Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Jul 15.
Published in final edited form as: Cancer Res. 2016 Jul 5;76(14):4032–4050. doi: 10.1158/0008-5472.CAN-16-0887

Efficacy and Mechanisms of Aerobic Exercise on Cancer Initiation, Progression, and Metastasis: A Critical Systematic Review of In Vivo Preclinical Data

Kathleen A Ashcraft 1, Ralph M Peace 1, Allison S Betof 2, Mark W Dewhirst 1,*, Lee W Jones 3,*
PMCID: PMC5378389  NIHMSID: NIHMS794584  PMID: 27381680

Abstract

A major objective of the emerging field of exercise-oncology research is to determine the efficacy of, and biological mechanisms by which, aerobic exercise affects cancer incidence, progression and/or metastasis. There is a strong inverse association between self-reported exercise and the primary incidence of several forms of cancer; similarly, emerging data suggest that exercise exposure following a cancer diagnosis may improve outcomes for early-stage breast, colorectal, or prostate cancer. Arguably, critical next steps in the development of exercise as a candidate treatment in cancer control require preclinical studies to validate the biological efficacy of exercise, identify the optimal “dose”, and pinpoint mechanisms of action. To evaluate the current evidence base, we conducted a critical systematic review of in vivo studies investigating the effects of exercise in cancer prevention and progression. Studies were evaluated on the basis of tumor outcomes (e.g., incidence, growth, latency, metastasis), dose-response, and mechanisms of action, when available. A total of 53 studies were identified and evaluated on tumor incidence (n=24), tumor growth (n=33) or metastasis (n=10). We report that the current evidence base is plagued by considerable methodological heterogeneity in all aspects of study design, end points, and efficacy. Such heterogeneity precludes meaningful comparisons and conclusions at present. To this end, we provide a framework of methodological and data reporting standards to strengthen the field to guide the conduct of high-quality studies required to inform translational, mechanism-driven clinical trials.

Keywords: Exercise, Physical Activity, Mouse Models, Rat Models, Cancer, Carcinogenesis, Metastasis, Mechanisms

INTRODUCTION

Structured exercise training (hereto referred to as exercise) is considered an integral component of “standard of care” therapy in primary and secondary prevention of numerous common chronic conditions (14). In comparison, the role of exercise has received surprisingly little attention in individuals at high-risk or following a diagnosis of cancer. Over the past two decades however, an increasing number of groups are investigating the effects of general physical activity as well as exercise in the oncology setting, a field now commonly referred to as “Exercise-Oncology” (5).

A strong body of observational evidence indicates that higher levels of self-reported exercise, physical activity, as well as cardiorespiratory fitness (i.e., objective assessment of exercise exposure) are inversely associated with the primary incidence of several forms of cancer. This evidence base is summarized by several excellent systematic reviews and meta-analyses (69). For example, exercise participation consistent with the national guidelines (i.e., 150 minutes of moderate-intensity or 75 minutes of vigorous-intensity exercise per week) is associated, on average, with 25%, and 30% to 40% risk reductions in breast and colon cancers, respectively compared with inactivity. There is also evidence of a linear dose-response relationship in prevention of breast and colon cancer. On this basis, the evidence for the exercise-prevention relationship is categorized as “convincing” for breast and colon cancer by several national agencies, and regarding as “encouraging” or “promising” for the prevention of prostate, lung, and endometrial cancers (10, 11). On the basis of this data, several phase II randomized controlled trials (RCTs) were initiated, predominantly in breast cancer prevention, to investigate the effects of highly-structured exercise on modulation of host-related factors (e.g., adiposity, circulating factors including sex-steroid and metabolic sex hormones, and the immune-inflammatory axis) that may underpin the exercise-cancer prevention relationship (6, 1214). In general, exercise was associated with modest changes in markers of adiposity and select circulating factors (6, 12, 13). Whether the observed alterations are of biological or clinical importance remains to be determined, as only one trial to date has investigated whether exercise-induced changes in circulating factors occur in conjunction with changes in factors/pathways in the organ/tissue of primary interest (i.e., colon crypts) (15). Whether exercise reduces cancer incidence or modulates established surrogate markers of cancer incidence has not been investigated.

In patients with cancer, a steadily growing and ever diversifying series of studies indicate that, in general, exercise is a tolerable adjunct therapy associated with significant benefit across a wide range of symptom control variables both during and after primary adjuvant therapy (5, 16, 17). This data combined with the powerful inverse relationships with primary cancer incidence has led researchers to investigate whether exercise influences disease outcomes after diagnosis (18, 19). Although not nearly as mature as the evidence in the prevention setting, emerging observational data suggest that regular exercise exposure is associated with between a 10%–50% reduction in the risk of recurrence and cancer-specific death in patients with colorectal, breast, and prostate cancer (reviewed in (20)), compared to inactivity.

In the development of potential drug candidates, data from observational studies is insufficient to support the initiation of human trials; appropriate preclinical evidence is first required prior to human testing (21). While exercise is not a drug and exhibits a markedly different safety profile than most anticancer agents, the use of preclinical models are of critical importance to confirm biological plausibility, establish the therapeutic window of efficacy, a biologically effective dose, and identify predictors of response (22). This evidence, combined with data from epidemiological and molecular epidemiological studies facilitates the design of early “signal-seeking” clinical studies and ultimately definitive RCTs. Accordingly, we conducted a critical systematic review of in vivo pre-clinical studies across the cancer continuum (i.e., prevention through metastasis). A secondary purpose was to provide recommendations to facilitate the standardization of the conduct of in vivo exercise-oncology studies as well as directions for future research.

METHODS

Search Strategy and Inclusion Criteria

A systematic literature search was conducted using OVID MEDLINE (1950 to May 2015), PUBMED (1962 to May 2015) and WEB OF SCIENCE (1950 to May 2015) with MeSH terms and keywords related to exercise, cancer and animals. Text words were searched and appropriate MeSH terms were found (Table S1). When possible, Boolean logic was used with MeSH terms to build searches. All results and reference lists were searched manually. Peer-reviewed research articles involving animals with cancer and exposed to chronic exercise (repeated bouts of more than 3 sessions) adopting either forced (i.e., treadmill running or swimming) endurance (aerobic) training or physical activity (i.e., voluntary wheel running) paradigms were considered eligible. All types of animal models of solid tumors were considered eligible including genetically predisposed models (transgenic, genetically engineered mouse models [GEMMs]), orthotopic, subcutaneous and intravenous injections, tumor transplant, and spontaneous or carcinogen-induced solid tumor models. Studies that included multiple treatments, such as study arms with dietary restrictions, were eligible, as long as it was possible to compare sedentary (control) and exercise alone groups. Studies that evaluated pre-neoplastic lesions (e.g., aberrant crypt foci, ApcMin), or hematopoietic and ascites tumors were excluded. Only mouse and rat studies were included. Papers unavailable in English were excluded.

Study Selection and Classification

Four authors (KAA, RMP, ASB and LWJ) assessed study eligibility by reviewing the titles and abstracts of all potential citations according to the inclusion criteria. KAA, RMP, MWD and ASB extracted and interpreted data from published papers. When necessary, effort was made to contact authors and acquire publications for evaluation. Studies are summarized by type of exercise model and method of tumor initiation. Exercise characteristics are described using common prescription elements: modality, frequency, duration, intensity and total length of intervention.

Tumor initiation vs. growth

Papers were classified by the endpoints reported in the individual studies: (1) “Incidence” (tumor presence or multiplicity) (2) “Growth” (data on tumor growth, latency or survival), and (3) “Metastasis” (evaluation of tumors distinct from the primary tumor or developing following the common metastasis model of intravenous tumor cell injection). Study categorization was not always mutually exclusive; thus studies that included multiple endpoints applicable to more than one categorization were included in both.

Analysis of mechanistic findings

For evaluation of mechanistic findings, an alternative classification was applied based on the initiation of the exercise intervention relative to the time of tumor cell transplant or application of carcinogens. “Prevention” was defined as exercise initiated prior to tumor transplant/induction. “Progression” was defined as exercise initiated ≥3 days post-transplant/induction. Studies involving GEMMs were categorized as prevention studies. Distinguishing between exercise initiation and tumor cell inoculation is important because exercise-induced adaptations prior to tumor injection could ‘prime’ the host and/or tissue microenvironment, making it physiological or biologically distinct from tumor inoculation into a sedentary host. Mechanistic findings were classified as either intratumoral or systemic.

Interpretation and analysis

Studies were assessed for changes in tumor growth (as well as the related parameters of time to tumor-related endpoint, and tumor size/mass at the end of the study), incidence (tumor presence) and multiplicity (number of tumors/metastases). All study results that were reported to be “statistically significant” achieved p<0.05 according to the authors of the original manuscripts. Preliminary analysis of included studies indicated considerable methodological heterogeneity in all aspects of study design, end points, and efficacy. As such, it was not possible to compare the efficacy of exercise across cancer setting (i.e., incidence, progression, metastasis), cancer histology, exercise paradigm (i.e., forced vs. voluntary), or exercise dose.

RESULTS

A total of 426 potential citations were initially identified using search terms. After secondary screening, 53 papers were deemed eligible and underwent full review (Figure S1). Classification of papers was as follows: (1) incidence (n=24; 45.3%) (2346), growth (n=33; 62.3%) (24, 26, 34, 37, 39, 40, 42, 4469), and metastasis (n=10; 18.9%) (53, 55, 63, 64, 7075).

Tumor Models and Exercise Prescription Characteristics

Included studies tested the effects of exercise on 15 different tumor types/cell lines, using six different tumor initiation methods (Table S2). Details of the exercise prescriptions are summarized in Tables 1, 2 and 3. The most common modalities included voluntary running (n=22; 41.5%) (2428, 31, 33, 35, 42, 43, 51, 52, 55, 57, 6466, 70, 71, 7375) forced running (n=25; 47.2%) (26, 29, 3641, 4446, 48, 49, 54, 56, 5860, 63, 6770, 72, 74) and swimming (n=10; 18.9%) (23, 30, 32, 34, 47, 50, 53, 54, 61, 62).

Table 1.

Tumor Incidence Studies

METHODS RESULTS
Study Reference Rodent
Model
Tumor Type /
Induction model
Exercise Protocol Tumor Incidence Results
Exercise Modality Exercise Prescription
Freq/week | Dur | Intens | Length
Exercise Initiation
Andrianopoulos,
1987
25 5w old male
Sprague-
Dawley rats
Intestinal / DMH, i.p.
q1w for 6w
Voluntary wheel
running
Wheel running 1w prior to first injection -Tumors present in 18/20 SED rats and 6/11 EX rats
Reddy, 1988 33 Male F344
rats
Intestinal / Subcutaneous
AOM 15mg/kg BW,
q1wx2w at 7 wk of age.
Voluntary wheel
running
Wheel running for 38 weeks 4d post-AOM -EX ↓ incidence and multiplicity adenocarcinomas, and liver foci. of colon and
Sugie, 1992 35 5w old F-344
rats
Hepatocellular / 15
mg/kg AOM s.c. q1w for
2w
Voluntary wheel
running
38w 4d post-injection. -Liver tumors noted in 7% of AOM treated SED only.
Ikuyama, 1993 28 Jc1:Wistar
rats
Hepatoma / 0.0177
g/day/kg BW dietary 3'-
Me-DAB for 35 weeks.
Voluntary wheel
running
Wheel running for 62 weeks, using
food as a reward for achieving
specified distances
17w prior to dietary 3'-Me-
DAB
-65% reduction in tumor incidence.
Zhu, 2008 42 21d old
female
Sprague-
Dawley rats
Mammary / 25 or 50
mg/kg MNU, i.p.
Voluntary wheel
running
Voluntary wheel running for 8w 1w post-injection -84.5% incidence vs 98.1%, (SED vs. EX)
Esser, 2009 27 C3(1)Tag
mice
Prostate / Transgenic
mouse model
Voluntary wheel
running
Wheel running for 10 weeks
Data analyzed based on mice that
ran >5K or <5K a day
10w of age -18% (>5K) and 55% (<5K) of animals with high grade neoplasia at 20 weeks
-57% reduction in incidence of high grade neoplasia in >5K vs <5K mice
Alessio, 2009 24 3w old
female
Sprague-
Dawley rats
Spontaneous tumors Voluntary wheel
running or activity box
Wheel: every other day, 24h access
throughout animals’ life

Activity box (PA): 1h in large
activity box twice a week for life.
Spontaneous tumors, animals
followed for life.
-During weeks 60–120, 38% of EX rats were tumor-bearing animals (vs. 42% PA rats and 54% SED rata).
-At week 88, tumor multiplicity was 0.69 for EX animals, 0.75 for PA, and 0.96 for SED.
Colbert, 2009 26 Female
heterozygous
(p53+/−):
MMTV-Wnt-
1 transgenic
mice
Mammary / Transgenic Voluntary wheel
running and forced
treadmill running
Treadmill running:
TREX 1: 5×|45min|20m/min at 5%
grade|until completion
TREX 2: 5×|45min|24m/min at 5%
grade|until completion
Voluntary wheel running with 24
hour access.
11w of age -Tumor incidence ↑ in wheel running mice by 32%
Mann, 2010 31 21d old
female
Sprague-
Dawley rats
Mammary / 50 mg/kg
MNU i.p.
Voluntary non-
motorized and
motorized wheel
running
Voluntary non-motorized and
motorized (40m/min) wheel
running
1w post-injection -96%, 74% and 70% incidence in controls, non-motorized and motorized mice, respectively
Zhu, 2012 43 21d old
female
Sprague-
Dawley rats
Mammary / 50 mg/kg
MNU i.p.
Voluntary wheel
running at a fixed daily
distance
Three levels:
WR-High: maximum 3500m/d
WR-Low: maximum 1750m/d.
SED control.
1w post-injection -97% tumor incidence in controls, 80% tumor incidence in WR-High
-Cannot compare WR-High and WR-Low, because dietary energy restriction was applied to WR-Low only
Thorling, 1993 36 5w old male
Fischer rats
Intestinal / 15mg/kg
AOM s.c. on Days 1, 4
and 8.
Forced treadmill
running
5×|2km/day|7m/min|38w.

