You are on page 1of 14

Cancer Treatment Reviews 40 (2014) 327–340

Contents lists available at SciVerse ScienceDirect

Cancer Treatment Reviews


journal homepage: www.elsevierhealth.com/journals/ctrv

General and Supportive Care

Evidence-based physical activity guidelines for cancer survivors: Current


guidelines, knowledge gaps and future research directions
L.M. Buffart a,⇑, D.A. Galvão b,1, J. Brug a,2, M.J.M. Chinapaw c,3, R.U. Newton b,4
a
EMGO Institute for Health and Care Research and the VU University Medical Center, Department of Epidemiology and Biostatistics, Van der Boechorststraat 7,
1081 BT Amsterdam, The Netherlands
b
Edith Cowan University Health and Wellness Institute, Edith Cowan University, Joondalup, WA 6027, Australia
c
EMGO Institute for Health and Care Research and the VU University Medical Center, Department of Public and Occupational Health, Van der Boechorststraat 7,
1081 BT Amsterdam, The Netherlands

a r t i c l e i n f o a b s t r a c t

Article history: Physical activity during and after cancer treatment has beneficial effects on a number of physical and psy-
Received 7 February 2013 chosocial outcomes. This paper aims to discuss the existing physical activity guidelines for cancer survi-
Received in revised form 20 June 2013 vors and to describe future research directions to optimize prescriptions. Studies on physical activity
Accepted 25 June 2013
during and after cancer treatment were searched in PubMed, Clinicaltrials.gov, Australian New Zealand
Clinical Trials Registry, and Dutch Trial registry. Physical activity guidelines for cancer survivors suggest
that physical activity should be an integral and continuous part of care for all cancer survivors. However,
Keywords:
the development of these guidelines has been limited by the research conducted. To be able to develop
Exercise
Health
more specific guidelines, future studies should focus on identifying clinical, personal, physical, psychoso-
Neoplasms cial, and intervention moderators explaining ‘for whom’ or ‘under what circumstances’ interventions
Physical activity work. Further, more insight into the working mechanisms of exercise interventions on health outcomes
Quality of life in cancer survivors is needed to improve the efficacy and efficiency of interventions. Finally, existing pro-
Review grams should embrace interests and preferences of patients to facilitate optimal uptake of interventions.
In conclusion, current physical activity guidelines for cancer survivors are generic, and research is needed
to develop more personalized physical activity guidelines.
Ó 2013 Elsevier Ltd. All rights reserved.

Introduction [6]. Several reviews and meta-analyses demonstrate beneficial ef-


fects of physical activity (PA) and exercise (i.e. form of PA that is
In developed countries, approximately one in three persons will planned, structured and repetitive and aims to improve fitness,
be directly affected by cancer before the age of 75 years, with breast performance or health [7]) in cancer survivors during and after
cancer, prostate cancer, lung cancer and colorectal cancer the most treatment on physical and psychosocial outcomes, [8,9] including
common diagnoses [1]. It has been estimated that 12.7 million can- increased aerobic fitness, [10] reduced fatigue [11,12] and depres-
cer cases and 7.6 million cancer deaths occurred worldwide in 2008 sion, [13] and improved QoL [14,15]. Sufficient levels of PA may
[1]. Advances in early detection and treatment have improved sur- also be important to improve disease free and overall survival.
vival rates over the past decades, with approximately 60% of pa- Observational studies showed higher levels of moderate-to-
tients living over 5 years after diagnosis [2,3]. However, cancer vigorous PA to be associated with lower mortality risk in survivors
and its treatment are often associated with physical and psychoso- [16] of breast, [17–19] colon, [20,21] and prostate cancer, [22,23]
cial problems, negatively affecting quality of life (QoL) [4,5]. with physically active survivors having approximately 50% lower
Anyone who has been diagnosed with cancer, from the time of mortality. However, to establish a causal relationship between PA
diagnosis through the rest of life, is considered a cancer survivor and survival, randomized controlled trials (RCTs) are needed. The
first RCT evaluating the effects of PA on survival is currently being
⇑ Corresponding author. Tel.: +31 20 444 9931; fax: +31 20 444 8387. conducted among survivors with colon cancer who have com-
E-mail addresses: l.buffart@vumc.nl (L.M. Buffart), d.galvao@ecu.edu.au pleted adjuvant chemotherapy in the Colon Health and Life-Long
(D.A. Galvão), j.brug@vumc.nl (J. Brug), m.chinapaw@vumc.nl (M.J.M. Chinapaw), Exercise Change (CHALLENGE) trial [24].
r.newton@ecu.edu.au (R.U. Newton). Given the increasing number of studies showing the safety and
1
Tel.: +61 8 6304 3420; fax: +61 8 6304 2499. benefits of PA, it should be part of standard care for all cancer sur-
2
Tel.: +31 20 444 8192; fax: +31 20 444 8171.
3 vivors. Evidence-based PA guidelines have been published, but the
Tel.: +31 20 444 8203; fax: +31 20 444 8387.
4
Tel.: +61 8 6304 3443; fax: +61 8 6304 5106. development of these guidelines is limited by the research

