JL, Day SD, Enos RT, Davis MJ, Singh UP, Murphy EA. Exercise effects on polyp burden and immune markers in the ApcMin/+ mouse model of intestinal tumorigenesis. Int J Oncol. 2014;45:861–8.PubMedPubMedCentral
60.
Zurück zum Zitat Wang J, Song H, Tang X, Yang Y, Vieira VJ, Niu Y, Ma Y. Effect of exercise training intensity on murine T-regulatory cells and vaccination response. Scand J Med Sci Sports. 2012;22:643–52.PubMed Wang J, Song H, Tang X, Yang Y, Vieira VJ, Niu Y, Ma Y. Effect of exercise training intensity on murine T-regulatory cells and vaccination response. Scand J Med Sci Sports. 2012;22:643–52.PubMed
61.
Zurück zum Zitat Hampras SS, Nesline M, Wallace PK, Odunsi K, Furlani N, Davis W, Moysich KB. Predictors of immunosuppressive regulatory T lymphocytes in healthy women. J Cancer Epidemiol. 2012;2012:191090.PubMedPubMedCentral Hampras SS, Nesline M, Wallace PK, Odunsi K, Furlani N, Davis W, Moysich KB. Predictors of immunosuppressive regulatory T lymphocytes in healthy women. J Cancer Epidemiol. 2012;2012:191090.PubMedPubMedCentral
62.
Zurück zum Zitat Davis JM, Kohut ML, Jackson DA, Colbert LH, Mayer EP, Ghaffar A. Exercise effects on lung tumor metastases and in vitro alveolar macrophage antitumor cytotoxicity. Am J Physiol Regul Integr Comp Physiol. 1998;274:R1454–9. Davis JM, Kohut ML, Jackson DA, Colbert LH, Mayer EP, Ghaffar A. Exercise effects on lung tumor metastases and in vitro alveolar macrophage antitumor cytotoxicity. Am J Physiol Regul Integr Comp Physiol. 1998;274:R1454–9.
63.
Zurück zum Zitat Frellstedt L, Waldschmidt I, Gosset P, Desmet C, Pirottin D, Bureau F, Farnir F, Franck T, Dupuis-Tricaud M-C, Lekeux P, Art T. Training modifies innate immune responses in blood monocytes and in pulmonary alveolar macrophages. Am J Respir Cell Mol Biol. 2014;51:135–42.PubMed Frellstedt L, Waldschmidt I, Gosset P, Desmet C, Pirottin D, Bureau F, Farnir F, Franck T, Dupuis-Tricaud M-C, Lekeux P, Art T. Training modifies innate immune responses in blood monocytes and in pulmonary alveolar macrophages. Am J Respir Cell Mol Biol. 2014;51:135–42.PubMed
64.
Zurück zum Zitat Fiuza-Luces C, Garatachea N, Berger NA, Lucia A. Exercise is the real polypill. Physiology. 2013;28:330–58.PubMed Fiuza-Luces C, Garatachea N, Berger NA, Lucia A. Exercise is the real polypill. Physiology. 2013;28:330–58.PubMed
65.
Zurück zum Zitat Aoi W, Naito Y, Takagi T, Tanimura Y, Takanami Y, Kawai Y, Sakuma K, Hang LP, Mizushima K, Hirai Y, Koyama R, Wada S, Higashi A, Kokura S, Ichikawa H, Yoshikawa T. A novel myokine, secreted protein acidic and rich in cysteine (SPARC), suppresses colon tumorigenesis via regular exercise. Gut. 2012;62:882–9.PubMed Aoi W, Naito Y, Takagi T, Tanimura Y, Takanami Y, Kawai Y, Sakuma K, Hang LP, Mizushima K, Hirai Y, Koyama R, Wada S, Higashi A, Kokura S, Ichikawa H, Yoshikawa T. A novel myokine, secreted protein acidic and rich in cysteine (SPARC), suppresses colon tumorigenesis via regular exercise. Gut. 2012;62:882–9.PubMed
66.
Zurück zum Zitat Benatti FB, Pedersen BK. Exercise as an anti-inflammatory therapy for rheumatic diseases—myokine regulation. Nat Rev Rheumatol. 2015;11:86–97.PubMed Benatti FB, Pedersen BK. Exercise as an anti-inflammatory therapy for rheumatic diseases—myokine regulation. Nat Rev Rheumatol. 2015;11:86–97.PubMed
67.
Zurück zum Zitat World Health Organization. Assessing national capacity for the prevention and control of noncommunicable diseases. Geneva: WHO; 2012. World Health Organization. Assessing national capacity for the prevention and control of noncommunicable diseases. Geneva: WHO; 2012.
68.
Zurück zum Zitat Leitzmann M, Powers H, Anderson AS, Scoccianti C, Berrino F, Boutron-Ruault M-C, Cecchini M, Espina C, Key TJ, Norat T, Wiseman M, Romieu I. European code against cancer 4th edition: physical activity and cancer. Cancer Epidemiol. 2015;39:S46–55.PubMed Leitzmann M, Powers H, Anderson AS, Scoccianti C, Berrino F, Boutron-Ruault M-C, Cecchini M, Espina C, Key TJ, Norat T, Wiseman M, Romieu I. European code against cancer 4th edition: physical activity and cancer. Cancer Epidemiol. 2015;39:S46–55.PubMed
69.
Zurück zum Zitat World Cancer Research Fund/American Institute for Cancer Research. Food, nutrition, physical activity, and the prevention of cancer: a global perspective. Washington, DC: AICR; 2007. World Cancer Research Fund/American Institute for Cancer Research. Food, nutrition, physical activity, and the prevention of cancer: a global perspective. Washington, DC: AICR; 2007.
71.
Zurück zum Zitat Moore SC, Lee IM, Weiderpass E, Campbell PT, Sampson JN, Kitahara CM, Keadle SK, Arem H, Berrington de Gonzalez A, Hartge P, Adami H-O, Blair CK, Borch KB, Boyd E, Check DP, Fournier A, Freedman ND, Gunter M, Johannson M, Khaw K-T, Linet MS, Orsini N, Park Y, Riboli E, Robien K, Schairer C, Sesso H, Spriggs M, Van Dusen R, Wolk A, Matthews CE, Patel AV. Association of leisure-time physical activity with risk of 26 types of cancer in 1.44 million adults. JAMA Intern Med. 2016;176:816–25.PubMedPubMedCentral Moore SC, Lee IM, Weiderpass E, Campbell PT, Sampson JN, Kitahara CM, Keadle SK, Arem H, Berrington de Gonzalez A, Hartge P, Adami H-O, Blair CK, Borch KB, Boyd E, Check DP, Fournier A, Freedman ND, Gunter M, Johannson M, Khaw K-T, Linet MS, Orsini N, Park Y, Riboli E, Robien K, Schairer C, Sesso H, Spriggs M, Van Dusen R, Wolk A, Matthews CE, Patel AV. Association of leisure-time physical activity with risk of 26 types of cancer in 1.44 million adults. JAMA Intern Med. 2016;176:816–25.PubMedPubMedCentral
72.