First week was acclimatization.
3d after last injection -EX ↓ colon neoplasia incidence, 53% vs 78% in
Woods, 1994 40 6w old male
C3H/HeN
mice
Mammary / 2.5 ×105
mammary SCA-1 cells
s.c. in the back.
Forced treadmill
running
Treadmill running: Moderate:
7×|30min|18m/min at 5% grade|1w.
Exhaustive: 7×|varied|18m/min for
30min, then 3m/min↑ every 30min
until exhausted|1w
3d prior to injection. -Increase in tumor incidence at Day 7 in both EX groups, but no differences in any subsequent time points.
Whittal, 1996 38 21d old
female
Sprague-
Dawley Rats
Mammary / 50 mg/kg
NMU i.p. at 50d of age
Forced treadmill
running
Progressive training to
5×|60min|18 m/min at 15%
grade|4w
21d of age (29d prior to
injection)
-Incidence/multiplicity at 24w post-NMU: 58 tumors in SED rats, 33 tumors in EX rats
-No significant difference in latency or incidence
Whittal-Strange,
1998
39 21d old
female
Sprague-
Dawley rats
Mammary / 37.5mg/kg
NMU i.p. at 50d of age,
(1 day after last bout of
EX)
Forced treadmill
running
Progressive training to
5×|60min|18 m/min at 15%
grade|4w
21d of age (29d prior to
injection)
-Incidences of carcinomas, high grade and low grade tumors were 29.2%, 10.4%, and 25% in SED, and 38%,
14.3% and 21.4% in EX
Westerlind,
2003
37 21d old
female
Sprague-
Dawley rats
Mammary / 25 or 50
mg/kg MNU, i.p.
Forced treadmill
running
Week 1: 5×|10–15min|30m/min|1w.
Week 2–9: 5×|30min|23–
25m/min|8w
1w post-injection -↑ Latency in EX (35.8d vs. 33.1d).
-No difference in median tumor-free survival time was observed in the EX versus sham-EX (SHAM), nor
were there any differences in multiplicity at either a high or moderate dose of MNU
Zielinski, 2004 44 6–8w old
female
BALB/c mice
Neoplastic Lymphoid
cells / 2×107 EL-4 cells
s.c. in the back behind
the neck
Forced treadmill
running
7×|3h or until volitional
fatigue|gradually increasing speed,
20–40m/min, 5% grade|5–14d
First session immediately
before injection.
-EX ↓ tumor appearance
Zhu, 2009 41 21d old
female
Sprague-
Dawley rats
Mammary / 50 mg/kg
MNU, i.p.
Forced wheel running Motorized running wheel; details
not provided
1w post-injection -66.7% and 92.6% incidence in EX and SED
Kato, 2011 29 5w old male
Fischer 344
rats
Renal / 5mg/kg BW Fe-
NTA i.p. once a day for
7 days, then 10mg/kg
Fe-NTA 3×/wk for 11w.
Forced treadmill
running
Short-term: 15m|8m/min, 0%|12w.

Long-term: 15m|8m/min, 0%|12w;
then 5×|30m|8m/min, 0%|12w for a
total of 24w training Exercise
continued until 40 weeks, but at
lower intensity to account for the
decline in rat health.
Training done 15m before each
injection
-No differences in number of rats with nodules, nodules/rat or mean area of nodules.
-Short-term EX ↑ rats with microcarcinomas,
- Long-term EX ↓ rats cinomas, karyomegalic cells and degenerative tubules compared to with microcar
short-term.
Malicka, 2015 45 4w old
female
Sprague
Dawley rats
Mammary / 180 mg/kg
MNU i.p.
Forced treadmill
running
Low intensity (LIT): 5×|10–
35min|0.48–1.34 km/h|12w

Moderate intensity (MIT): 5×|10–
35min|0.6–1.68 km/h|12w

High intensity (HIT): 5×|10–
35min|0.72–2.0 km/h|12w
Immediately after MNU
injection
-Incidence 64%, 67%, 40% and 43% in SED, LIT, MIT and HIT groups, respectively (Not significant)
-Multiplicity: Rats with tumors had an average of 2.4, 1.6, 1 and 1 tumors in SED, LIT, MIT and HIT
groups, respectively (Statistics not performed.)
Piguet, 2015 46 7–9w old
male
AlbCrePten
flox/flox
mice
Hepatocellular
carcinoma / Transgenic
Forced treadmill
running
5w acclimation period followed by:
5×|60m|12.5m/min|27w
7–9w of age -Tumor incidence 100% in SED vs. 71% in EX
Lunz, 2008 30 11w old male
Wistar Rats
Intestinal / 4 s.c.
injections DMH 3 days
apart.
Forced
swimming with 0%,
2% or 4% BW load
Week 1–2: 5×|5–20min|- load|2w
Week 3–5: 5×|5–20min|+ load|3w
Week 6– 35: 5×|20min|+load|30w
24h post first injection -No difference in tumor incidence
-Aerobic swimming training with 2% body weight of load protected against the DMH-induced preneoplastic
colon lesions, but not against tumor development in the rat
Paceli, 2012 32 Adult male
Balb/c mice
Lung / 1.5mg/kg BW
urethane i.p. twice, 2
days apart
Forced swimming Aerobic:
4×|20m|--|19w
Week 1: 10m/d to 50m in 5 days
Anaerobic:
3×|20m (2m swimming/2m
resting)|progressive loading of 5–
20%BW|20w
Within a week after injection -No significant effects of aerobic training on lung cancer incidence.
-Aerobic training resulted in 8 lung nodules per animal vs 52 in the control. Not significant.
-Median control nodules was 2.0, median aerobic control nodules was 0.0. Not significant.
Abdalla, 2013 23 8w old
female Balb/c
mice
Mammary / 1mg/ml
DMBA p.o. once weekly
for 6 weeks
Forced swimming 5×|45m|--|8 weeks
Water temperature 30+/−4°C
Same as tumor initiation -EX ↓ tumor incidence
Sáez Mdel,
2007
34 50d old
female
Sprague-
Dawley rats
Mammary / 5mg/w
DMBA gastric
intubation for 4w
Forced swimming 30m/d, 5d/w for 38–65d. 1d after appearance of first
tumor
-No difference in survival time or tumor multiplicity

EX = exercise or activity groups; SED = Sedentary controls; NMU = nitrosomethylurea; MNU= 1-methyl-1-nitrosourea; AOM= azoxymethane; DMBA= 7,12-dimethylbenz(a)anthracene; DMH= 1,2-dimethylhydrazine; 3’-Me-DAB= 3’-methyl-4-dimethylaminoazobenzene; Fe-NTA= ferric nitrilotriacetate; s.c.= sub-cutaneous; i.v.= intravenous

Table 2.

Tumor Growth Studies

METHODS RESULTS
Study Reference Rodent Model Tumor Type /
Induction model
Exercise Protocol Tumor Progression Results
Exercise Modality Exercise Prescription
Freq/week | Dur | Intens | Length
Exercise Initiation
Daneryd, 1995 51 Female Wistar
Furth rats
Leydig cell / 1.5mm3
injection of
Leydig cell sarcoma (LTW)
Voluntary wheel
running
13d Immediately after injection -EX ↓ tumor volume by 34%.
Daneryd, 1995 52 Female Wistar
Furth rats
Leydig cell / 1.5mm3
injection of Leydig
cell sarcoma (LTW)
or Nitrosoguanine-
induced
adenocarcinoma s.c.
into each flank
Voluntary wheel
running
32d Immediately after injection - 13.4g vs 16.4g EX vs SED final LTW weights
Zhu, 2008 42 21d old female
Sprague-Dawley
rats
Mammary / 25 or 50
mg/kg MNU, i.p.
Voluntary wheel
running
Voluntary wheel running for 8w 1w post-injection -Tumor weight 0.62g vs. 1.16g (SED vs. EX)
Jones, 2010 57 3–4w old female
athymic mice
Mammary / 1×106
MDA-MB-231 cells,
injected
orthotopically
Voluntary wheel
running
Voluntary wheel running for 41–
48d
2d post-implant No change in primary tumor growth (EX ↑21%)
Yan, 2011 64 3w old male
C57BL/6 mice
Lung / 2.5×105/50
µl/mouse Lewis lung
carcinoma cells s.c.
lower dorsal region
Voluntary wheel
running
Primary tumors excised at 1cm
diameter, access to wheels
continued for additional 2w.
9w before tumor implantation -No difference in tumor cross-sectional area and tumor volume.
Jones, 2012 55 6–8w old male
C57BL/6 mice
Prostate / 5×105
mouse prostate C-1
cells, orthotopically
Voluntary wheel
running
Wheel running for 8 weeks 14d after tumor transplant -Primary tumor growth was comparable between groups
Goh, 2014 66 18m old Balb/cBy
mice
Mammary / 1×104
cells in 4th mammary
fat pad
Voluntary wheel
running
Voluntary wheel running for 90
days
60 days prior to tumor transplant,
followed by 30 days post-transplant
-Inverse relationship between distance run and final tumor mass
Betof, 2015 65 Female Balb/c or
female C57Bl/6
mice
Mammary / 5×105
4T1-luc or 2.5×105
E0771 cells in dorsal
mammary fat pad
Voluntary wheel
running
Voluntary wheel running beginning
either 9 weeks prior to tumor
transplant, or at the time of tumor
transplant
SS: SED before and after transplant
RS: EX for 9 weeks prior to
transplant; SED after transplant
SR: SED before transplant; EX
after transplant
RR: EX 9 weeks prior to transplant,
continuing after transplant
- Growth rates of SS and RS were similar, and growth rates of SR and RR were similar
- EX slowed tumor growth compared to SED (both tumor models)
Alessio, 2009 24 3w old female
Sprague-Dawley
rats
Spontaneous tumors Voluntary wheel
running or activity box
Wheel: every other day, 24h access
throughout animals’ life.