0305-7372/$ - see front matter Ó 2013 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.ctrv.2013.06.007
328 L.M. Buffart et al. / Cancer Treatment Reviews 40 (2014) 327–340

conducted in this area. This paper reviews the current evidence- conducted more systematically and based on categories A–D, out-
based PA guidelines for cancer survivors, identifies current knowl- lined by the National Heart, Lung and Blood Institute. Overall, the
edge gaps and describes the research needed to fill these gaps. This ACSM guidelines fall into evidence category level B, indicating that
synopsis may inform future studies evaluating exercise as medi- few RCTs exist or they are small and results are inconsistent. The
cine for cancer, as well as the development of more personalized expert panel reported consistent evidence regarding the safety of
PA guidelines for cancer survivors. exercise during and after cancer treatment (including intensive
treatments such as bone marrow transplant) and reported that
Current guidelines and their empirical basis improvements can be expected in aerobic fitness, muscle strength,
QoL and fatigue in survivors of breast, prostate, and hematologic
In 2003, the American Cancer Society (ACS) published a report in- cancer. The expert panel acknowledged that the extent to which
tended to present health care providers with the best possible infor- these findings may generalize to other cancer survivor groups re-
mation on which to assist cancer survivors and their families to mains unknown, and that exercise prescriptions should be individ-
make informed choices related to nutrition and PA [25]. This was ualized according to cancer survivors’ pre-treatment aerobic
ACS’s second report on nutrition, but the first to also include infor- fitness, medical co-morbidities, response to treatment, and the
mation on PA. Although evidence was generally insufficient to draw immediate or persistent negative effects of treatment.
conclusions about the benefits and risks of PA, a probable beneficial The ACSM generally recommends avoiding inactivity, returning to
effect of PA on QoL was found for survivors of breast and colorectal normal daily activities as quickly as possible after surgery, and to con-
cancer during and after cancer treatment, and for survivors of pros- tinue normal daily activities and exercise as much as possible during
tate cancer after cancer treatment, as well as for overall survival after and after nonsurgical treatments. Recommendations for aerobic,
treatment for colorectal, lung, and prostate cancer. Importantly, no resistance, and flexibility exercises are the same as the age-appropri-
evidence of harm was reported. The ACS report was updated in ate PA guidelines for the general population [28,29] (Table 1), with
2006 [26]. Despite methodological limitations and small sample several alterations for patients with and at risk for lymphedema, skel-
sizes, evidence strongly suggested that exercise is safe and feasible etal fractures or infections. In addition, several contraindications for
during cancer treatment and can improve physical functioning and exercise were identified, including arm and shoulder problems sec-
some aspects of QoL [26]. It was also suggested that regular PA after ondary to breast cancer treatment, having an ostomy after colon can-
cancer treatment is essential for recovery and to improve fitness. cer, or swelling or inflammation in the abdomen, groin, or lower
Also, after recovery it was concluded that PA is important to promote extremity following gynecologic cancer (Table 2).
overall health, QoL and longevity. For the prevention of cancer, the In 2011, PA guidelines for cancer survivors were developed in
ACS recommends at least 30 min, and preferably 45–60 min, of the Netherlands recommending at least moderate intensity exer-
moderate-to-vigorous PA above usual activities, on 5 or more days cise to all cancer survivors and to tailor exercise programs to indi-
of the week (Table 1). Although these PA levels have not been studied vidual fitness levels to obtain optimal training effects [30,31].
systematically in cancer survivors, these recommendations may Individual cardiopulmonary fitness levels can be assessed by mea-