Zurück zum Zitat Keum N, Bao Y, Smith-Warner SA, Orav J, Wu K, Fuchs CS, Giovannucci EL. Association of physical activity by type and intensity with digestive system cancer risk. JAMA Oncol. 2016;2:1146–53.PubMedPubMedCentral Keum N, Bao Y, Smith-Warner SA, Orav J, Wu K, Fuchs CS, Giovannucci EL. Association of physical activity by type and intensity with digestive system cancer risk. JAMA Oncol. 2016;2:1146–53.PubMedPubMedCentral
73.
Zurück zum Zitat Kyu HH, Bachman VF, Alexander LT, Mumford JE, Afshin A, Estep K, Veerman JL, Delwiche K, Iannarone ML, Moyer ML, Cercy K, Vos T, Murray CJL, Forouzanfar MH. Physical activity and risk of breast cancer, colon cancer, diabetes, ischemic heart disease, and ischemic stroke events: systematic review and dose-response meta-analysis for the Global Burden of Disease Study 2013. BMJ. 2016;354:i3857.PubMedPubMedCentral Kyu HH, Bachman VF, Alexander LT, Mumford JE, Afshin A, Estep K, Veerman JL, Delwiche K, Iannarone ML, Moyer ML, Cercy K, Vos T, Murray CJL, Forouzanfar MH. Physical activity and risk of breast cancer, colon cancer, diabetes, ischemic heart disease, and ischemic stroke events: systematic review and dose-response meta-analysis for the Global Burden of Disease Study 2013. BMJ. 2016;354:i3857.PubMedPubMedCentral
75.
Zurück zum Zitat Catsburg C, Kirsh VA, Soskolne CL, Kreiger N, Bruce E, Ho T, Leatherdale ST, Rohan TE. Associations between anthropometric characteristics, physical activity, and breast cancer risk in a Canadian cohort. Breast Cancer Res Treat. 2014;145:545–52.PubMed Catsburg C, Kirsh VA, Soskolne CL, Kreiger N, Bruce E, Ho T, Leatherdale ST, Rohan TE. Associations between anthropometric characteristics, physical activity, and breast cancer risk in a Canadian cohort. Breast Cancer Res Treat. 2014;145:545–52.PubMed
76.
Zurück zum Zitat Lope V, Martín M, Castelló A, Casla S, Ruiz A, Baena-Cañada JM, Casas AM, Calvo L, Bermejo B, Muñoz M, Ramos M, de Juan-Ferré A, Jara C, Antón A, Jimeno MÁ, Lluch A, Antolín S, García-Sáenz JÁ, Estévez P, Arriola-Arellano E, Gavilá J, Pérez-Gómez B, Carrasco E, Pollán M. Physical activity and breast cancer risk by pathological subtype. Gynecol Oncol. 2017;144:577–85.PubMed Lope V, Martín M, Castelló A, Casla S, Ruiz A, Baena-Cañada JM, Casas AM, Calvo L, Bermejo B, Muñoz M, Ramos M, de Juan-Ferré A, Jara C, Antón A, Jimeno MÁ, Lluch A, Antolín S, García-Sáenz JÁ, Estévez P, Arriola-Arellano E, Gavilá J, Pérez-Gómez B, Carrasco E, Pollán M. Physical activity and breast cancer risk by pathological subtype. Gynecol Oncol. 2017;144:577–85.PubMed
78.
Zurück zum Zitat Schmid D, Behrens G, Keimling M, Jochem C, Ricci C, Leitzmann M. A systematic review and meta-analysis of physical activity and endometrial cancer risk. Eur J Epidemiol. 2015;30:397–412.PubMed Schmid D, Behrens G, Keimling M, Jochem C, Ricci C, Leitzmann M. A systematic review and meta-analysis of physical activity and endometrial cancer risk. Eur J Epidemiol. 2015;30:397–412.PubMed
79.
Zurück zum Zitat Brenner DR, Yannitsos DH, Farris MS, Johansson M, Friedenreich CM. Leisure-time physical activity and lung cancer risk: a systematic review and meta-analysis. Lung Cancer. 2016;95:17–27.PubMed Brenner DR, Yannitsos DH, Farris MS, Johansson M, Friedenreich CM. Leisure-time physical activity and lung cancer risk: a systematic review and meta-analysis. Lung Cancer. 2016;95:17–27.PubMed
82.
Zurück zum Zitat Behrens G, Jochem C, Keimling M, Ricci C, Schmid D, Leitzmann MF. The association between physical activity and gastroesophageal cancer: systematic review and meta-analysis. Eur J Epidemiol. 2014;29:151–70.PubMed Behrens G, Jochem C, Keimling M, Ricci C, Schmid D, Leitzmann MF. The association between physical activity and gastroesophageal cancer: systematic review and meta-analysis. Eur J Epidemiol. 2014;29:151–70.PubMed
85.
Zurück zum Zitat Shephard RJ. Physical activity and prostate cancer: an updated review. Sports Med. 2017;47:1055–73.PubMed Shephard RJ. Physical activity and prostate cancer: an updated review. Sports Med. 2017;47:1055–73.PubMed
87.
Zurück zum Zitat Cannioto RA, Moysich KB. Epithelial ovarian cancer and recreational physical activity: a review of the epidemiological literature and implications for exercise prescription. Gynecol Oncol. 2015;137:559–73.PubMedPubMedCentral Cannioto RA, Moysich KB. Epithelial ovarian cancer and recreational physical activity: a review of the epidemiological literature and implications for exercise prescription. Gynecol Oncol. 2015;137:559–73.PubMedPubMedCentral
88.