Activity box (PA): 1h in large
activity box twice a week for life.
Spontaneous tumors, animals
followed for life.
- EX ↓tumor growth rate
Colbert, 2009 26 Female
heterozygous
(p53+/−):
MMTV-Wnt-1
transgenic mice
Mammary /
Transgenic
Voluntary wheel
running and forced
treadmill running
Treadmill running:
TREX 1: 5×|45min|20m/min at 5%
grade|until completion
TREX 2: 5×|45min|24m/min at 5%
grade|until completion
Voluntary wheel running with 24
hour access.
11w of age - Time to tumor size of 1.5cm: 24.8d, 13.8d, and 19.5d in control, TREX1 and TREX2 animals.
- Treadmill running led to faster tumor growth, no difference due to voluntary wheel running
- Treadmill running ↓ survival
Newton, 1965 60 45d old Sprague-
Dawley rats
Carcinoma / Equal
volumes of Walker-
256 cells s.c. into the
right flank.
Forced treadmill
running
Pre-Tumor: 50h over 5d at
950ft/hr.
Post-Tumor: 138h over 10d at 950
ft/hr.
5d before tumor implantation +/−
4d after tumor implantation
-EX ↓ final tumor weight vs SED control.
-Early life manipulation + EX ↓ final tumor weight vs EX alone and SED.
Uhlenbruck,
1991
63 BALB/c mice Sarcoma / 2.4×104 L-
1 cells s.c.
Forced treadmill
running
7×|distances of
200m/400m/800m|0.3m/s|2w
4w before and 2w after injection -200m group ↓ tumor weight
Woods, 1994 40 6w old male
C3H/HeN mice
Mammary / 2.5 ×105
mammary SCA-1
cells s.c. in the back.
Forced treadmill
running
Treadmill running: Moderate:
7×|30min|18m/min at 5% grade|1w.
Exhaustive: 7×|varied|18m/min for
30min, then 3m/min↑ every 30min
until exhausted|1w
3d prior to injection. No difference in growth rate of tumor size at time of euthanasia (two weels)
Whittal-Strange,
1998
39 21d old female
Sprague-Dawley
rats
Mammary /
37.5mg/kg NMU i.p.
at 50d of age, (1 day
after last bout of EX)
Forced treadmill
running
Progressive training to
5×|60min|18 m/min at 15%
grade|4w
21d of age (29d prior to injection) -Tumor growth rate at 22w post-NMU: 0.043g/day in SED vs. 0.107g/day in EX
-Final tumor weight: (3.2g in SED vs. 1.2g in EX
Bacurau, 2000 49 12w old male
Wistar rats
Carcinoma / 2×107
Walker-256 cells s.c.
in the flank
Forced treadmill
running
5×|60min|60% of VO2 peak|10w 8w prior to injection -Day 15 tumor weight 1.82% vs 19% of BW (EX vs SED)
-EX prolonged survival by 1.9 fold
Westerlind,
2003
37 21d old female
Sprague-Dawley
rats
Mammary / 25 or 50
mg/kg MNU, i.p.
Forced treadmill
running
Week 1: 5×|10–15min|30m/min|1w.
Week 2–9: 5×|30min|23–
25m/min|8w
1w post-injection -↑ Latency in EX (35.8d vs. 33.1d).
-No difference in median tumor-free survival time was observed in the EX versus sham-EX (SHAM),
nor were there any differences in multiplicity at either a high or moderate dose of MNU
Zielinski, 2004 44 6–8w old female
BALB/c mice
Neoplastic lymphoid
cells / 2×107 EL-4
cells s.c. in the back
behind the neck
Forced treadmill
running
7×|3h or until volitional
fatigue|gradually increasing speed,
20–40m/min, 5% grade|5–14d
First session immediately before
injection.
-No difference in tumor density
Jones, 2005 56 3–4w old female
athymic mice
Mammary / Flank
injection of 5×106
MDA-MB-231 cells
Forced treadmill
running
5d/w|10m/min for 10min up to
18m/min for 45 min|0% grade|8w
14d post-injection -No change in tumor growth.
Bacurau, 2007 48 8w old male
Wistar rats
Carcinoma / 2×107
Walker-256 cells s.c.
in the flank
Forced treadmill
running
5×|30min|85% of VO2 max|10w 8w prior to injection -Survival: 16d for SED, 45d for EX
-EX tumors were 6.9% of final body mass vs. 17.33% for SED control.
Lira, 2008 58 Male Wistar rats Carcinoma / 2×107
Walker-256 cells s.c.
in the flank
Forced treadmill
running
5×|60min|60–65% of VO2
peak|10w

2w pre-training period: rats ran
progressively from 15 to 60min at
10m/min. Running was increased
to 20m/min for two weeks after
injection.
8w prior to injection -Tumor weight: 17.2g in SED; 1.9g in EX
Murphy, 2011 59 4w old
C3(1)SV40Tag
mice
Mammary /
Transgenic (tumors
began developing at
12w of age).
Forced treadmill
running
6×|60m|20m/min at 5%|20w 4w of age -Tumor volume: ↓ in EX at 21 and 22w.
Gueritat, 2014 69 10–12w old
Copenhagen rats
Prostate / surgical s.c.
implantation of
R3327 Dunning AT1
tumor fragment
Forced treadmill
running
5×|15–60m|20–25 m/min|5w 15d after tumor implantation - EX rats had smaller tumors at 14 and 21 days compared to SED controls
- Tumor doubling time was significantly slower in EX vs. SED (6.19d vs. 8.81d)
Shalamzari,
2014
67 4–6w old Balb/c
mice
Mammary / 1×106
MC4-L2 s.c. in the
flank
Forced treadmill
running
-|20–40min|6–20 m/min|15w RTR: Sedentary before and after
transplant
RTE: EX after tumor transplant
only
ETR: EX before tumor transplant
only
ETE: EX before and after
transplant

9w before tumor transplant, and/or
6w after transplant
- ETE had significantly slower growth compared to RTR
- No difference in final tumor volume of RTE and ETR groups
Aveseh, 2015 68 5w old female
Balb/c mice
Mammary / 1.2×106
MC4-L2 cells in right
dorsal mammary fat
pad
Forced treadmill
running
7×|20–55min|10–20 m/min|7w 10d after tumor transplant -EX decreased tumor volume
Malicka, 2015 45 4w old female
Sprague Dawley
rats
Mammary / 180
mg/kg MNU i.p.
Forced treadmill
running
Low intensity (LIT): 5×|10–
35min|0.48–1.34 km/h|12w

Moderate intensity (MIT): 5×|10–
35min|0.6–1.68 km/h|12w

High intensity (HIT): 5×|10–
35min|0.72–2.0 km/h|12w
Immediately after MNU injection -No difference in final volume of total tumor volume
Piguet, 2015 46 7–9w old male
AlbCrePten
flox/flox mice
Hepatocellular
carcinoma /
Transgenic
Forced treadmill
running
5w acclimation period followed by:
5×|60m|12.5m/min|27w
7–9w of age -EX decreased total volume of liver tumors
Hoffman, 1962 54 Wistar rats Carcinoma / 2mL
Walker 256 cell
suspension s.c. into
the right thigh.
Continuous running on
a 20ft runway +
swimming + revolving
drum
21d of EX, all EX did all 3
modalities each day:
Runway: continuous running on
20ft runway, duration and intensity
not clear
Swimming: increasing 20min/day
to 4h/day
Revolving drum: 5.4 mi in 12h
Immediately after injection -97% inhibition of tumor growth.
-Tumor weight ↓ in EX group
Gershbein, 1974 53 Holtzman rats Carcinosarcoma /
Walker-256 tumor
i.m. into both
hindlimbs.
Forced swimming 10×|15min|--|10d Immediately after injection. -EX ↓ tumor size.
-No change in survival rates.
Baracos, 1989 50 Sprague-Dawley
rats
Hepatoma / 20uL
Morris hepatoma 777
s.c.
Forced swimming Low: 5×|5min/d, increased by
5min/d for 3w.
Medium: 5×|10min/d, increased by
10min/d for 3w.
High: 5×|15min/d, increased by
15min/d for 3w.
2 Groups
- 3w of swimming, tumor
transplant, then 3w additional
swimming.
-3w of swimming beginning 3d
post-transplant
-EX ↓ final tumor weight, both groups.
Radak, 2002 61 Adult female
hybrid BDF1
mice
Solid leukemia /
5×106 P-388
lymphoid leukemia
cells s.c.
Forced swimming 5×|60min|--|10w

ET: training terminated at tumor
implantation.
EC: training continued for 18d after
tumor implantation.
10w before injection. -EC animals had slower tumor growth than ET and control, (endpoints were ~1.24cm3 vs. 2cm3 and
2.4cm3)
SáezMdel, 2007 34 50d old female
Sprague-Dawley
rats
Mammary / 5mg/w
DMBA gastric
intubation for 4w
Forced swimming 30m/d, 5d/w for 38–65d. 1d after appearance of first tumor -EX ↑ tumor growth by 200%.
Almeida, 2009 47 7w old male
Swiss mice
Carcinoma / 2×106
Ehrlich tumor cells
s.c. in the dorsum.
Forced swimming 5×|60min|50/80% max test|6w.
Progressive load test began after
1w: load increased by 2% BW
every 3min until exhaustion.
4w before injection -50% workload ↓ tumor weight and tumor volume (0.18mg/g and 0.11mm3) vs control (0.55mg/g and
0.48mm3)
-Tumor volume and weight were 270% and 280% ↑ in SED mice.
Sasvari, 2011 62 Adult female
BDF1 mice
Sarcoma / 5×106 S-
180 cells s.c.
injected.
Forced swimming 5×|60m|--|10w

ETT: cells injected after EX.
ETC: EX (10w), cells injected, EX
(18 additional days)
10w before tumor implantation. -ETC ↓ final tumor weight vs. SED control.

EX = exercise or activity groups; SED = Sedentary controls; NMU = nitrosomethylurea; MNU= 1-methyl-1-nitrosourea; AOM= azoxymethane; DMBA= 7,12-dimethylbenz(a)anthracene; DMH= 1,2-dimethylhydrazine; 3’-Me-DAB= 3’-methyl-4-dimethylaminoazobenzene; Fe-NTA= ferric nitrilotriacetate; s.c.= sub-cutaneous; i.v.= intravenous

Table 3.

Tumor Metastasis Studies

METHODS RESULTS
Study Reference Rodent Model Tumor Type /
Induction model
Exercise Protocol Tumor Metastasis Results
Exercise Modality Exercise Prescription
Freq/week | Dur | Intens | Length
Exercise Initiation
Naturally occurring Gershbein, 1974 53 Holtzman rats Carcinosarcoma /
Walker-256 tumor
i.m. into both
hindlimbs.
Forced swimming 10×|15min|--|10d Immediately after injection. -Older EX rats ↑ lower abdominal and inguinal secondary tumor nodules.
Yan, 2011 64 3w old male
C57BL/6 mice
Lung / 2.5×105/50
µl/mouse Lewis lung
carcinoma cells s.c.
lower dorsal region
Voluntary wheel
running
Tumors excised at 1cm diameter,
access to wheels continued for
additional 2w.
9w before tumor implantation -Trend to inverse relationship between running distance and metastatic tumor yield
Jones, 2012 55 6–8w old male
C57BL/6 mice
Prostate / 5×105
mouse prostate C-1
cells, orthotopically
Voluntary wheel
running
Wheel running for 8 weeks 14d after tumor transplant -EX ↓ tumor nodal involvement by 36%, metastases weight by 88% and number of metastases by
34% (None were significant)
Artificial (intravenous injection of tumor cells) Uhlenbruck,
1991
63 BALB/c mice Sarcoma / 2.4×104
L-1 cells i.v.
Forced treadmill
running
7×|predetermined distances of
200–850m|0.3–0.5m/s|4–6w
4w before injection, followed by
either rest or an additional 2w
running
-EX decreased lung tumor multiplicity
-Differences in running prescriptions make it difficult to determine if exercise cessation after tumor
cell transplant was different from continuous exercise
MacNeil, 1993a 74 4–5w old male
C3H/He mice
Transformed
fibroblasts / 1.5×106
CIRAS 1 tumor cells
i.v. via tail vein.
Forced treadmill and
voluntary wheel
running
Treadmill: 5×|30min|15m/min,
0%|9w.
Voluntary Wheel: 24h access to
wheel for 9w.
All animals remained SED after
injection for 3w.
9w before injection -No significant effect of activity on lung tumor retention.
-Significant but weak correlation of distance run and multiplicity of lung metastases.
-Median number of lung metastases per animal was greater in EX mice (21 vs 10 SED control).
MacNeil, 1993b 73 C3H/He mice Transformed
fibroblasts / 3×105
CIRAS1 cells i.v.
Voluntary wheel
running
3–12w. 9w prior to and/or 3w after
injection
-EX had no effect on lung tumor density
-EX prior to tumor injection ↑ incidence in the lowest tertile of tumor distribution vs SED controls.
Hoffmann-
Goetz, 1994a
71 C3H/He-bg2J/+
and C3H/HeJ
mice
Transformed
fibroblasts / 5×105
CIRAS 1 or CIRAS
3 radiolabeled
transformed
fibroblast cells i.v.
via tail vein.
Voluntary wheel
running
24h access to running wheel for
9w.
9w before injection -EX ↓ retention of radioactivity in lungs 5min and 30min post-injection.
Hoffman-
Goetz, 1994b
70 3w old female
BALB/c mice
Mammary / Lateral
tail vein injection of
1×104 MMT 66 cells
Forced treadmill
running or voluntary
wheel running
Treadmill running:
5×|30min|18m/min at 0%| 8w or
3w.
Groups:
WS: wheel mice, 24h access for
8w, then SED for 3w.
TS: treadmill mice, then SED for
3w.
TT/WT: Continuous EX for 11w
total.
ST or SW: SED for 8w then
treadmill (ST) or wheel (SW) for
3w.
SS: SED for 11w total.
8w prior or 36h after injection for
3w
-No EX effect on number of lung tumors.
-TT tended to have ↑ tumor multiplicity.
-ST and SW tended to have ↓ tumor multiplicity.
Jadeski, 1996 72 4–9w old
C3H/He-bg2J/+
and C3H/HeJ
mice
Transformed
fibroblasts / 5×105
CIRAS 1 or CIRAS
3 transformed
fibroblast cells i.v.
via tail vein.
Forced treadmill
running
5×|30min|20m/min, 0%|9w