also benefit cancer survivors, as it may reduce the risk of recurrence suring peak oxygen uptake (peakVO2) which is feasible and safe for
or developing secondary cancers and extend survival. Therefore, ACS cancer survivors prior to an exercise program [32]. In case of logis-
recommended that daily and regular PA is preferred, but any steps tical problems related to cost, lack of appropriate equipment or
that are taken to move from a sedentary to a more active lifestyle experienced health care professionals to use this test in clinical
should be encouraged, with higher levels of PA leading to more practice, [32] a 6 min walk-test, shuttle walk test, or steep ramp
health benefits [25,26] (Table 1). However, specific precautions test [33] is recommended [32]. To adequately tailor resistance
should be heeded for survivors with severe anemia, compromised exercises to individual patients, the Dutch guidelines recommend
immune function, severe fatigue, indwelling catheters and periphe- using indirect 1-repetition maximum (1-RM) assessments to as-
ral neuropathies or ataxia [25,26] (Table 2). sess muscle strength (Table 1) [33,31].
In 2009, Exercise and Sport Science Australia (ESSA) published In 2012, the ACS updated their report from 2006 and 2003 [34].
exercise guidelines for cancer survivors with the intention of guid- Generally, they recommended cancer survivors to engage in regu-
ing exercise practitioners in their work with cancer survivors [27]. lar PA, to avoid inactivity and return to normal daily activities as
The document provided specific recommendations for aerobic soon as possible following diagnosis. The ACS recommends cancer
exercise and for the first time, also for resistance exercise (Table 1), survivors to aim for exercising at least 150 min per week and to in-
particularly important for cancer survivors experiencing loss of clude strength exercises following the comprehensive guidelines
lean mass during and following treatment. The ESSA position state- suggested by the ACSM expert panel, after consideration of specific
ment indicated that the optimal exercise prescription remains un- precautions (Table 2). In the same year, the British Association of
known, and may depend on the type of cancer, treatment and Sport and Exercise Science (BASES) published an expert statement
characteristics of the patient. It was recognized that participation on exercise and cancer survivorship [35]. They reported that there
in some PA is better than none, and that more is generally better is consistent evidence confirming that exercise is safe during and
than less, at least up to levels meeting national PA guidelines after cancer treatment, provided that individual limitations and
[27]. It was also recommended to avoid certain types of aerobic specific side effects associated with cancer therapies are consid-
or resistance exercises (e.g. high impact activities) during periods ered and monitored, and that improvements in aerobic and muscu-
of increased risk of infection, ataxia, dizziness, peripheral sensory lar fitness, QoL and fatigue can be expected. They recommended all
neuropathy, low platelet counts, bone pain, or in patients with pri- cancer survivors be encouraged to avoid being sedentary, and un-
mary or metastatic bone cancer (Table 2). less advised otherwise, follow the PA guidelines of the general UK
In 2010, the American College of Sports Medicine (ACSM) orga- population [36] (Table 1), as no formal guidelines for cancer survi-
nized a roundtable on exercise guidelines for cancer survivors given vors have been published in the UK.
the proliferation of exercise programs, and the lack of structured
exercise guidelines for health and fitness professionals to draw Knowledge gaps to be addressed
upon in working with cancer survivors [28]. Evidence was reviewed
for adult survivors of breast, prostate, colon, hematologic, and gyne- Although the significant work conducted to formulate the above
cologic cancers. This was the first time that evaluation of the evi- mentioned guidelines is important and has changed clinical
dence in exercise oncology for development of guidelines was practice for management of cancer, current recommendations are
L.M. Buffart et al. / Cancer Treatment Reviews 40 (2014) 327–340 329