Zurück zum Zitat Huang T, Eliassen AH, Hankinson SE, Okereke OI, Kubzansky LD, Wang M, Poole EM, Chavarro JE, Tworoger SS. A prospective study of leisure-time physical activity and risk of incident epithelial ovarian cancer: impact by menopausal status. Int J Cancer. 2016;138:843–52.PubMed Huang T, Eliassen AH, Hankinson SE, Okereke OI, Kubzansky LD, Wang M, Poole EM, Chavarro JE, Tworoger SS. A prospective study of leisure-time physical activity and risk of incident epithelial ovarian cancer: impact by menopausal status. Int J Cancer. 2016;138:843–52.PubMed
89.
Zurück zum Zitat World Cancer Research Fund/American Institute for Cancer Research. Continous Update Project Expert Report 2018. Food, nutrition, physical activity, and the prevention of pancreatic cancer. Available at http://dietandcancerreport.org. Accesed 4 June 2019. World Cancer Research Fund/American Institute for Cancer Research. Continous Update Project Expert Report 2018. Food, nutrition, physical activity, and the prevention of pancreatic cancer. Available at http://​dietandcancerrep​ort.​org. Accesed 4 June 2019.
90.
Zurück zum Zitat Farris MS, Mosli MH, McFadden AA, Friedenreich CM, Brenner DR. The association between leisure time physical activity and pancreatic cancer risk in adults: a systematic review and meta-analysis. Cancer Epidemiol Biomark Prev. 2015;24:1462–73. Farris MS, Mosli MH, McFadden AA, Friedenreich CM, Brenner DR. The association between leisure time physical activity and pancreatic cancer risk in adults: a systematic review and meta-analysis. Cancer Epidemiol Biomark Prev. 2015;24:1462–73.
91.
Zurück zum Zitat Noor NM, Banim PJR, Luben RN, Khaw K-T, Hart AR. Investigating physical activity in the etiology of pancreatic cancer. Pancreas. 2016;45:388–93.PubMedPubMedCentral Noor NM, Banim PJR, Luben RN, Khaw K-T, Hart AR. Investigating physical activity in the etiology of pancreatic cancer. Pancreas. 2016;45:388–93.PubMedPubMedCentral
92.
93.
Zurück zum Zitat Behrens G, Leitzmann MF. The association between physical activity and renal cancer: systematic review and meta-analysis. Br J Cancer. 2013;108:798–811.PubMedPubMedCentral Behrens G, Leitzmann MF. The association between physical activity and renal cancer: systematic review and meta-analysis. Br J Cancer. 2013;108:798–811.PubMedPubMedCentral
95.
Zurück zum Zitat Keimling M, Behrens G, Schmid D, Jochem C, Leitzmann MF. The association between physical activity and bladder cancer: systematic review and meta-analysis. Br J Cancer. 2014;110:1862–70.PubMedPubMedCentral Keimling M, Behrens G, Schmid D, Jochem C, Leitzmann MF. The association between physical activity and bladder cancer: systematic review and meta-analysis. Br J Cancer. 2014;110:1862–70.PubMedPubMedCentral
96.
Zurück zum Zitat Jochem C, Leitzmann MF, Keimling M, Schmid D, Behrens G. Physical activity in relation to risk of hematologic cancers: a systematic review and meta-analysis. Cancer Epidemiol Biomark Prev. 2014;23:833–46. Jochem C, Leitzmann MF, Keimling M, Schmid D, Behrens G. Physical activity in relation to risk of hematologic cancers: a systematic review and meta-analysis. Cancer Epidemiol Biomark Prev. 2014;23:833–46.
97.
Zurück zum Zitat 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. 2015;54:635–54.PubMed 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. 2015;54:635–54.PubMed
98.
Zurück zum Zitat Lahart IM, Metsios GS, Nevill AM, Carmichael AR. Physical activity for women with breast cancer after adjuvant therapy. Cochrane Database Syst Rev. 2018;1:CD011292.PubMed Lahart IM, Metsios GS, Nevill AM, Carmichael AR. Physical activity for women with breast cancer after adjuvant therapy. Cochrane Database Syst Rev. 2018;1:CD011292.PubMed
99.
Zurück zum Zitat Holmes MD, Chen WY, Feskanich D, Kroenke CH, Colditz GA. Physical activity and survival after breast cancer diagnosis. JAMA. 2005;293:2479–86.PubMed Holmes MD, Chen WY, Feskanich D, Kroenke CH, Colditz GA. Physical activity and survival after breast cancer diagnosis. JAMA. 2005;293:2479–86.PubMed
100.
Zurück zum Zitat Irwin ML, Smith AW, McTiernan A, Ballard-Barbash R, Cronin K, Gilliland FD, Baumgartner RN, Baumgartner KB, Bernstein L. Influence of pre- and postdiagnosis physical activity on mortality in breast cancer survivors: the health, eating, activity, and lifestyle study. J Clin Oncol. 2008;26:3958–64.PubMedPubMedCentral Irwin ML, Smith AW, McTiernan A, Ballard-Barbash R, Cronin K, Gilliland FD, Baumgartner RN, Baumgartner KB, Bernstein L. Influence of pre- and postdiagnosis physical activity on mortality in breast cancer survivors: the health, eating, activity, and lifestyle study. J Clin Oncol. 2008;26:3958–64.PubMedPubMedCentral
101.
Zurück zum Zitat Bao P-P, Zhao G-M, Shu X-O, Peng P, Cai H, Lu W, Zheng Y. Modifiable lifestyle factors and triple-negative breast cancer survival. Epidemiology. 2015;26:909–16.PubMedPubMedCentral Bao P-P, Zhao G-M, Shu X-O, Peng P, Cai H, Lu W, Zheng Y. Modifiable lifestyle factors and triple-negative breast cancer survival. Epidemiology. 2015;26:909–16.PubMedPubMedCentral
102.
Zurück zum Zitat Schmidt ME, Chang-Claude J, Vrieling A, Seibold P, Heinz J, Obi N, Flesch-Janys D, Steindorf K. Association of pre-diagnosis physical activity with recurrence and mortality among women with breast cancer. Int J Cancer. 2013;133:1431–40.PubMed Schmidt ME, Chang-Claude J, Vrieling A, Seibold P, Heinz J, Obi N, Flesch-Janys D, Steindorf K. Association of pre-diagnosis physical activity with recurrence and mortality among women with breast cancer. Int J Cancer. 2013;133:1431–40.PubMed
103.
Zurück zum Zitat Holick CN, Newcomb PA, Trentham-Dietz A, Titus-Ernstoff L, Bersch AJ, Stampfer MJ, Baron JA, Egan KM, Willett WC. Physical activity and survival after diagnosis of invasive breast cancer. Cancer Epidemiol Biomark Prev. 2008;17:379–86. Holick CN, Newcomb PA, Trentham-Dietz A, Titus-Ernstoff L, Bersch AJ, Stampfer MJ, Baron JA, Egan KM, Willett WC. Physical activity and survival after diagnosis of invasive breast cancer. Cancer Epidemiol Biomark Prev. 2008;17:379–86.