Acclimatization period of 1 week.
9w before injection -EX ↓ tumor cell lung retention (44.2 vs 52.8% control).
-EX ↓ lung retention of the less aggressive CIRAS 1, no difference in CIRAS 3 cells.
-EX did not alter final tumor lung metastases numbers, (subgroup evaluated 3w after EX and
injection).
Yan, 2011 64 3w old male
C57BL/6 mice
Melanoma /
0.75×105/200
µl/mouse B16BL/6
cells in lateral tail
vein
Voluntary wheel
running
Melanoma: 24h access to wheels,
continued 2w post-implantation.
9w before tumor implantation -No difference in number of lung metastasis
Higgins, 2014 75 6w old male
scid-beige mice
Lung / 5×105 A549-
luc-C8 cells i.v. via
tail vein
Voluntary wheel
running
28d
Mice averaged 600–1200 m/day
After lung tumors were detectable - EX decreased tumor volume, as measured by bioluminescent imaging

EX= exercise or activity groups; SED= Sedentary controls; i.v.= intravenous

Effect of exercise on tumor outcomes

In incidence studies, 58% reported that exercise inhibited tumor initiation or multiplicity (2325, 27, 28, 31, 33, 35, 36, 41, 4346), 8% reported that exercise accelerated tumor incidence (26, 42), and 3% found null effects (29, 30, 32, 34, 3740). In the growth category, exercise was associated with tumor inhibition in 64% of studies (24, 39, 4754, 5863, 6569), while 21% (37, 40, 44, 5557, 64) and 9% (26, 34, 42) of studies reported null and accelerated tumor growth, respectively. Finally, in the tumor metastasis category, three studies utilized models in which metastases arose from a primary tumor; of these, two reported non-significant inhibition of tumor growth (55, 64), while the other found accelerated tumor growth with exercise (53). Eight studies utilized intravenously injected tumor cell model of metastases. MacNeil and Hoffman-Goetz found that exercise did not alter retention of lung tumor cells, but increased the median number of lung metastases (74). Conversely, Jadeski and Hoffman-Goetz reported that exercise decreased retention of lung tumor cells, but had no effect on the number of lung metastases (72). One additional paper reported no difference in tumor cell retention in the lungs (71), whereas three papers reported no effect of exercise on lung metastasis (64, 70, 73). Two papers reported that exercise inhibited lung metastasis multiplicity (63) or total volume (75).

Effects on the intratumoral microenvironment

Mechanistic findings are summarized in Table 4 and Figure 1. Studies were classified as prevention (n=20; 37.7%), progression (n=26; 49.1%) or metastasis (n=7; 13.2%). Eight prevention studies (40%) reported significant effects of exercise in modulation of local immune response, tumor metabolism and tumor physiology/angiogenesis. Ten progression studies (38.5%) examined mechanisms underlying the effects of exercise on tumor progression (32, 41, 42, 44, 52, 55, 57, 61, 68, 69). The mechanisms examined included multiple different factors/pathways such as apoptosis, perfusion and immune cell infiltration. Only one metastasis study examined changes in tumor biology: Higgins et al. found that exercise favored a pro-apoptotic environment (75), reflecting changes in immune cell populations/function, tumor physiology and signaling cascades.

Table 4.

Mechanistic results

Study Reference Tumor type Exercise Modality Mechanistic findings
Intratumoral Systemic Potential Implications
Prevention Ikuyama, 1993 28 Carcinogen induced hepatoma Voluntary wheel
running
-None reported. -EX ↓ gamma-glutamyl transpeptidase and ↑ ALP. Reduced liver damage
Alessio, 2009 24 Spontaneous tumors Voluntary wheel
running
-None reported. -Mean serum prolactin levels were ↓ in exercising rats and ↑
in SED rats at 24 and 52w of age.
Prolactin has been associated with tumor growth
Goh, 2014 66 Orthotopic mammary tumor Voluntary wheel
running
-Inverse correlation between distance run and mitotic
index within tumors
-EX increased VO2 and respiratory exchange rate Decreased tumor cell proliferation
Betof, 2015 65 Orthotopic mammary tumor Voluntary wheel
running
-EX increased colocalization of desmin and CD31 and
decreased tumor hypoxia and necrosis
-EX increased expression of Fas, caspase 8 and cleaved
caspase 3
-None reported Increased microvessel density and maturity, which
leads to improved tumor perfusion; increased tumor
cell apoptosis
Woods, 1994 40 s.c. mammary tumor transplant Forced treadmill
running
-Moderate EX: ↑ phagocytic cells
-Exhaustive EX: ↓ total and highly phagocytic cells
-None reported Increased tumoricidal immune response
Bacurau, 2000 48 s.c. carcinoma transplant Forced treadmill
running
-None reported -EX ↓ glucose consumption and production, and lactate
production and ↑ glutamine consumption of macrophages
-EX ↓ H2O2 production by macrophages, and ↑
phagocytosis.
-EX ↑ lymphocyte proliferation
Increased tumoricidal immune response
Bacurau, 2007 49 s.c. carcinoma transplant Forced treadmill
running
-EX impairs tumor cell glucose and glutamine
metabolism.
-EX, non-tumor ↑ plasma corticosterone vs. control.
-EX prevents tumor-induced reduction in body weight and
food intake, activation of glutamine metabolism in
macrophages and lymphocytes.
Modulation of physiology
Lira, 2008 58 s.c. carcinoma transplant Forced treadmill
running
-None reported. -EX ↓ liver triacylglycerol (TAG) content compared to
SED.
-SED tumor animals had ↑ serum and liver TAG, and ↓ rate
of VLDL secretion, apoB expression and microsomal TAG
transfer protein compared to control.
Associated with reduced cachexia
Murphy, 2011 59 Transgenic mammary tumors Forced treadmill
running
-None reported. -↓ spleen weight compared to wild-type mice.
-No difference in spleen weight due to EX.
-EX ↓ plasma IL-6 and MP-1.
Increased tumoricidal immune response
Kato, 2011 29 Carcinogen-induced renal tumors Forced treadmill
running
-None reported. -Long-term EX and Fe-NTA ↓ renal brown droplets
compared to SED, ↑ adrenal weight compared to both other
groups.
Brown droplets could reflect renal damage caused by
carcinogen applied. Increases in adrenal weight have
been linked to psychological stress.
Shalamzari, 2014 67 s.c. mammary tumor transplant Forced treadmill
running
- EX after tumor transplant decreased tumor IL-6
and VEGF
- IL-6 and VEGF levels in ETR mice were not different
from RTR (sedentary) mice.
-None reported Reduced inflammation and subsequent angiogenesis
Piguet, 2015 46 Transgenic hepatocellular
carcinoma
Forced treadmill
running
- EX decreased proliferation (Ki67 staining) in tumor
nodules >15mm3.
- EX altered gene expression of fatty acid metabolism
pathways
Altered lipogenesis. Some lipogenesis pathways are
prognostic indicators following HCC surgical
removal
Malicka, 2015 45 Carcinogen-induced mammary
tumor
Forced treadmill
running
-EX increased TUNEL positive cells - None reported
Almeida, 2009 47 s.c. carcinoma transplant Swimming -50% workload ↓ macrophage infiltration and neutrophil accumulation. -80% workload induced cardiac hypertrophy vs. 50%. Altered immune response; future analysis of
macrophage subsets would be beneficial to the field
Sasvari, 2011 62 s.c. sarcoma transplant Swimming -None reported. -EX ↓ oxidative modification of protein in the liver as
measured by protein carbonyls.
Reduced oxidative stress or improved anti-oxidant
activity
Progression Daneryd, 1995 51 Carcinogen-induced or s.c. leydig
cell transplant
Voluntary wheel
running
-None reported - EX normalized insulin sensitivity compared to SED
- EX ↑ skeletal muscle metabolism
- EX attenuated ↓ of reverse triiodothyronine secretion by
the thyroid
- EX ↑ insulin:glucagon ratio
- EX ↓ corticosterone
Reflects metabolic adaptations to prevent hypo- or
hyperglycemia, which may develop in cancer
patients.
Daneryd, 1995 52 s.c. leydig cell transplant Voluntary wheel
running
-EX 1.31-fold ↑ in tumor cell energy charge and uric acid
content
-None reported. Uric acid has since been shown to stimulate dendritic
cell activation, and is elevated in tumors with anti-
tumor immune responses
Zhu, 2008 42 Carcinogen-induced mammary
tumor
Voluntary wheel
running
-EX activated AMPK
-EX ↓ VEGF
-EX ↓ Bcl-2, X-linked inhibitor of apoptosis pathway
(XIAP) while ↑ Bax, apoptosis peptidase-activating
factor-1
↓ Insulin, IGF-1, CRP, leptin and estradiol
↑ Corticosterone
EX increases cell metabolism and skews the BCL-2
family member protein profile pro-apoptotic.
Jones, 2010 57 Orthotopic mammary tumor Voluntary wheel
running
-EX ↑ perfused vessels and HIF-1 protein levels -None reported Improved tumor perfusion, oxygenation
Yan, 2011 64 s.c. lung tumor transplant Voluntary wheel
running
-None reported. -T umor presence ↑ plasma VEGF, PDGF-AB, MCP-1.
-Running: ↓ plasma insulin and leptin, ↑ adiponectin, ↑
plasma VEGF.
VEGF expression has been linked to worse outcome
in patients, but could instead represent improved
microvessel density and vascular normalization in
tumors.
Jones, 2012 55 Orthotopic prostate tumor Voluntary wheel
running
-EX ↑vascularization, stabilization of HIF-1a → 40%
↑regulation of VEGF
-Expression of prometastatic genes was significantly
modulated in exercising animals with a shift toward
reduced metastasis
-↑ activity of protein kinases within MEK/MAPK and
PI3/mTOR
Improved tumor perfusion/oxygenation
Mann, 2010 31 Carcinogen-induced mammary
tumor
Voluntary non-
motorized or
motorized wheel
running
-None reported -Induced citrate synthase activity Reflects training response
Zhu, 2012 43 Carcinogen-induced mammary
tumor
Voluntary wheel
running at a fixed daily
distance
-None reported -↓ Insulin-like growth factor-1 (IGF-1), insulin, interleukin-
6, serum amyloid protein, TNF-α, and leptin
-↑ IGF-binding protein 3 (IGFBP-3) and adiponectin
Increased tumoricidal immune response
Westerlind, 2003 37 Carcinogen-induced mammary
tumor
Forced treadmill
running
-None reported - ↑heart and soleus weights Reflects training response
Zielinski, 2004 44 s.c. neoplastic lymphoid cell
transplant
Forced treadmill
running
-No difference in the fluid content of tumor
-EX ↓ in vessel density
-EX ↓ inflammatory cells (macrophages and neutrophils)
but increased lymphocytes
-None reported Altered immune response; future analysis of
macrophage subsets would be beneficial to the field
Zhu, 2009 41 Carcinogen-induced mammary
tumor
Forced wheel running -Apoptosis induced via mitochondrial pathway in EX
-Cell cycle progression suppressed at G(1)/S transition in
EX
-EX ↓ blood vessel density.
-None reported EX promotes tumor cell apoptosis and prevents
proliferations and angiogenesis
Gueritat 2014 69 s.c. prostate tumor transplant Forced treadmill
running
- EX decreased tumor cell proliferation (Ki67 staining), p-
ERK/ERK ratio and 8-OHdG
- No difference in tumor SOD, protein carbonylation or
lipid oxidattion
-None reported ERK phosphorylation is increased by oxidative stress
Aveseh, 2015 68 Orthotopic mammary tumor Forced treadmill
running
- EX shifted tumor lactate dehydrogenase expression
towards the LDH1 isoenzyme
- EX decreased tumor monocarboxylate transporter 1
(MCT1) and CD147 expression.
-None reported A shift towards LDH-1 may signal reduced lactate
concentrations in the tumor. Lack of MCT1 may
result in the tumor cells utilizing glucose instead of
lactose, and eventually starving the tumor cells.
CD147 is required for MCT1 expression
Radak, 2002 61 s.c. solid leukemia transplant Swimming -EC tumors had ↑ Ras protein compared to control.
-EC tumors had ↑ I-kappaB protein than ET and control.
-None reported May be associated with lymphocyte proliferation and
activity, but additional analyses are needed
Abdalla, 2013 23 Carcinogen-induced mammary
tumor
Swimming -None reported. -Physical activity ↑ lymphocytes producing IFN-γ, IL-2, IL-
12, and TNF-α.
-Physical activity promoted immune system polarization
toward an antitumor Th1 response pattern profile.
Increased tumoricidal immune response
Metastasis MacNeil, 1993a 74 i.v. injection of transformed
fibroblasts
Forced treadmill or
voluntary wheel
running
-None reported. -Wheel group ↑ splenic NK response vs control. Increased tumoricidal immune response
Hoffmann-Goetz,
1994a
71 i.v. injection of transformed
fibroblasts
Voluntary wheel
running
-None reported. -Running ↓recovery of radioactivity from liver, spleen and
kidney at 30min and 90min post-injection.
May reflect decreased retention of circulating tumor
cells in organs, thereby lowering the risk of
metastasis.
Hoffman-Goetz,
1994b
70 i.v. injection of mammary tumor
cells
Forced treadmill
running or voluntary
wheel running
-None reported -Pre-tumor EX ↑ LAK cell activity.
-NK activity lower in animals that stopped EX at tumor
injection.
Increased tumoricidal immune response
Jadeski, 1996 72 i.v. injection of transformed
fibroblasts
Forced treadmill
running
-None reported -EX ↑ citrate synthase activity in the soleus. Indicates a training effect developed
Higgins, 2014 75 i.v. injection of lung tumor cells Voluntary wheel
running
-EX tumors had increased levels of p53, Bax and Bak.
-EX tumors had increased staining for cleaved caspase 3
-EX increased serum BUN and decreased ALT, but both
were still within normal ranges
Supports increased apoptosis of tumor cells

Figure 1.