Table 1
Overview of PA guidelines for cancer survivors.

Year Institute PA recommendations


2003/ ACS  Follow guidelines for the general population for cancer prevention, i.e. at least 30 min of moderate-to-vigorous PA, above
2006 [25,26] the usual activities, on 5 or more days of the week. Forty-five to 60 min of intentional PA are preferable
 Daily and regular activity may be preferred and may be the goal. Any steps that are taken to move from a sedentary to an
active lifestyle should be encouraged

2009 ESSA Aerobic exercise:  Type: exercises involving large muscle groups
[27]  Frequency: at least 3–5 times per week, but daily exercise may be preferable for deconditioned patients who do lower
intensity and shorter duration exercise sessions
 Intensity: moderate, depending on current fitness level. Guidelines recommend 50–75% VO2max, or HRreserve, 60–80%
HRmax, or a RPE of 11–14 (original Borg scale)
 Duration: at least 20–30 min continuous exercise; however, deconditioned patients or those experiencing severe side
effects of treatment may need to combine short bouts (e.g. 3–5 min) with rest intervals
 Progression: would be slower and more gradual for deconditioned patients or those who are experiencing severe side-
effects of treatment

Resistance exercise:  Type: exercises using machine-weights, free weights, body weight and/or elastic bands that involve major functional
lower- and upper-body muscle groups. Targeting large muscle groups. Exercises should be dynamic in nature using both
concentric and eccentric muscle contractions
 Frequency: 1–3 times per week, with minimum one rest day between sessions
 Intensity: 50–80% of 1-RM or 6–12 RM
 Duration: 6–10 exercises, 1–4 sets per muscle group
 Progression: as described for aerobic exercise

2010 ACSM Aerobic exercise:  Avoid inactivity and be as physically active as abilities and conditions allow
[28]  For patients with breast, prostate, colon, hematologic (no HSCT) and gynaecologic cancer, recommendations are the same
as age-appropriate guidelines for the general American population, i.e. to engage in at least 150 min per week of moderate
intensity or 75 min per week of vigorous intensity aerobic PA or an equivalent combination of moderate and vigorous
intensity aerobic PA, for adults aged 18–64 years. Activity should be done in episodes of at least 10 min per session
and preferably spread throughout the week; some activity is better than nothing and exceeding the guideline is likely
to provide additional health benefits
 For patients with HSCT, it is ok to exercise everyday, however lighter intensity and lower progression of intensity is
recommended
 Women with gynaecologic cancer who are morbidity obese may require additional supervision and altered programming

Resistance exercise:  For patients with prostate, colon, and hematologic cancer, recommendations for resistance training are the same as age-
appropriate guidelines for the general American population, i.e. Muscle-strengthening activities involving all major mus-
cle groups at least 2 days per week for adults aged 18–64 years
 For patients with breast cancer, start with a supervised program of at least 16 sessions and very low resistance; progress
resistance at small increments
 For patients with prostate cancer: add pelvic floor exercises for those who undergo radical prostatectomy
 For patients with colon cancer with a stoma, start with low resistance and progress resistance slowly to avoid herniation
at the stoma
 For bone marrow transplant patients, resistance exercises might be more important than aerobic exercise

2011 CCCN  Systematically identify physical and psychosocial problems using for example the distress thermometer
[30,31]  At least moderate intensity physical exercise should be recommended to all patients with cancer, using the Dutch phys-
ical activity norms as a guideline: at least 30 min moderate intensity (4 MET, and 3 MET for people aged P55 years) on at
least 5 but preferable all days per week
 Physical exercise programs should be tailored to patients’ individual cardiopulmonary fitness levels to obtain optimal
training effects
 Assessments of CPET, 6-min walk test, shuttle walk test or steep ramp test should be performed to tailor aerobic exercises
to individual disease characteristics, preferences and goals
 Assessments of 1-RM should be performed to tailor resistance exercises to individual disease characteristics, preferences
and goals

2012 ACS [34]  Engage in regular PA


 Avoid inactivity and return to normal daily activities as soon as possible following diagnosis
 Aim to exercise at least 150 min per week
 Include strength training exercises at least 2 days per week
 Follow the survivor-specific guidelines written by the ACSM expert panel

2012 BASES  All cancer survivors should be encouraged, as a minimum, to avoid being sedentary
[35]  Unless advised otherwise, cancer survivors should follow the health-related PA guidelines provided for the general UK
population i.e. 150 min each week of moderate to vigorous intensity PA for adults (and adults should aim to do some
PA every day) and include muscle strengthening activities twice a week

ACS = American Cancer Society; ACSM = American College of Sports Medicine; BASES = British Association of Sport and Exercise Science; CCCN = Comprehensive Cancer
Center the Netherlands; CPET = cardiopulmonary exercise testing; ESSA = Exercise and Sport Science Australia; HR = heart rate; HSCT = hematopoietic stem cell transplan-
tation; MET = metabolic equivalent; RM = repetition maximum; RPE = rating of perceived exertion; UK = United Kingdom; VO2 = oxygen uptake.

rather general due to gaps in specific areas of exercise oncology. meta-analysis by Speck et al. [9] concluded that the current vol-
The ultimate aim is to have specific guidelines to inform a given ume of literature was not large enough to summarize findings
exercise intervention (e.g. mode, frequency, intensity, duration), per time point within PA across the Cancer Continuum (PACC)
for a given cancer site at a particular phase of the cancer trajectory framework, [37] which proposes four post-diagnosis (pre-treat-
(e.g. during treatment, survivorship, end of life), for specific end ment, treatment, survivorship, and end of life) cancer-related time
points (e.g. fatigue, physical function, QoL, survival) and to address periods [37]. Nevertheless, results of exercise during and post
specific treatment side-effects and co-morbidities (e.g. osteoporo- cancer treatment were separately presented in this meta-analysis.
sis, metabolic syndrome, sarcopenia, functional decline) [28]. A During treatment, small-to-moderate effects of PA were reported
330 L.M. Buffart et al. / Cancer Treatment Reviews 40 (2014) 327–340

Table 2
Precautions for exercise.