104.
Zurück zum Zitat Ammitzbøll G, Søgaard K, Karlsen RV, Tjønneland A, Johansen C, Frederiksen K, Bidstrup P. Physical activity and survival in breast cancer. Eur J Cancer. 2016;66:67–74.PubMed Ammitzbøll G, Søgaard K, Karlsen RV, Tjønneland A, Johansen C, Frederiksen K, Bidstrup P. Physical activity and survival in breast cancer. Eur J Cancer. 2016;66:67–74.PubMed
105.
Zurück zum Zitat Friedenreich CM, Gregory J, Kopciuk KA, Mackey JR, Courneya KS. Prospective cohort study of lifetime physical activity and breast cancer survival. Int J Cancer. 2009;124:1954–62.PubMed Friedenreich CM, Gregory J, Kopciuk KA, Mackey JR, Courneya KS. Prospective cohort study of lifetime physical activity and breast cancer survival. Int J Cancer. 2009;124:1954–62.PubMed
106.
Zurück zum Zitat Sternfeld B, Weltzien E, Quesenberry CP, Castillo AL, Kwan M, Slattery ML, Caan BJ. Physical activity and risk of recurrence and mortality in breast cancer survivors: findings from the LACE study. Cancer Epidemiol Biomark Prev. 2009;18:87–95. Sternfeld B, Weltzien E, Quesenberry CP, Castillo AL, Kwan M, Slattery ML, Caan BJ. Physical activity and risk of recurrence and mortality in breast cancer survivors: findings from the LACE study. Cancer Epidemiol Biomark Prev. 2009;18:87–95.
107.
Zurück zum Zitat Irwin ML, McTiernan A, Manson JE, Thomson CA, Sternfeld B, Stefanick ML, Wactawski-Wende J, Craft L, Lane D, Martin LW, Chlebowski R. Physical activity and survival in postmenopausal women with breast cancer: results from the women’s health initiative. Cancer Prev Res. 2011;4:522–9. Irwin ML, McTiernan A, Manson JE, Thomson CA, Sternfeld B, Stefanick ML, Wactawski-Wende J, Craft L, Lane D, Martin LW, Chlebowski R. Physical activity and survival in postmenopausal women with breast cancer: results from the women’s health initiative. Cancer Prev Res. 2011;4:522–9.
108.
Zurück zum Zitat Bertram LAC, Stefanick ML, Saquib N, Natarajan L, Patterson RE, Bardwell W, Flatt SW, Newman VA, Rock CL, Thomson CA, Pierce JP. Physical activity, additional breast cancer events, and mortality among early-stage breast cancer survivors: findings from the WHEL Study. Cancer Causes Control. 2011;22:427–35.PubMed Bertram LAC, Stefanick ML, Saquib N, Natarajan L, Patterson RE, Bardwell W, Flatt SW, Newman VA, Rock CL, Thomson CA, Pierce JP. Physical activity, additional breast cancer events, and mortality among early-stage breast cancer survivors: findings from the WHEL Study. Cancer Causes Control. 2011;22:427–35.PubMed
109.
Zurück zum Zitat Bradshaw PT, Ibrahim JG, Khankari N, Cleveland RJ, Abrahamson PE, Stevens J, Satia JA, Teitelbaum SL, Neugut AI, Gammon MD. Post-diagnosis physical activity and survival after breast cancer diagnosis: the Long Island Breast Cancer Study. Breast Cancer Res Treat. 2014;145:735–42.PubMedPubMedCentral Bradshaw PT, Ibrahim JG, Khankari N, Cleveland RJ, Abrahamson PE, Stevens J, Satia JA, Teitelbaum SL, Neugut AI, Gammon MD. Post-diagnosis physical activity and survival after breast cancer diagnosis: the Long Island Breast Cancer Study. Breast Cancer Res Treat. 2014;145:735–42.PubMedPubMedCentral
110.
Zurück zum Zitat Walter V, Jansen L, Knebel P, Chang-Claude J, Hoffmeister M, Brenner H. Physical activity and survival of colorectal cancer patients: population-based study from Germany. Int J Cancer. 2017;140:1985–97.PubMed Walter V, Jansen L, Knebel P, Chang-Claude J, Hoffmeister M, Brenner H. Physical activity and survival of colorectal cancer patients: population-based study from Germany. Int J Cancer. 2017;140:1985–97.PubMed
111.
Zurück zum Zitat Arem H, Pfeiffer RM, Engels EA, Alfano CM, Hollenbeck A, Park Y, Matthews CE. Pre- and postdiagnosis physical activity, television viewing, and mortality among patients with colorectal cancer in the National Institutes of Health—AARP diet and health study. J Clin Oncol. 2015;33:180–8.PubMed Arem H, Pfeiffer RM, Engels EA, Alfano CM, Hollenbeck A, Park Y, Matthews CE. Pre- and postdiagnosis physical activity, television viewing, and mortality among patients with colorectal cancer in the National Institutes of Health—AARP diet and health study. J Clin Oncol. 2015;33:180–8.PubMed
112.
Zurück zum Zitat Meyerhardt JA, Giovannucci EL, Holmes MD, Chan AT, Chan JA, Colditz GA, Fuchs CS. Physical activity and survival after colorectal cancer diagnosis. J Clin Oncol. 2006;24:3527–34.PubMed Meyerhardt JA, Giovannucci EL, Holmes MD, Chan AT, Chan JA, Colditz GA, Fuchs CS. Physical activity and survival after colorectal cancer diagnosis. J Clin Oncol. 2006;24:3527–34.PubMed
113.
Zurück zum Zitat van Blarigan EL, Fuchs CS, Niedzwiecki D, Zhang S, Saltz LB, Mayer RJ, Mowat RB, Whittom R, Hantel A, Benson A, Atienza D, Messino M, Kindler H, Venook A, Ogino S, Giovannucci EL, Ng K, Meyerhardt JA. Association of survival with adherence to the american cancer society nutrition and physical activity guidelines for cancer survivors after colon cancer diagnosis. JAMA Oncol. 2018;4:783–90.PubMedPubMedCentral van Blarigan EL, Fuchs CS, Niedzwiecki D, Zhang S, Saltz LB, Mayer RJ, Mowat RB, Whittom R, Hantel A, Benson A, Atienza D, Messino M, Kindler H, Venook A, Ogino S, Giovannucci EL, Ng K, Meyerhardt JA. Association of survival with adherence to the american cancer society nutrition and physical activity guidelines for cancer survivors after colon cancer diagnosis. JAMA Oncol. 2018;4:783–90.PubMedPubMedCentral
114.