Figure 1

Postulated intratumoral and systemic mechanisms underlying exercise oncology across the cancer continuum. Note that few reports combined measurements in tumor with systemic changes. The link between intratumoral changes and systemic changes is largely unknown.

Effects on systemic (host) pathways

Correlative systemic pathways examined are summarized in Table 4. Ten prevention (24, 28, 29, 40, 4649, 59, 62), seven progression (23, 31, 37, 42, 51, 55, 64, 76) and five metastasis studies (7072, 74, 75) reported significant effects of exercise on changes in systemic effectors, predominantly changes in factors involved in immune surveillance or metabolism.

DISCUSSION

The findings of this critical review indicate that the current evidence base is plagued by heterogeneity in all aspects of study methodology and data reporting. Unfortunately, such heterogeneity precludes rigorous evaluation of essential questions such as the biologically effective exercise dose to modulate specific tumor pathways or inhibit tumor growth, the effects of manipulating exercise intensity or duration or the differential impact of exercise across tumor subtypes (22). Nevertheless, our review did permit identification of the most salient methodological issues that prudent investigators may consider when designing in-vivo pre-clinical exercise-oncology studies. These aspects are described in the proceeding sections and summarized in Table 5.

Table 5.

Recommendations for Preclinical Exercise Oncology Research

Concern Recommendation
Use of xenograft models in immune deficit
animals
Orthotopic implantation of syngeneic tumor cell lines or
induction of orthotopic tumors via transgenic or
chemical methods in immune competent animals
Poor description of exercise intervention
characteristics
Describe frequency, intensity, duration, and progression,
as appropriate. Avoid vague terms such as “exercise to
exhaustion”. Confirmation of ‘training’ effect via
muscle fiber or mitochondrial function analysis
Handling of control (sedentary) animals Handling, social interaction, and environment should be
similar to animals randomized to exercise conditions.
This includes differences in cage size and social
housing. If possible, animals should be acclimated to
exercise, or introduced to the activity gradually.
Tail vein models of metastasis Consider using orthotopic implantation of syngeneic
tumor cell lines or transgenic models that spontaneously
develop metastasis. However, tail vein models of
metastasis may still be useful for assessing the effects of
exercise at later time points in the metastatic cascade.
Lack of assessment of systemic and molecular
mechanisms
Investigate effects on systemic mechanisms (metabolic
and sex hormones, inflammation, immunity, and
products of oxidation) postulated to underlie effects of
exercise on tumorigenesis as well as potential mediating
molecular mechanisms (e.g., cell signaling pathways,
angiogenesis, metabolism, migration). Findings should
be validated by the use of knock-out/knock-in transgenic
animals.
Lack of assessment of tumor biology beyond
tumor incidence, weight, or volume
Report on other common markers of the neoplastic
phenotype (e.g., apoptosis, proliferation, microvessel
density, necrosis, angiogenesis)
Lack of concern regarding the psychological
differences between voluntary physical
activity vs. forced exercise
Comprehensive study on hypothalamic- pituitary-
adrenal axis activation in response to different exercise
prescriptions and the effects that associated hormones
have on tumor progression/prevention in sedentary
controls.

Heterogeneity in Exercise Prescription

The components of the exercise prescription being investigated in preclinical studies should mirror, as closely as possible, exercise prescription parameters tested in human studies (22). Key parameters include: (1) modality/paradigm (i.e., forced vs. voluntary), (2) dose (i.e., intensity, session duration, frequency of training sessions/week, and length of treatment), and (3) schedule (i.e., time of initiation).

Exercise Modality (Forced vs. Voluntary Paradigms)

A foremost decision is the use of forced versus voluntary exercise paradigms. An often overlooked key point is that voluntary wheel running is a model of physical activity whereas forced paradigms are a model of exercise training (i.e., structured and purposeful physical activity). Observational (epidemiological) studies typically measure both physical activity and exercise, whereas efficacy-based clinical trials largely examine highly structured exercise training. The decision of which exercise modality is selected should depend on whether the investigators envisage the study findings directly informing clinical translation or plausibility/mechanisms of a clinical observation.

An important caveat to consider in all exercise paradigms is the degree of associated negative stress, as evidenced by exercise-induced increases in serum corticosterone and fecal corticosterone metabolites (77, 78), as observed in both voluntary and forced exercise paradigms. Furthermore, voluntary running may result in “addictive behavior” with negative effects on the hypothalamic-pituitary-adrenal (HPA) axis and animal health (79). Human studies have taught us that exercise induces spikes in cortisol at the beginning of exercise and immediately after bout of exercise ceases (80). Stress-induced activation of the HPA axis may accelerate tumor growth (81), and have diverse effects on the immune response (reviewed in (82)), thereby confounding the efficacy of exercise on tumor growth characteristics. HPA-mediated changes on the immune response could contribute to discordance between studies with respect to immune function. For example Zhu et al. reported a decrease in TNF-α (43) whereas Abdalla et al. reported an increase (23).

Though changes in glucocorticoids have been seen using all exercise modes and intensities, such considerations may be especially important when investigating the effects of high-intensity or high-volume exercise treatment doses. This is relevant because, as reviewed here, studies have tested the effects of exhaustive exercise doses (40) (e.g., forced swimming for four hours followed by 12 hours of physical activity (54)), or forced swimming of animals loaded with external weights (30, 32, 47). The latter exercise paradigms may have additional safety and ethical concerns. Indeed, one study investigating the effect of forced swimming reported four animal deaths due to drowning (30). Researchers should also be cognizant of the impact of the exercise on the animals’ natural circadian rhythms. Fuss et al. demonstrated that voluntary wheel running occurs predominantly during the animals’ dark cycle (78), therefore it would be prudent to conduct exercise training during the dark cycle. Regardless of exercise prescription, corticosterone analysis would be helpful to include in all study designs; such levels could be correlated with tumor growth end points. In this review, three studies analyzed systemic corticosterone levels (42, 48, 51); of these, two studies reported increases in corticosterone levels whereas the other study reported a decrease. Interestingly, tumor growth was inhibited in all studies.

Exercise Dose (Intensity, Session Duration, Frequency of Training, and Length of Treatment)

Exercise at different intensities confers remarkably different physiological and gene expression adaptations in mammals (83), however a key question is whether these differences translate into differential effects on tumor outcomes. In clinical trials, the efficacy of different exercise intensities varying from 50% to 100% of a patient-derived physiological parameter (e.g., age-predicted maximum heart rate, measured exercise capacity) have been investigated. Such an approach permits personalized exercise prescriptions, which is important because exercise abilities (and therefore appropriate exercise intensity) vary considerably in patients with cancer (83). Animal-specific exercise prescriptions are challenging to implement in pre-clinical studies, but could be important, as findings reviewed here suggest that exercise intensity may differentially modulate tumor end points. For example, using a transgenic model of p53+/− MMTV-Wnt-1, Colbert et al. found that exposure to forced treadmill training of different intensities accelerated tumor growth rate compared to sedentary controls, with the highest tumor multiplicity in the lowest intensity group (26). Conversely, Almeida et al. reported diminishing returns in the context of increasing exercise intensity using two different swimming intensities (50% or 80% of maximal workload; CT50 and CT80, respectively, defined using a maximum load test conducted one week prior to exercise initiation) (47). CT50 caused significant tumor growth inhibition whereas CT80 was ineffective. Using a model of forced treadmill running, Woods et al. compared the effects of “moderate” vs “exhaustive” exercise prescriptions, with no differences in mammary tumor growth or overall incidence (40). In contrast, Malicka et al. reported a trend towards a negative correlation between exercise intensity (varied by progressively adjusting the speed of the treadmill [low: 0.48–1.34 km/h, moderate: 0.6–1.68 km/h, high: 0.72–2.0 km/h]) and tumor incidence and multiplicity, as well as an increase in tumor cell apoptosis across all exercise prescriptions, compared to controls (45). Kato et al. examined duration of prescription of forced treadmill running using a nitrilotriacetate-induced renal carcinoma model. Rats were assigned to exercise exposure for either 12 weeks or 40 weeks (29). Twelve weeks of exercise increased microcarcinoma incidence and multiplicity compared to sedentary controls with no differences (compared to control) in 40 weeks of exercise.

Schedule of Exercise Exposure

A critical question is whether exercise exposure prior to diagnosis improves disease outcomes compared to inactivity (prior to diagnosis). Similarly, do previously sedentary patients initiating exercise after diagnosis have superior outcomes compared to those remaining sedentary or decreasing exercise levels after diagnosis? Exploratory findings from epidemiological studies suggest that timing or initiation of exercise exposure could be important (84). Two studies have empirically investigated this question: Betof et al. and Shalamzari et al. found that exercise initiated after tumor transplantation (equivalent to post-diagnosis setting) inhibited tumor growth, independent of exposure to exercise prior to transplantation (65, 67). Similarly, Radak et al. found slower tumor growth with exercise pre- as well as post-tumor implantation in comparison with no exercise after transplant or sedentary control (61). Such findings stress the importance of maintaining exercise post-diagnosis. Finally, whether the effects of exercise are different across the steps in the metastatic cascade (altered adhesion [invasion]; intravasation; survival in the circulation; extravasation, and seeding at a distant site [metastatic colonization] (85)) has not been investigated. Addressing this question has significant clinical implications, and further research in this area is warranted.

Common Methodological/Experimental Weaknesses

Several salient methodological issues across studies were identified (see Table 5). Of these, two common issues that require particular attention regarded selection of appropriate in vivo tumor models, and lack of mechanistic studies.

In Vivo Tumor Models

Preclinical oncology studies have generally focused on subcutaneous tumor implantation models. These models permit monitoring of tumor growth kinetics but do not accurately recapitulate the tissue microenvironment of the ectopic (orthotopic) or distant organ or mimic the natural evolution of cancer. In addition, blood flow to subcutaneous tumors may not reflect that of orthotopic tumors. As the benefits/risks of using subcutaneous models are beyond the scope of this review, here we will simply caution investigators against using subcutaneous tumors. This model is not a perfect surrogate for spontaneously occurring tumors.

The majority of studies reviewed herein investigated the effects of exercise on tumor growth characteristics in the primary organ site. In contrast, studies of metastasis, the primary cause of cancer-related death, has received limited attention to date. Additionally, 73% of the metastasis studies we reviewed used an intravenous (tail-vein) injection of tumor cells as a model of metastasis which evaluates the ability of tumor cells to survive in circulation and colonize an organ but does not enable investigation of the early steps in the metastatic cascade.(86) Indeed, three studies reported on tumor cell retention in the lungs following intravenous injection (71, 72, 74), with two studies reporting a decrease in tumor cell retention in exercising animals, but no change in the final number of metastases. Interestingly, MacNeil et al. reported that exercise did not affect tumor cell retention in the lung. These three studies differed with respect to type of exercise and number of tumor cells injected; thus no meaningful comparisons can be made between them. The impact of the methodological differences can only be evaluated by completing the studies-side-by-side, comparing all combinations of exercise/tumor inoculation models.

An alternative, and arguably more appropriate model of metastasis, is surgical excision of the primary tumor which stimulates spontaneous metastasis, predominantly to the lungs; Only one study to date (Yan et al.) has adopted this model to examine exercise effects (64); they reported a trend towards an inverse relationship between distance run and metastatic burden. Conversely, Jones et al. examined the metastasis response in a prostate cancer model by quantifying the mass of “non-contiguous external masses that were grossly visible independent from the primary prostate tissue.” In this model, exercise was not associated with a significant reduction in the number or weight of metastases (55).

Emerging technology has provided researchers with a considerable number of in vivo models beyond cell line xenografts, such as patient-derived xenografts (PDXs), syngeneic allografts, and genetically-engineered mouse models (GEMMs) (87), as well as non-rodent models (e.g., zebrafish, Drosophila). The advantages and disadvantages of each model have been reviewed elsewhere (8891). Ultimately, no one in vivo model will be appropriate to address all exercise-oncology questions, with model selection being contingent on the scientific question at hand as well as translational importance.