Zurück zum Zitat Campbell PT, Patel AV, Newton CC, Jacobs EJ, Gapstur SM. Associations of recreational physical activity and leisure time spent sitting with colorectal cancer survival. J Clin Oncol. 2013;31:876–85.PubMed Campbell PT, Patel AV, Newton CC, Jacobs EJ, Gapstur SM. Associations of recreational physical activity and leisure time spent sitting with colorectal cancer survival. J Clin Oncol. 2013;31:876–85.PubMed
115.
Zurück zum Zitat Richman EL, Kenfield SA, Stampfer MJ, Paciorek A, Carroll PR, Chan JM. Physical activity after diagnosis and risk of prostate cancer progression: data from the cancer of the prostate strategic urologic research endeavor. Cancer Res. 2011;71:3889–95.PubMedPubMedCentral Richman EL, Kenfield SA, Stampfer MJ, Paciorek A, Carroll PR, Chan JM. Physical activity after diagnosis and risk of prostate cancer progression: data from the cancer of the prostate strategic urologic research endeavor. Cancer Res. 2011;71:3889–95.PubMedPubMedCentral
116.
Zurück zum Zitat Friedenreich CM, Wang Q, Neilson HK, Kopciuk KA, McGregor SE, Courneya KS. Physical activity and survival after prostate cancer. Eur Urol. 2016;70:576–85.PubMed Friedenreich CM, Wang Q, Neilson HK, Kopciuk KA, McGregor SE, Courneya KS. Physical activity and survival after prostate cancer. Eur Urol. 2016;70:576–85.PubMed
117.
Zurück zum Zitat Kenfield SA, Stampfer MJ, Giovannucci E, Chan JM. Physical activity and survival after prostate cancer diagnosis in the health professionals follow-up study. J Clin Oncol. 2011;29:726–32.PubMedPubMedCentral Kenfield SA, Stampfer MJ, Giovannucci E, Chan JM. Physical activity and survival after prostate cancer diagnosis in the health professionals follow-up study. J Clin Oncol. 2011;29:726–32.PubMedPubMedCentral
118.
Zurück zum Zitat Liss M, Natarajan L, Hasan A, Noguchi JL, White M, Parsons JK. Physical activity decreases kidney cancer mortality. Curr Urol. 2017;10:193–8.PubMedPubMedCentral Liss M, Natarajan L, Hasan A, Noguchi JL, White M, Parsons JK. Physical activity decreases kidney cancer mortality. Curr Urol. 2017;10:193–8.PubMedPubMedCentral
119.
Zurück zum Zitat Sloan JA, Cheville AL, Liu H, Novotny PJ, Wampfler JA, Garces YI, Clark MM, Yang P. Impact of self-reported physical activity and health promotion behaviors on lung cancer survivorship. Health Qual Outcomes. 2016;14:66. Sloan JA, Cheville AL, Liu H, Novotny PJ, Wampfler JA, Garces YI, Clark MM, Yang P. Impact of self-reported physical activity and health promotion behaviors on lung cancer survivorship. Health Qual Outcomes. 2016;14:66.
120.
Zurück zum Zitat Wang L, Wang C, Wang J, Huang X, Cheng Y. Longitudinal, observational study on associations between postoperative nutritional vitamin D supplementation and clinical outcomes in esophageal cancer patients undergoing esophagectomy. Sci Rep. 2016;6:1572–81. Wang L, Wang C, Wang J, Huang X, Cheng Y. Longitudinal, observational study on associations between postoperative nutritional vitamin D supplementation and clinical outcomes in esophageal cancer patients undergoing esophagectomy. Sci Rep. 2016;6:1572–81.
121.
Zurück zum Zitat Ballard-Barbash R, Friedenreich CM, Courneya KS, Siddiqi SM, McTiernan A, Alfano CM. Physical activity, biomarkers, and disease outcomes in cancer survivors: a systematic review. J Natl Cancer Inst. 2012;104:815–40.PubMedPubMedCentral Ballard-Barbash R, Friedenreich CM, Courneya KS, Siddiqi SM, McTiernan A, Alfano CM. Physical activity, biomarkers, and disease outcomes in cancer survivors: a systematic review. J Natl Cancer Inst. 2012;104:815–40.PubMedPubMedCentral
122.
Zurück zum Zitat Gunnell AS, Joyce S, Tomlin S, Taaffe DR, Cormie P, Newton RU, Joseph D, Spry N, Einarsdóttir K, Galvão DA. Physical activity and survival among long-term cancer survivor and non-cancer cohorts. Front Public Health. 2017;5:19.PubMedPubMedCentral Gunnell AS, Joyce S, Tomlin S, Taaffe DR, Cormie P, Newton RU, Joseph D, Spry N, Einarsdóttir K, Galvão DA. Physical activity and survival among long-term cancer survivor and non-cancer cohorts. Front Public Health. 2017;5:19.PubMedPubMedCentral
123.
Zurück zum Zitat Li T, Wei S, Shi Y, Pang S, Qin Q, Yin J, Deng Y, Chen Q, Wei S, Nie S, Liu L. The dose–response effect of physical activity on cancer mortality: findings from 71 prospective cohort studies. Br J Sports Med. 2016;50:339–45.PubMed Li T, Wei S, Shi Y, Pang S, Qin Q, Yin J, Deng Y, Chen Q, Wei S, Nie S, Liu L. The dose–response effect of physical activity on cancer mortality: findings from 71 prospective cohort studies. Br J Sports Med. 2016;50:339–45.PubMed
124.
Zurück zum Zitat Schmid D, Leitzmann MF. Association between physical activity and mortality among breast cancer and colorectal cancer survivors: a systematic review and meta-analysis. Ann Oncol. 2014;25:1293–311.PubMed Schmid D, Leitzmann MF. Association between physical activity and mortality among breast cancer and colorectal cancer survivors: a systematic review and meta-analysis. Ann Oncol. 2014;25:1293–311.PubMed
125.