The Importance of Appropriate Control Groups

Consideration of the nature of control (non-exercising) groups should not be overlooked. To the extent possible, control animals should be exposed to the same variables as exercise counterparts including aspects related to housing, transportation to different facilities, or procedures to reinforce exercise behavior (i.e., prodding, shock). Taken even further, animals could be housed in cages with locked wheels, placed on stationary treadmills, or made to stand in very shallow pools of water, depending upon the exercise regimens used. We stress that exposing control and experimental mice to different housing or environmental conditions is a study weakness. For example, one study housed control animals in unusually small cages (5 inches in diameter and 6 inches high) to restrict activity (54), whereas exercise treatment animals were not confined. The differences in housing size and daily movement could have either induced a stress response or altered the animals’ resting metabolism, thereby affecting tumor growth. We advise researchers reviewing extant publications for planning of their own exercise oncology studies to consider whether controls were properly handled before modeling their own work off of previous studies.

Mechanistic Studies/Analyses

The majority of studies reviewed here examined the effects of exercise on tumor growth characteristics as evaluated by tumor volume or growth rates. While such end points are clearly important, subtle but important modulations of intratumoral physiological or biological alterations can be masked. For instance, our group observed differences in perfusion and expression of key factors regulating metabolism and hypoxia, despite comparable primary tumor growth rates between exercised and sedentary animals (57). Elegant studies by McCollough et al. demonstrated that exercise improved blood flow and oxygen delivery to orthotopic prostate tumors in rats, but not to the normal prostate tissue in either tumor-bearing or control rats (92). These changes were reflected by decreased hypoxia within the tumor during exercise. While in-depth explications of the systemic or local molecular mechanisms underpinning the exercise-tumor prevention/progression relationship remains in its infancy, studies such as this one may set the precedent for future mechanistic studies in this field. The current working hypothesis is that exercise modulates tumor progression via modulation of the host -tumor interaction (19). Tumor progression is regulated by complex, multifaceted interactions between the systemic milieu (host), tumor microenvironment, and cancer cells (93). The microenvironments of primary and metastatic tumors are subject to modulation by systemic and local growth/angiogenic factors, cytokines, hormones, and resident cells (94, 95). Factors such as hepatocyte growth factor (HGF), tumor necrosis factor (TNF), interleukin (IL)-6, insulin, and leptin (9698) have already been associated with higher risk of recurrence and cancer-specific mortality in a number of solid malignancies.(99) Clearly, manipulation of such factors by physical activity could alter aspects of the cancer continuum (100).

As reviewed here, multiple systemic factors are perturbed by exercise, including metabolism, inflammatory-immune, and reactive oxygen species-mediated pathways (101). The breadth of these factors likely contributes to the pleiotropic benefits of exercise in health and disease (102, 103) and likely the potential antitumor effects of exercise. (19) Notably, most cancer studies investigate single pathways in isolation, without consideration of overlap/synergism between pathways. Because the host/tumor interaction is modulated by numerous host-related factors and multiple pathways (100), an ideal study approach would be to investigate exercise effects on multiple pathways simultaneously. This would fill in the missing gaps of the “multi-targeted” effects of exercise. A recent analysis of pre-clinical exercise oncology studies by Pedersen et al. reported that only 30.7% evaluated systemic changes in animals (104). In addition, intratumoral signaling, and changes in tumor vascularity were examined by only 29.5% and 6.8%, respectively (104).

Future Recommendations

With a view towards future studies, we encourage the exercise-oncology field to consider certain guidelines for preclinical exercise-oncology research (see Table 5). However, we realize that it is impractical for all exercise-oncology experiments to standardize all study procedures. Nevertheless, it may behoove the field to develop a means of quantifying the exercise prescription applied, which will then permit comparisons between studies. Such an approach is readily used in the fields of radiation oncology (i.e., the biologically equivalent dose [BED], which allows comparison of different dose/fractionation schemes (105)) and hyperthermia (i.e., the cumulative equivalent minutes at 43°C [CEM43], which standardizes the thermal killing effect of hyperthermia, regardless of variations in heating efficiencies between tumors (106)). Establishing a biological equivalent exercise dose (BEED) would facilitate comparisons across studies as well as provide an opportunity to test elements of prescriptions such as different schedules, timing, and duration.

Exercise investigations should also strive to adopt the experimental procedures and models being utilized in the general tumor biology literature. Indeed, it appears that more recent studies reviewed here have started to utilize clinically-relevant tumor models, favoring transgenic mice and orthotopic tumors over carcinogen-induction. The general cancer field is also starting to favor the use of patient-derived xenograft (PDX) models. PDX have certain weaknesses, including increased heterogeneity, and a requirement for immunocompromised mice, but could represent an important step towards personalized medicine. Furthermore, PDXs do not accrue additional mutations through in vitro culture and tend to be slower growing than murine-derived tumor lines. Delayed growth curves may highlight slight changes in tumor growth following manipulations of exercise dose. GEM models should also be considered, especially in mechanistic studies. A second trend in cancer biology is use of biomarkers. The discovery of blood, imaging, and/or genomic biomarker(s) to predict or monitor exercise response is of obvious importance.

Finally, researchers must be cognizant of clinical care, and design studies that reflect the clinical scenario. For example, the vast majority of the current literature investigates the effects of exercise as monotherapy. The majority of clinical scenarios would be applying exercise as an adjuvant therapy to surgery, radiation, chemotherapy, or immunotherapies. As such, it is important for the next generation of preclinical studies aiming to study tumor progression to evaluate the interaction between exercise and the pharmacodynamic or pharmacokinetic activity of conventional and novel therapies to guide the design and interpretation of clinical studies. We advise researchers to proceed with caution and carefully include all possible controls groups, because the additional therapies could introduce new confounding factors that complicate data interpretations Conversely, studies on tumor incidence may be strengthened by eliminating additional factors such as concomitant manipulation of dietary composition.

Conclusion

A sound foundation of basic and translational studies will optimize the therapeutic potential of exercise on symptom control and clinical outcomes across the cancer continuum. Despite its importance, we found that the current evidence base is plagued by considerable methodological heterogeneity in all aspects of study design, end points, and efficacy thereby precluding meaningful comparisons and conclusions. To this end, we have provided an overview of methodological and data reporting standards that we hope will set the platform for the next generation of preclinical studies required for the continued development and progression of exercise-oncology research.

Supplementary Material

1
2
3

Figure 2.

Figure 2

Hypothesized pathways by which endurance exercise may impact tumor progression and metastasis. Key host tissues such as skeletal muscle, adipose tissue, bone marrow, and the liver mediate the effect of exercise on a variety of systemic pathways. Exercise-induced alterations in systemic and circulating factors, in turn, influences ligand availability in the tumor microenvironment which alters cellular signaling modulating the hallmarks of cancer.

Acknowledgments

LWJ is supported in part by grants from the National Cancer Institute, AKTIV Against Cancer, and the Memorial Sloan Kettering Cancer Center Support Grant/Core Grant (P30 CA008748). MWD is supported by NIH CA40355.

Footnotes

Conflict of Interest: The authors report no conflicts of interest

REFERENCES

  • 1.Booth FW, Gordon SE, Carlson CJ, Hamilton MT. Waging war on modern chronic diseases: primary prevention through exercise biology. Journal of applied physiology. 2000;88(2):774–787. doi: 10.1152/jappl.2000.88.2.774. [DOI] [PubMed] [Google Scholar]
  • 2.Giannuzzi P, Mezzani A, Saner H, Bjornstad H, Fioretti P, Mendes M, et al. Physical activity for primary and secondary prevention. Position paper of the Working Group on Cardiac Rehabilitation and Exercise Physiology of the European Society of Cardiology. European journal of cardiovascular prevention and rehabilitation : official journal of the European Society of Cardiology, Working Groups on Epidemiology & Prevention and Cardiac Rehabilitation and Exercise Physiology. 2003;10(5):319–327. doi: 10.1097/01.hjr.0000086303.28200.50. [DOI] [PubMed] [Google Scholar]
  • 3.Hawley JA, Hargreaves M, Joyner MJ, Zierath JR. Integrative biology of exercise. Cell. 2014;159(4):738–749. doi: 10.1016/j.cell.2014.10.029. [DOI] [PubMed] [Google Scholar]
  • 4.Warburton DE, Nicol CW, Bredin SS. Health benefits of physical activity: the evidence. CMAJ : Canadian Medical Association journal = journal de l'Association medicale canadienne. 2006;174(6):801–809. doi: 10.1503/cmaj.051351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Jones LW, Alfano CM. Exercise-oncology research: past, present, and future. Acta oncologica. 2013;52(2):195–215. doi: 10.3109/0284186X.2012.742564. [DOI] [PubMed] [Google Scholar]
  • 6.Friedenreich CM, Woolcott CG, McTiernan A, Ballard-Barbash R, Brant RF, Stanczyk FZ, et al. Alberta physical activity and breast cancer prevention trial: sex hormone changes in a year-long exercise intervention among postmenopausal women. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2010;28(9):1458–1466. doi: 10.1200/JCO.2009.24.9557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lynch BM, Friedenreich CM, Winkler EA, Healy GN, Vallance JK, Eakin EG, et al. Associations of objectively assessed physical activity and sedentary time with biomarkers of breast cancer risk in postmenopausal women: findings from NHANES (2003–2006) Breast cancer research and treatment. 2011;130(1):183–194. doi: 10.1007/s10549-011-1559-2. [DOI] [PubMed] [Google Scholar]
  • 8.Gammon MD, Schoenberg JB, Britton JA, Kelsey JL, Coates RJ, Brogan D, et al. Recreational physical activity and breast cancer risk among women under age 45 years. American journal of epidemiology. 1998;147(3):273–280. doi: 10.1093/oxfordjournals.aje.a009447. [DOI] [PubMed] [Google Scholar]
  • 9.Bernstein L, Henderson BE, Hanisch R, Sullivan-Halley J, Ross RK. Physical exercise and reduced risk of breast cancer in young women. Journal of the National Cancer Institute. 1994;86(18):1403–1408. doi: 10.1093/jnci/86.18.1403. [DOI] [PubMed] [Google Scholar]
  • 10.Byers T, Nestle M, McTiernan A, Doyle C, Currie-Williams A, Gansler T, et al. American Cancer Society guidelines on nutrition and physical activity for cancer prevention: Reducing the risk of cancer with healthy food choices and physical activity. CA: a cancer journal for clinicians. 2002;52(2):92–119. doi: 10.3322/canjclin.52.2.92. [DOI] [PubMed] [Google Scholar]
  • 11.Kushi LH, Doyle C, McCullough M, Rock CL, Demark-Wahnefried W, Bandera EV, et al. American Cancer Society Guidelines on nutrition and physical activity for cancer prevention: reducing the risk of cancer with healthy food choices and physical activity. CA: a cancer journal for clinicians. 2012;62(1):30–67. doi: 10.3322/caac.20140. [DOI] [PubMed] [Google Scholar]
  • 12.Friedenreich CM, Neilson HK, O'Reilly R, Duha A, Yasui Y, Morielli AR, et al. Effects of a High vs Moderate Volume of Aerobic Exercise on Adiposity Outcomes in Postmenopausal Women: A Randomized Clinical Trial. JAMA oncology. 2015 doi: 10.1001/jamaoncol.2015.2239. [DOI] [PubMed] [Google Scholar]
  • 13.Irwin ML, Yasui Y, Ulrich CM, Bowen D, Rudolph RE, Schwartz RS, et al. Effect of exercise on total and intra-abdominal body fat in postmenopausal women: a randomized controlled trial. JAMA : the journal of the American Medical Association. 2003;289(3):323–330. doi: 10.1001/jama.289.3.323. [DOI] [PubMed] [Google Scholar]
  • 14.McTiernan A, Ulrich CM, Yancey D, Slate S, Nakamura H, Oestreicher N, et al. The Physical Activity for Total Health (PATH) Study: rationale and design. Medicine and science in sports and exercise. 1999;31(9):1307–1312. doi: 10.1097/00005768-199909000-00012. [DOI] [PubMed] [Google Scholar]
  • 15.Campbell KL, McTiernan A, Li SS, Sorensen BE, Yasui Y, Lampe JW, et al. Effect of a 12-month exercise intervention on the apoptotic regulating proteins Bax and Bcl-2 in colon crypts: a randomized controlled trial. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2007;16(9):1767–1774. doi: 10.1158/1055-9965.EPI-07-0291. [DOI] [PubMed] [Google Scholar]
  • 16.Mishra SI, Scherer RW, Geigle PM, Berlanstein DR, Topaloglu O, Gotay CC, et al. Exercise interventions on health-related quality of life for cancer survivors. The Cochrane database of systematic reviews. 2012;8:CD007566. doi: 10.1002/14651858.CD007566.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Schmitz KH, Holtzman J, Courneya KS, Masse LC, Duval S, Kane R. Controlled physical activity trials in cancer survivors: a systematic review and meta-analysis. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2005;14(7):1588–1595. doi: 10.1158/1055-9965.EPI-04-0703. [DOI] [PubMed] [Google Scholar]
  • 18.Jones LW, Dewhirst MW. Therapeutic properties of aerobic training after a cancer diagnosis: more than a one-trick pony? Journal of the National Cancer Institute. 2014;106(4) doi: 10.1093/jnci/dju042. dju042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Betof AS, Dewhirst MW, Jones LW. Effects and potential mechanisms of exercise training on cancer progression: a translational perspective. Brain, behavior, and immunity. 2013;30(Suppl):S75–S87. doi: 10.1016/j.bbi.2012.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.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. Journal of the National Cancer Institute. 2012;104(11):815–840. doi: 10.1093/jnci/djs207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Gorski DH. Integrative oncology: really the best of both worlds? Nature reviews Cancer. 2014;14(10) doi: 10.1038/nrc3822. [DOI] [PubMed] [Google Scholar]
  • 22.Jones LW. Precision Oncology Framework for Investigation of Exercise as Treatment for Cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2015 doi: 10.1200/JCO.2015.62.7687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Abdalla DR, Murta EF, 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. European journal of cancer prevention : the official journal of the European Cancer Prevention Organisation. 2013;22(3):251–258. doi: 10.1097/CEJ.0b013e3283592cbb. [DOI] [PubMed] [Google Scholar]
  • 24.Alessio HM, Schweitzer NB, Snedden AM, Callahan P, Hagerman AE. Revisiting influences on tumor development focusing on laboratory housing. Journal of the American Association for Laboratory Animal Science : JAALAS. 2009;48(3):258–262. [PMC free article] [PubMed] [Google Scholar]
  • 25.Andrianopoulos G, Nelson RL, Bombeck CT, Souza G. The influence of physical activity in 1,2 dimethylhydrazine induced colon carcinogenesis in the rat. Anticancer research. 1987;7(4B):849–852. [PubMed] [Google Scholar]
  • 26.Colbert LH, Westerlind KC, Perkins SN, Haines DC, Berrigan D, Donehower LA, et al. Exercise effects on tumorigenesis in a p53-deficient mouse model of breast cancer. Medicine and science in sports and exercise. 2009;41(8):1597–1605. doi: 10.1249/MSS.0b013e31819f1f05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Esser KA, Harpole CE, Prins GS, Diamond AM. Physical activity reduces prostate carcinogenesis in a transgenic model. The Prostate. 2009;69(13):1372–1377. doi: 10.1002/pros.20987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ikuyama T, Watanabe T, Minegishi Y, Osanai H. Effect of voluntary exercise on 3'-methyl-4-dimethylaminoazobenzene-induced hepatomas in male Jc1:Wistar rats. Proceedings of the Society for Experimental Biology and Medicine Society for Experimental Biology and Medicine (New York, NY) 1993;204(2):211–215. doi: 10.3181/00379727-204-43655. [DOI] [PubMed] [Google Scholar]
  • 29.Kato T, Kawaguchi H, Miyoshi N, Aoyama K, Komatsu M, Horiuchi M, et al. Effect of habitual exercise on renal carcinogenesis by ferric nitrilotriacetate. Environmental health and preventive medicine. 2011;16(4):232–238. doi: 10.1007/s12199-010-0191-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Lunz W, Peluzio MC, Dias CM, Moreira AP, Natali AJ. Long-term aerobic swimming training by rats reduces the number of aberrant crypt foci in 1,2-dimethylhydrazine-induced colon cancer. Brazilian journal of medical and biological research = Revista brasileira de pesquisas medicas e biologicas / Sociedade Brasileira de Biofisica [et al] 2008;41(11):1000–1004. doi: 10.1590/s0100-879x2008001100009. [DOI] [PubMed] [Google Scholar]
  • 31.Mann PB, Jiang W, Zhu Z, Wolfe P, McTiernan A, Thompson HJ. Wheel running, skeletal muscle aerobic capacity and 1-methyl-1-nitrosourea induced mammary carcinogenesis in the rat. Carcinogenesis. 2010;31(7):1279–1283. doi: 10.1093/carcin/bgq063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Paceli RB, Cal RN, dos Santos CH, Cordeiro JA, Neiva CM, Nagamine KK, et al. The influence of physical activity in the progression of experimental lung cancer in mice. Pathology, research and practice. 2012;208(7):377–381. doi: 10.1016/j.prp.2012.04.006. [DOI] [PubMed] [Google Scholar]
  • 33.Reddy BS, Sugie S, Lowenfels A. Effect of voluntary exercise on azoxymethane-induced colon carcinogenesis in male F344 rats. Cancer research. 1988;48(24 Pt 1):7079–7081. [PubMed] [Google Scholar]
  • 34.Saez Mdel C, Barriga C, Garcia JJ, Rodriguez AB, Ortega E. Exercise-induced stress enhances mammary tumor growth in rats: beneficial effect of the hormone melatonin. Molecular and cellular biochemistry. 2007;294(1–2):19–24. doi: 10.1007/s11010-005-9067-5. [DOI] [PubMed] [Google Scholar]
  • 35.Sugie S, Reddy BS, Lowenfels A, Tanaka T, Mori H. Effect of voluntary exercise on azoxymethane-induced hepatocarcinogenesis in male F344 rats. Cancer letters. 1992;63(1):67–72. doi: 10.1016/0304-3835(92)90091-9. [DOI] [PubMed] [Google Scholar]
  • 36.Thorling EB, Jacobsen NO, Overvad K. Effect of exercise on intestinal tumour development in the male Fischer rat after exposure to azoxymethane. European journal of cancer prevention : the official journal of the European Cancer Prevention Organisation. 1993;2(1):77–82. doi: 10.1097/00008469-199301000-00012. [DOI] [PubMed] [Google Scholar]
  • 37.Westerlind KC, McCarty HL, Schultheiss PC, Story R, Reed AH, Baier ML, et al. Moderate exercise training slows mammary tumour growth in adolescent rats. European journal of cancer prevention : the official journal of the European Cancer Prevention Organisation. 2003;12(4):281–287. doi: 10.1097/00008469-200308000-00007. [DOI] [PubMed] [Google Scholar]
  • 38.Whittal KS, Parkhouse WS. Exercise during adolescence and its effects on mammary gland development, proliferation, and nitrosomethylurea (NMU) induced tumorigenesis in rats. Breast cancer research and treatment. 1996;37(1):21–27. doi: 10.1007/BF01806628. [DOI] [PubMed] [Google Scholar]
  • 39.Whittal-Strange KS, Chadan S, Parkhouse WS. Exercise during puberty and NMU induced mammary tumorigenesis in rats. Breast cancer research and treatment. 1998;47(1):1–8. doi: 10.1023/a:1005838721890. [DOI] [PubMed] [Google Scholar]
  • 40.Woods JA, Davis JM, Kohut ML, Ghaffar A, Mayer EP, Pate RR. Effects of exercise on the immune response to cancer. Medicine and science in sports and exercise. 1994;26(9):1109–1115. [PubMed] [Google Scholar]
  • 41.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. Journal of applied physiology. 2009;106(3):911–918. doi: 10.1152/japplphysiol.91201.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.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 epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2008;17(8):1920–1929. doi: 10.1158/1055-9965.EPI-08-0175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.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 prevention research. 2012;5(3):414–422. doi: 10.1158/1940-6207.CAPR-11-0454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Zielinski MR, Muenchow M, Wallig MA, Horn PL, Woods JA. Exercise delays allogeneic tumor growth and reduces intratumoral inflammation and vascularization. Journal of applied physiology. 2004;96(6):2249–2256. doi: 10.1152/japplphysiol.01210.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Malicka I, Siewierska K, Pula B, Kobierzycki C, Haus D, Paslawska U, et al. The effect of physical training on the N-methyl-N-nitrosourea-induced mammary carcinogenesis of Sprague-Dawley rats. Experimental biology and medicine (Maywood, NJ) 2015 doi: 10.1177/1535370215587532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Piguet AC, Saran U, Simillion C, Keller I, Terracciano L, Reeves HL, et al. Regular exercise decreases liver tumors development in hepatocyte-specific PTEN-deficient mice independently of steatosis. Journal of hepatology. 2015;62(6):1296–1303. doi: 10.1016/j.jhep.2015.01.017. [DOI] [PubMed] [Google Scholar]
  • 47.Almeida PW, Gomes-Filho A, Ferreira AJ, Rodrigues CE, Dias-Peixoto MF, Russo RC, et al. Swim training suppresses tumor growth in mice. Journal of applied physiology. 2009;107(1):261–265. doi: 10.1152/japplphysiol.00249.2009. [DOI] [PubMed] [Google Scholar]
  • 48.Bacurau AV, Belmonte MA, Navarro F, Moraes MR, Pontes FL, Jr, Pesquero JL, et al. Effect of a high-intensity exercise training on the metabolism and function of macrophages and lymphocytes of walker 256 tumor bearing rats. Experimental biology and medicine (Maywood, NJ) 2007;232(10):1289–1299. doi: 10.3181/0704-RM-93. [DOI] [PubMed] [Google Scholar]
  • 49.Bacurau RF, Belmonte MA, Seelaender MC, Costa Rosa LF. Effect of a moderate intensity exercise training protocol on the metabolism of macrophages and lymphocytes of tumour-bearing rats. Cell biochemistry and function. 2000;18(4):249–258. doi: 10.1002/1099-0844(200012)18:4<249::AID-CBF879>3.0.CO;2-2. [DOI] [PubMed] [Google Scholar]
  • 50.Baracos VE. Exercise inhibits progressive growth of the Morris hepatoma 7777 in male and female rats. Canadian journal of physiology and pharmacology. 1989;67(8):864–870. doi: 10.1139/y89-135. [DOI] [PubMed] [Google Scholar]