Zurück zum Zitat Scott JM, Li N, Liu Q, Yasui Y, Leisenring W, Nathan PC, Gibson T, Armenian SH, Nilsen TS, Oeffinger KC, Ness KK, Adams SC, Robison LL, Armstrong GT, Jones LW. Association of exercise with mortality in adult survivors of childhood cancer. JAMA Oncol. 2018;4:1352–8.PubMedPubMedCentral Scott JM, Li N, Liu Q, Yasui Y, Leisenring W, Nathan PC, Gibson T, Armenian SH, Nilsen TS, Oeffinger KC, Ness KK, Adams SC, Robison LL, Armstrong GT, Jones LW. Association of exercise with mortality in adult survivors of childhood cancer. JAMA Oncol. 2018;4:1352–8.PubMedPubMedCentral
126.
Zurück zum Zitat Beasley JM, Kwan ML, Chen WY, Weltzien EK, Kroenke CH, Lu W, Nechuta SJ, Cadmus-Bertram L, Patterson RE, Sternfeld B, Shu X-O, Pierce JP, Caan BJ. Meeting the physical activity guidelines and survival after breast cancer: findings from the after breast cancer pooling project. Breast Cancer Res Treat. 2012;131:637–43.PubMed Beasley JM, Kwan ML, Chen WY, Weltzien EK, Kroenke CH, Lu W, Nechuta SJ, Cadmus-Bertram L, Patterson RE, Sternfeld B, Shu X-O, Pierce JP, Caan BJ. Meeting the physical activity guidelines and survival after breast cancer: findings from the after breast cancer pooling project. Breast Cancer Res Treat. 2012;131:637–43.PubMed
127.
Zurück zum Zitat Jeon J, Sato K, Niedzwiecki D, Ye X, Saltz LB, Mayer RJ, Mowat RB, Whittom R, Hantel A, Benson A, Wigler DS, Atienza D, Messino M, Kindler H, Venook A, Fuchs CS, Meyerhardt JA. Impact of physical activity after cancer diagnosis on survival in patients with recurrent colon cancer: findings from CALGB 89803/alliance. Clin Colorectal Cancer. 2013;12:233–8.PubMed Jeon J, Sato K, Niedzwiecki D, Ye X, Saltz LB, Mayer RJ, Mowat RB, Whittom R, Hantel A, Benson A, Wigler DS, Atienza D, Messino M, Kindler H, Venook A, Fuchs CS, Meyerhardt JA. Impact of physical activity after cancer diagnosis on survival in patients with recurrent colon cancer: findings from CALGB 89803/alliance. Clin Colorectal Cancer. 2013;12:233–8.PubMed
128.
Zurück zum Zitat Meyerhardt JA, Heseltine D, Niedzwiecki D, Hollis D, Saltz LB, Mayer RJ, Thomas J, Nelson H, Whittom R, Hantel A, Schilsky RL, Fuchs CS. Impact of physical activity on cancer recurrence and survival in patients with stage III colon cancer: findings from CALGB 89803. J Clin Oncol. 2006;24:3535–41.PubMed Meyerhardt JA, Heseltine D, Niedzwiecki D, Hollis D, Saltz LB, Mayer RJ, Thomas J, Nelson H, Whittom R, Hantel A, Schilsky RL, Fuchs CS. Impact of physical activity on cancer recurrence and survival in patients with stage III colon cancer: findings from CALGB 89803. J Clin Oncol. 2006;24:3535–41.PubMed
129.
Zurück zum Zitat Haykowsky MJ, Scott JM, Hudson K, Denduluri N. Lifestyle interventions to improve cardiorespiratory fitness and reduce breast cancer recurrence. Am Soc Clin Oncol Educ Book. 2017;37:57–64.PubMed Haykowsky MJ, Scott JM, Hudson K, Denduluri N. Lifestyle interventions to improve cardiorespiratory fitness and reduce breast cancer recurrence. Am Soc Clin Oncol Educ Book. 2017;37:57–64.PubMed
130.
Zurück zum Zitat Casla S, Hojman P, Márquez-Rodas I, López-Tarruella S, Jerez Y, Barakat R, Martín M. Running away from side effects: physical exercise as a complementary intervention for breast cancer patients. Clin Transl Oncol. 2015;17:180–96.PubMed Casla S, Hojman P, Márquez-Rodas I, López-Tarruella S, Jerez Y, Barakat R, Martín M. Running away from side effects: physical exercise as a complementary intervention for breast cancer patients. Clin Transl Oncol. 2015;17:180–96.PubMed
133.
Zurück zum Zitat Schmitz KH, Courneya KS, Matthews C, Demark-Wahnefried W, GalvÃO DA, Pinto BM, Irwin ML, Wolin KY, Segal RJ, Lucia A, Schneider CM, Von Gruenigen VE, Schwartz AL. American college of sports medicine roundtable on exercise guidelines for cancer survivors. Med Sci Sports Exerc. 2010;42:1409–26.PubMed Schmitz KH, Courneya KS, Matthews C, Demark-Wahnefried W, GalvÃO DA, Pinto BM, Irwin ML, Wolin KY, Segal RJ, Lucia A, Schneider CM, Von Gruenigen VE, Schwartz AL. American college of sports medicine roundtable on exercise guidelines for cancer survivors. Med Sci Sports Exerc. 2010;42:1409–26.PubMed
134.
Zurück zum Zitat Jones LW, Eves ND, Peddle CJ, Courneya KS, Haykowsky M, Kumar V, Winton TW, Reiman T. Effects of presurgical exercise training on systemic inflammatory markers among patients with malignant lung lesions. Appl Physiol Nutr Metabol. 2009;34:197–202. Jones LW, Eves ND, Peddle CJ, Courneya KS, Haykowsky M, Kumar V, Winton TW, Reiman T. Effects of presurgical exercise training on systemic inflammatory markers among patients with malignant lung lesions. Appl Physiol Nutr Metabol. 2009;34:197–202.
135.
Zurück zum Zitat Ligibel JA, Irwin M, Dillon D, Barry W, Giobbie-Hurder A, Frank E, Winer EP, McTiernan A, Cornwell M, Pun M, Brown M, Jeselsohn R. Impact of pre-operative exercise on breast cancer gene expresision. In: Proceedings of the thirty-ninth annual CTRC–AACR San Antonio breast cancer symposium. San Antonio: American Association for Cancer Research; 2017. p. 77. Ligibel JA, Irwin M, Dillon D, Barry W, Giobbie-Hurder A, Frank E, Winer EP, McTiernan A, Cornwell M, Pun M, Brown M, Jeselsohn R. Impact of pre-operative exercise on breast cancer gene expresision. In: Proceedings of the thirty-ninth annual CTRC–AACR San Antonio breast cancer symposium. San Antonio: American Association for Cancer Research; 2017. p. 77.
136.
Zurück zum Zitat Furmaniak AC, Menig M, Markes MH. Exercise for women receiving adjuvant therapy for breast cancer. Cochrane Database Syst Rev. 2016;9:CD005001.PubMed Furmaniak AC, Menig M, Markes MH. Exercise for women receiving adjuvant therapy for breast cancer. Cochrane Database Syst Rev. 2016;9:CD005001.PubMed
137.
Zurück zum Zitat Loughney L, West MA, Kemp GJ, Grocott MPW, Jack S. Exercise intervention in people with cancer undergoing neoadjuvant cancer treatment and surgery: a systematic review. Eur J Surg Oncol. 2016;42:28–38.PubMed Loughney L, West MA, Kemp GJ, Grocott MPW, Jack S. Exercise intervention in people with cancer undergoing neoadjuvant cancer treatment and surgery: a systematic review. Eur J Surg Oncol. 2016;42:28–38.PubMed
138.
Zurück zum Zitat Loughney L, West MA, Kemp GJ, Grocott MPW, Jack S. Exercise intervention in people with cancer undergoing adjuvant cancer treatment following surgery: a systematic review. Eur J Surg Oncol. 2015;41:1590–602.PubMed Loughney L, West MA, Kemp GJ, Grocott MPW, Jack S. Exercise intervention in people with cancer undergoing adjuvant cancer treatment following surgery: a systematic review. Eur J Surg Oncol. 2015;41:1590–602.PubMed
139.
Zurück zum Zitat Beaudry RI, Liang Y, Boyton ST, Tucker WJ, Brothers RM, Daniel KM, Rao R, Haykowsky MJ. Meta-analysis of exercise training on vascular endothelial function in cancer survivors. Integr Cancer Ther. 2018;17:192–9.PubMedPubMedCentral Beaudry RI, Liang Y, Boyton ST, Tucker WJ, Brothers RM, Daniel KM, Rao R, Haykowsky MJ. Meta-analysis of exercise training on vascular endothelial function in cancer survivors. Integr Cancer Ther. 2018;17:192–9.PubMedPubMedCentral
140.
Zurück zum Zitat Nelson ME, Rejeski WJ, Blair SN, Duncan PW, Judge JO, King AC, Macera CA, Castaneda-Sceppa C. Physical activity and public health in older adults. Med Sci Sports Exerc. 2007;39:1435–45.PubMed Nelson ME, Rejeski WJ, Blair SN, Duncan PW, Judge JO, King AC, Macera CA, Castaneda-Sceppa C. Physical activity and public health in older adults. Med Sci Sports Exerc. 2007;39:1435–45.PubMed
141.
Zurück zum Zitat Turner RR, Steed L, Quirk H, Greasley RU, Saxton JM, Taylor SJC, Rosario DJ, Thaha MA, Bourke L. Interventions for promoting habitual exercise in people living with and beyond cancer. Cochrane Database Syst Rev. 2018;9:CD010192.PubMed Turner RR, Steed L, Quirk H, Greasley RU, Saxton JM, Taylor SJC, Rosario DJ, Thaha MA, Bourke L. Interventions for promoting habitual exercise in people living with and beyond cancer. Cochrane Database Syst Rev. 2018;9:CD010192.PubMed
142.
Zurück zum Zitat Jones LW, Liu Q, Armstrong GT, Ness KK, Yasui Y, Devine K, Tonorezos E, Soares-Miranda L, Sklar CA, Douglas PS, Robison LL, Oeffinger KC. Exercise and risk of major cardiovascular events in adult survivors of childhood hodgkin lymphoma: a report from the childhood cancer survivor study. J Clin Oncol. 2014;32:3643–50.PubMedPubMedCentral Jones LW, Liu Q, Armstrong GT, Ness KK, Yasui Y, Devine K, Tonorezos E, Soares-Miranda L, Sklar CA, Douglas PS, Robison LL, Oeffinger KC. Exercise and risk of major cardiovascular events in adult survivors of childhood hodgkin lymphoma: a report from the childhood cancer survivor study. J Clin Oncol. 2014;32:3643–50.PubMedPubMedCentral
143.
Zurück zum Zitat Sasso JP, Eves ND, Christensen JF, Koelwyn GJ, Scott J, Jones LW. A framework for prescription in exercise-oncology research. J Cachexia Sarcopenia Muscle. 2015;6:115–24.PubMedPubMedCentral Sasso JP, Eves ND, Christensen JF, Koelwyn GJ, Scott J, Jones LW. A framework for prescription in exercise-oncology research. J Cachexia Sarcopenia Muscle. 2015;6:115–24.PubMedPubMedCentral
144.
Zurück zum Zitat Padilha CS, Marinello PC, Galvão DA, Newton RU, Borges FH, Frajacomo F, Deminice R. Evaluation of resistance training to improve muscular strength and body composition in cancer patients undergoing neoadjuvant and adjuvant therapy: a meta-analysis. J Cancer Surviv. 2017;11:339–49.PubMed Padilha CS, Marinello PC, Galvão DA, Newton RU, Borges FH, Frajacomo F, Deminice R. Evaluation of resistance training to improve muscular strength and body composition in cancer patients undergoing neoadjuvant and adjuvant therapy: a meta-analysis. J Cancer Surviv. 2017;11:339–49.PubMed
145.
Zurück zum Zitat Jones LW, Courneya KS, Vallance JKH, Ladha AB, Mant MJ, Belch AR, Stewart DA, Reiman T. Association between exercise and quality of life in multiple myeloma cancer survivors. Support Care Cancer. 2004;12:780–8.PubMed Jones LW, Courneya KS, Vallance JKH, Ladha AB, Mant MJ, Belch AR, Stewart DA, Reiman T. Association between exercise and quality of life in multiple myeloma cancer survivors. Support Care Cancer. 2004;12:780–8.PubMed
146.
Zurück zum Zitat Mills RC. Breast cancer survivors, common markers of inflammation, and exercise: a narrative review. Breast Cancer Basic Clin Res. 2017;11:117822341774397. Mills RC. Breast cancer survivors, common markers of inflammation, and exercise: a narrative review. Breast Cancer Basic Clin Res. 2017;11:117822341774397.
147.