  • 51.Daneryd P, Hafstrom L, Svanberg E, Karlberg I. Insulin sensitivity, hormonal levels and skeletal muscle protein metabolism in tumour-bearing exercising rats. European journal of cancer. 1995;31A(1):97–103. doi: 10.1016/0959-8049(94)00344-5. [DOI] [PubMed] [Google Scholar]
  • 52.Daneryd P, Westin T, Edstrom S, Soussi B. Tumour purine nucleotides and cell proliferation in response to exercise in rats. European journal of cancer. 1995;31a(13–14):2309–2312. doi: 10.1016/0959-8049(95)00441-6. [DOI] [PubMed] [Google Scholar]
  • 53.Gershbein LL, Benuck I, Shurrager PS. Influence of stress of lesion growth and on survival of animals bearing parenteral and intracerebral leukemia L1210 and Walker tumors. Oncology. 1974;30(5):429–435. doi: 10.1159/000224983. [DOI] [PubMed] [Google Scholar]
  • 54.Hoffman SA, Paschkis KE, Debias DA, Cantarow A, Williams TL. The influence of exercise on the growth of transplanted rat tumors. Cancer research. 1962;22:597–599. [PubMed] [Google Scholar]
  • 55.Jones LW, Antonelli J, Masko EM, Broadwater G, Lascola CD, Fels D, et al. Exercise modulation of the host-tumor interaction in an orthotopic model of murine prostate cancer. Journal of applied physiology. 2012;113(2):263–272. doi: 10.1152/japplphysiol.01575.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Jones LW, Eves ND, Courneya KS, Chiu BK, Baracos VE, Hanson J, et al. Effects of exercise training on antitumor efficacy of doxorubicin in MDA-MB-231 breast cancer xenografts. Clinical cancer research : an official journal of the American Association for Cancer Research. 2005;11(18):6695–6698. doi: 10.1158/1078-0432.CCR-05-0844. [DOI] [PubMed] [Google Scholar]
  • 57.Jones LW, Viglianti BL, Tashjian JA, Kothadia SM, Keir ST, Freedland SJ, et al. Effect of aerobic exercise on tumor physiology in an animal model of human breast cancer. Journal of applied physiology. 2010;108(2):343–348. doi: 10.1152/japplphysiol.00424.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Lira FS, Tavares FL, Yamashita AS, Koyama CH, Alves MJ, Caperuto EC, et al. Effect of endurance training upon lipid metabolism in the liver of cachectic tumour-bearing rats. Cell biochemistry and function. 2008;26(6):701–708. doi: 10.1002/cbf.1495. [DOI] [PubMed] [Google Scholar]
  • 59.Murphy EA, Davis JM, Barrilleaux TL, McClellan JL, Steiner JL, Carmichael MD, et al. Benefits of exercise training on breast cancer progression and inflammation in C3(1)SV40Tag mice. Cytokine. 2011;55(2):274–279. doi: 10.1016/j.cyto.2011.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Newton G. TUMOR SUSCEPTIBILITY IN RATS: ROLE OF INFANTILE MANIPULATION AND LATER EXERCISE. Psychological reports. 1965;16:127–132. doi: 10.2466/pr0.1965.16.1.127. [DOI] [PubMed] [Google Scholar]
  • 61.Radak Z, Gaal D, Taylor AW, Kaneko T, Tahara S, Nakamoto H, et al. Attenuation of the development of murine solid leukemia tumor by physical exercise. Antioxidants & redox signaling. 2002;4(1):213–219. doi: 10.1089/152308602753625979. [DOI] [PubMed] [Google Scholar]
  • 62.Sasvari M, Taylor AW, Gaal D, Radak Z. The effect of regular exercise on development of sarcoma tumor and oxidative damage in mice liver. Journal of sports science & medicine. 2011;10(1):93–96. [PMC free article] [PubMed] [Google Scholar]
  • 63.Uhlenbruck G, Order U. Can endurance sports stimulate immune mechanisms against cancer and metastasis? International journal of sports medicine. 1991;12(Suppl 1):S63–S68. doi: 10.1055/s-2007-1024753. [DOI] [PubMed] [Google Scholar]
  • 64.Yan L, Demars LC. Effects of non-motorized voluntary running on experimental and spontaneous metastasis in mice. Anticancer research. 2011;31(10):3337–3344. [PubMed] [Google Scholar]
  • 65.Betof AS, Lascola CD, Weitzel D, Landon C, Scarbrough PM, Devi GR, et al. Modulation of murine breast tumor vascularity, hypoxia and chemotherapeutic response by exercise. Journal of the National Cancer Institute. 2015;107(5) doi: 10.1093/jnci/djv040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Goh J, Endicott E, Ladiges WC. Pre-tumor exercise decreases breast cancer in old mice in a distance-dependent manner. American journal of cancer research. 2014;4(4):378–384. [PMC free article] [PubMed] [Google Scholar]
  • 67.Shalamzari SA, Agha-Alinejad H, Alizadeh S, Shahbazi S, Khatib ZK, Kazemi A, et al. The effect of exercise training on the level of tissue IL-6 and vascular endothelial growth factor in breast cancer bearing mice. Iranian journal of basic medical sciences. 2014;17(4):231–258. [PMC free article] [PubMed] [Google Scholar]
  • 68.Aveseh M, Nikooie R, Aminaie M. Exercise-induced changes in tumour LDH-B and MCT1 expression are modulated by oestrogen-related receptor alpha in breast cancer-bearing BALB/c mice. The Journal of physiology. 2015 doi: 10.1113/JP270463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Gueritat J, Lefeuvre-Orfila L, Vincent S, Cretual A, Ravanat JL, Gratas-Delamarche A, et al. Exercise training combined with antioxidant supplementation prevents the antiproliferative activity of their single treatment in prostate cancer through inhibition of redox adaptation. Free radical biology & medicine. 2014;77:95–105. doi: 10.1016/j.freeradbiomed.2014.09.009. [DOI] [PubMed] [Google Scholar]
  • 70.Hoffman-Goetz L, May KM, Arumugam Y. Exercise training and mouse mammary tumour metastasis. Anticancer research. 1994;14(6B):2627–2631. [PubMed] [Google Scholar]
  • 71.Hoffmann-Goetz L, MacNeil B, Arumugam Y. Tissue distribution of radiolabelled tumor cells in wheel exercised and sedentary mice. International journal of sports medicine. 1994;15(5):249–253. doi: 10.1055/s-2007-1021055. [DOI] [PubMed] [Google Scholar]
  • 72.Jadeski L, Hoffman-Goetz L. Exercise and in vivo natural cytotoxicity against tumour cells of varying metastatic capacity. Clinical & experimental metastasis. 1996;14(2):138–144. doi: 10.1007/BF00121210. [DOI] [PubMed] [Google Scholar]
  • 73.MacNeil B, Hoffman-Goetz L. Exercise training and tumour metastasis in mice: influence of time of exercise onset. Anticancer research. 1993;13(6A):2085–2088. [PubMed] [Google Scholar]
  • 74.MacNeil B, Hoffman-Goetz L. Effect of exercise on natural cytotoxicity and pulmonary tumor metastases in mice. Medicine and science in sports and exercise. 1993;25(8):922–928. [PubMed] [Google Scholar]
  • 75.Higgins KA, Park D, Lee GY, Curran WJ, Deng X. Exercise-induced lung cancer regression: mechanistic findings from a mouse model. Cancer. 2014;120(21):3302–3310. doi: 10.1002/cncr.28878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Zhao G, Zhou S, Davie A, Su Q. Effects of moderate and high intensity exercise on T1/T2 balance. Exercise immunology review. 2012;18:98–114. [PubMed] [Google Scholar]
  • 77.Roberts CJ, Stuhr KL, Hillard CJ. Swim stress differentially affects limbic contents of 2-arachidonoylglycerol and 2-oleoylglycerol. Neuroscience. 2012;204:74–82. doi: 10.1016/j.neuroscience.2011.11.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Fuss J, Ben Abdallah NM, Vogt MA, Touma C, Pacifici PG, Palme R, et al. Voluntary exercise induces anxiety-like behavior in adult C57BL/6J mice correlating with hippocampal neurogenesis. Hippocampus. 2010;20(3):364–376. doi: 10.1002/hipo.20634. [DOI] [PubMed] [Google Scholar]
  • 79.Richter SH, Gass P, Fuss J. Resting Is Rusting: A Critical View on Rodent Wheel-Running Behavior. The Neuroscientist : a review journal bringing neurobiology, neurology and psychiatry. 2014;20(4):313–325. doi: 10.1177/1073858413516798. [DOI] [PubMed] [Google Scholar]
  • 80.Hackney AC, Viru A. Twenty-four-hour cortisol response to multiple daily exercise sessions of moderate and high intensity. Clinical physiology. 1999;19(2):178–182. doi: 10.1046/j.1365-2281.1999.00157.x. [DOI] [PubMed] [Google Scholar]
  • 81.Thaker PH, Han LY, Kamat AA, Arevalo JM, Takahashi R, Lu C, et al. Chronic stress promotes tumor growth and angiogenesis in a mouse model of ovarian carcinoma. Nature medicine. 2006;12(8):939–944. doi: 10.1038/nm1447. [DOI] [PubMed] [Google Scholar]
  • 82.Zen M, Canova M, Campana C, Bettio S, Nalotto L, Rampudda M, et al. The kaleidoscope of glucorticoid effects on immune system. Autoimmunity reviews. 2011;10(6):305–310. doi: 10.1016/j.autrev.2010.11.009. [DOI] [PubMed] [Google Scholar]
  • 83.Sasso JP, Eves ND, Christensen JF, Koelwyn GJ, Scott J, Jones LW. A framework for prescription in exercise-oncology research. Journal of cachexia, sarcopenia and muscle. 2015;6(2):115–124. doi: 10.1002/jcsm.12042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Irwin ML, Smith AW, McTiernan A, Ballard-Barbash R, Cronin K, Gilliland FD, et al. Influence of pre- and postdiagnosis physical activity on mortality in breast cancer survivors: the health, eating, activity, and lifestyle study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2008;26(24):3958–3964. doi: 10.1200/JCO.2007.15.9822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Mina LA, Sledge GW., Jr Rethinking the metastatic cascade as a therapeutic target. Nature reviews Clinical oncology. 2011;8(6):325–332. doi: 10.1038/nrclinonc.2011.59. [DOI] [PubMed] [Google Scholar]
  • 86.Chambers AF, Groom AC, MacDonald IC. Dissemination and growth of cancer cells in metastatic sites. Nature reviews Cancer. 2002;2(8):563–572. doi: 10.1038/nrc865. [DOI] [PubMed] [Google Scholar]
  • 87.Day CP, Merlino G, Van Dyke T. Preclinical mouse cancer models: a maze of opportunities and challenges. Cell. 2015;163(1):39–53. doi: 10.1016/j.cell.2015.08.068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Gonzalez C. Drosophila melanogaster: a model and a tool to investigate malignancy and identify new therapeutics. Nature reviews Cancer. 2013;13(3):172–183. doi: 10.1038/nrc3461. [DOI] [PubMed] [Google Scholar]
  • 89.Gould SE, Junttila MR, de Sauvage FJ. Translational value of mouse models in oncology drug development. Nature medicine. 2015;21(5):431–439. doi: 10.1038/nm.3853. [DOI] [PubMed] [Google Scholar]
  • 90.Sharpless NE, Depinho RA. The mighty mouse: genetically engineered mouse models in cancer drug development. Nature reviews Drug discovery. 2006;5(9):741–754. doi: 10.1038/nrd2110. [DOI] [PubMed] [Google Scholar]
  • 91.White R, Rose K, Zon L. Zebrafish cancer: the state of the art and the path forward. Nature reviews Cancer. 2013;13(9):624–636. doi: 10.1038/nrc3589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.McCullough DJ, Stabley JN, Siemann DW, Behnke BJ. Modulation of blood flow, hypoxia, and vascular function in orthotopic prostate tumors during exercise. Journal of the National Cancer Institute. 2014;106(4) doi: 10.1093/jnci/dju036. dju036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Straussman R, Morikawa T, Shee K, Barzily-Rokni M, Qian ZR, Du J, et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature. 2012;487(7408):500–504. doi: 10.1038/nature11183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.McAllister SS, Gifford AM, Greiner AL, Kelleher SP, Saelzler MP, Ince TA, et al. Systemic endocrine instigation of indolent tumor growth requires osteopontin. Cell. 2008;133(6):994–1005. doi: 10.1016/j.cell.2008.04.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.McAllister SS, Weinberg RA. Tumor-host interactions: a far-reaching relationship. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2010;28(26):4022–4028. doi: 10.1200/JCO.2010.28.4257. [DOI] [PubMed] [Google Scholar]
  • 96.Goodwin PJ, Ennis M, Pritchard KI, Trudeau ME, Koo J, Taylor SK, et al. Insulin- and obesity-related variables in early-stage breast cancer: correlations and time course of prognostic associations. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2012;30(2):164–171. doi: 10.1200/JCO.2011.36.2723. [DOI] [PubMed] [Google Scholar]
  • 97.Sheen-Chen SM, Chen WJ, Eng HL, Chou FF. Serum concentration of tumor necrosis factor in patients with breast cancer. Breast cancer research and treatment. 1997;43(3):211–215. doi: 10.1023/a:1005736712307. [DOI] [PubMed] [Google Scholar]
  • 98.Sheen-Chen SM, Liu YW, Eng HL, Chou FF. Serum levels of hepatocyte growth factor in patients with breast cancer. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2005;14(3):715–717. doi: 10.1158/1055-9965.EPI-04-0340. [DOI] [PubMed] [Google Scholar]
  • 99.Hursting SD, Berger NA. Energy balance, host-related factors, and cancer progression. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2010;28(26):4058–4065. doi: 10.1200/JCO.2010.27.9935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Glass OK, Inman BA, Broadwater G, Courneya KS, Mackey JR, Goruk S, et al. Effect of aerobic training on the host systemic milieu in patients with solid tumours: an exploratory correlative study. British journal of cancer. 2015;112(5):825–831. doi: 10.1038/bjc.2014.662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Joyner MJ, Green DJ. Exercise protects the cardiovascular system: effects beyond traditional risk factors. The Journal of physiology. 2009;587(Pt 23):5551–5558. doi: 10.1113/jphysiol.2009.179432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Kodama S, Saito K, Tanaka S, Maki M, Yachi Y, Asumi M, et al. Cardiorespiratory fitness as a quantitative predictor of all-cause mortality and cardiovascular events in healthy men and women: a meta-analysis. JAMA : the journal of the American Medical Association. 2009;301(19):2024–2035. doi: 10.1001/jama.2009.681. [DOI] [PubMed] [Google Scholar]
  • 103.Lee IM, Shiroma EJ, Lobelo F, Puska P, Blair SN, Katzmarzyk PT, et al. Effect of physical inactivity on major non-communicable diseases worldwide: an analysis of burden of disease and life expectancy. Lancet. 2012;380(9838):219–229. doi: 10.1016/S0140-6736(12)61031-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Pedersen L, Christensen JF, Hojman P. Effects of exercise on tumor physiology and metabolism. Cancer journal. 2015;21(2):111–116. doi: 10.1097/PPO.0000000000000096. [DOI] [PubMed] [Google Scholar]
  • 105.Fowler JF. 21 years of biologically effective dose. The British journal of radiology. 2010;83(991):554–568. doi: 10.1259/bjr/31372149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Dewhirst MW, Viglianti BL, Lora-Michiels M, Hanson M, Hoopes PJ. Basic principles of thermal dosimetry and thermal thresholds for tissue damage from hyperthermia. International journal of hyperthermia : the official journal of European Society for Hyperthermic Oncology, North American Hyperthermia Group. 2003;19(3):267–294. doi: 10.1080/0265673031000119006. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1
2
3

RESOURCES