Zurück zum Zitat Gil-Rey E, Quevedo-Jerez K, Maldonado-Martin S, Herrero-Román F. Exercise intensity guidelines for cancer survivors: a comparison with reference values. Int J Sports Med. 2014;35:e1–9.PubMed Gil-Rey E, Quevedo-Jerez K, Maldonado-Martin S, Herrero-Román F. Exercise intensity guidelines for cancer survivors: a comparison with reference values. Int J Sports Med. 2014;35:e1–9.PubMed
148.
Zurück zum Zitat Toohey K, Pumpa K, McKune A, Cooke J, Semple S. High-intensity exercise interventions in cancer survivors: a systematic review exploring the impact on health outcomes. J Cancer Res Clin Oncol. 2018;144:1–12.PubMed Toohey K, Pumpa K, McKune A, Cooke J, Semple S. High-intensity exercise interventions in cancer survivors: a systematic review exploring the impact on health outcomes. J Cancer Res Clin Oncol. 2018;144:1–12.PubMed
149.
Zurück zum Zitat National Academies of Sciences EM, Health and Medicine Division. Board on health care services, national cancer policy forum. Incorporating weight management and physical activity throughout the cancer care continuum. In: Proceedings of a workshop. Washington, DC: National Academies Press; 2018. National Academies of Sciences EM, Health and Medicine Division. Board on health care services, national cancer policy forum. Incorporating weight management and physical activity throughout the cancer care continuum. In: Proceedings of a workshop. Washington, DC: National Academies Press; 2018.
150.
Zurück zum Zitat Tudor-Locke C, Bassett DR. How many steps/day are enough? Sports Med. 2004;34:1–8.PubMed Tudor-Locke C, Bassett DR. How many steps/day are enough? Sports Med. 2004;34:1–8.PubMed
151.
Zurück zum Zitat Chen X, Lu W, Zheng W, Gu K, Matthews CE, Chen Z, Zheng Y, Shu XO. Exercise after diagnosis of breast cancer in association with survival. Cancer Prev Res. 2011;4:1409–18. Chen X, Lu W, Zheng W, Gu K, Matthews CE, Chen Z, Zheng Y, Shu XO. Exercise after diagnosis of breast cancer in association with survival. Cancer Prev Res. 2011;4:1409–18.
152.
Zurück zum Zitat Carayol M, Bernard P, Boiché J, Riou F, Mercier B, Cousson-Gélie F, Romain AJ, Delpierre C, Ninot G. Psychological effect of exercise in women with breast cancer receiving adjuvant therapy: what is the optimal dose needed? Ann Oncol. 2013;24:291–300.PubMed Carayol M, Bernard P, Boiché J, Riou F, Mercier B, Cousson-Gélie F, Romain AJ, Delpierre C, Ninot G. Psychological effect of exercise in women with breast cancer receiving adjuvant therapy: what is the optimal dose needed? Ann Oncol. 2013;24:291–300.PubMed
153.
Zurück zum Zitat Valle C. Enhancing benefits of exercise for the survivor using emerging technology. In: ASCO Annual Meeting. Chicago; 201. Valle C. Enhancing benefits of exercise for the survivor using emerging technology. In: ASCO Annual Meeting. Chicago; 201.
154.
Zurück zum Zitat Joyner MJ, Sanchis-Gomar F, Lucia A. Exercise medicine education should be expanded. Br J Sports Med. 2017;51:625–6.PubMed Joyner MJ, Sanchis-Gomar F, Lucia A. Exercise medicine education should be expanded. Br J Sports Med. 2017;51:625–6.PubMed
155.
Zurück zum Zitat McDermott LA, Murphy MH, McNeilly AM, Rankin JP, Gracey JH. Biological markers as an outcome measure of exercise in cancer rehabilitation: a systematic review. J Cancer Res Ther. 2018;14:267–77.PubMed McDermott LA, Murphy MH, McNeilly AM, Rankin JP, Gracey JH. Biological markers as an outcome measure of exercise in cancer rehabilitation: a systematic review. J Cancer Res Ther. 2018;14:267–77.PubMed
156.
Zurück zum Zitat 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. Med Sci Sports Exerc. 2019;51:2375–90.PubMedPubMedCentral 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. Med Sci Sports Exerc. 2019;51:2375–90.PubMedPubMedCentral
157.
Zurück zum Zitat Jones LW, Peddle CJ, Eves ND, Haykowsky MJ, Courneya KS, Mackey JR, et al. Effects of presurgical exercise training on cardiorespiratory fitness among patients undergoing thoracic surgery for malignant lung lesions. Cancer. 2007;110:590–8.PubMed Jones LW, Peddle CJ, Eves ND, Haykowsky MJ, Courneya KS, Mackey JR, et al. Effects of presurgical exercise training on cardiorespiratory fitness among patients undergoing thoracic surgery for malignant lung lesions. Cancer. 2007;110:590–8.PubMed
158.
Zurück zum Zitat Jones LW, Hornsby WE, Goetzinger A, Forbes LM, Sherrard EL, Quist M, et al. Prognostic significance of functional capacity and exercise behavior in patients with metastatic non-small cell lung cancer. Lung Cancer. 2012;76:248–52.PubMed Jones LW, Hornsby WE, Goetzinger A, Forbes LM, Sherrard EL, Quist M, et al. Prognostic significance of functional capacity and exercise behavior in patients with metastatic non-small cell lung cancer. Lung Cancer. 2012;76:248–52.PubMed
159.
Zurück zum Zitat Travier N, Velthuis MJ, Steins Bisschop CN, van den Buijs B, Monninkhof EM, Backx F, et al. Effects of an 18-week exercise programme started early during breast cancer treatment: a randomised controlled trial. BMC Med. 2015;13:121.PubMedPubMedCentral Travier N, Velthuis MJ, Steins Bisschop CN, van den Buijs B, Monninkhof EM, Backx F, et al. Effects of an 18-week exercise programme started early during breast cancer treatment: a randomised controlled trial. BMC Med. 2015;13:121.PubMedPubMedCentral
Metadaten
Titel
Exercise and cancer: a position statement from the Spanish Society of Medical Oncology
verfasst von
M. Pollán
S. Casla-Barrio
J. Alfaro
C. Esteban
M. A. Segui-Palmer
A. Lucia
M. Martín
Publikationsdatum
13.02.2020
Verlag
Springer International Publishing
Erschienen in
Clinical and Translational Oncology / Ausgabe 10/2020
Print ISSN: 1699-048X
Elektronische ISSN: 1699-3055
DOI
https://doi.org/10.1007/s12094-020-02312-y

Weitere Artikel der Ausgabe 10/2020

Clinical and Translational Oncology 10/2020 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.