Year Institute Precautions


2003/ ACS  Survivors with severe anemia should delay exercise, other than activities of daily living, until the anemia is improved
2006 [25,26]  Survivors with compromised immune function should avoid public gyms and public pools until their white blood cell counts return to safe
levels. Survivors who have completed a bone marrow transplant are usually advised to avoid such exposures for one year after
transplantation
 Survivors suffering from severe fatigue from their therapy may not feel up to an exercise program, so they may be encouraged to do 10 min
of stretching exercises daily
 Survivors undergoing radiation should avoid chlorine exposure to irradiated skin (e.g. from swimming pools)
 Survivors with indwelling catheters or feeding tubes should be cautious or avoid pool, lake, or ocean water or other microbial exposures that
may result in infections, as well as resistance training of muscles in the area of the catheter to avoid dislodgment
 Survivors with significant peripheral neuropathies or ataxia may have a reduced ability to use the affected limbs because of weakness or loss
of balance. They may do better with a stationary reclining bicycle, for example, than walking on a treadmill

2009 ESSA  Patients are recommended to avoid swimming or the use of public facilities such as machine-weights in gymnasiums during periods of
[27] increased infections (e.g. low absolute neutrophil counts, when catheters are being used, during wound recovery from surgery)
 Patients with primary or metastatic bone cancer, patients with low platelet counts, or patients experiencing bone pain should avoid high-
impact activities or contact sports
 Patients with ataxia, dizziness, or peripheral sensory neuropathy are recommended to avoid activities requiring balance and coordination
(e.g. treadmill exercise, cycling) and specific resistance exercises such as those using free weights
 Patients should avoid the use of public facilities (e.g. local gymnasium) during periods of increased infection risk
 When nausea, dyspnoea, fatigue and/or muscle weakness exist, exercise intensity and duration should be prescribed to tolerance

2010 ACSM  Beware of fracture risk in patients with breast and prostate cancer who are treated with hormone of androgen deprivation therapy, respec-
[28] tively, or diagnosed with osteoporosis or bone metastases
 Women with immediate arm or shoulder problems secondary to breast cancer treatment should seek medical care to resolve those issues
before exercise training with the upper body
 If an ostomy is present in patients with colon cancer, physician permission is recommended before participation in contact sport and weight
training, excessive intra-abdominal pressure should be avoided, and modifications will be needed for swimming and contact sports
 In adults with HSCT, care should be taken to avoiding overtraining given immune effects of vigorous exercise
 If peripheral neuropathy is present in patients with gynaecologic cancer, a stationary bike might be preferable over weight bearing exercise
 Women with gynaecologic cancer with swelling or inflammation of the abdomen, groin or lower extremities should seek medical care to
resolve these issues before exercise training with the lower body
 Patients with bone metastases may need to alter their exercise program given the increased risk for skeletal fractures
 Care should be taken to reduce infection risk in fitness centers for patients who are currently undergoing chemotherapy or radiation treat-
ment or have compromised immune function
 Patients with cardiac conditions will require modifications and may require increased supervision for safety

2012 ACS [34]  See also precautions from ACS published in 2003 and 2006
 Survivors with multiple or uncontrolled comorbidities need to consider modifications to their exercise program in consultation with their
physicians

ACS = American Cancer Society; ACSM = American College of Sports Medicine; ESSA = Exercise and Sport Science Australia.

for aerobic fitness, upper and lower body muscle strength, body on QoL in cancer survivors. Greater QoL benefits were found in
weight, functional QoL, anxiety, and self-esteem. Post treatment, lymphoma patients who had no preference for exercise, were
large effects were found for upper and lower body muscle strength unmarried, had normal weight or were obese, or had poor/fair
and breast cancer-specific concerns, and small to moderate effects health [41]. During chemotherapy for breast cancer, aerobic train-
for PA level, aerobic fitness, overall QoL, insulin-like growth factor ing was most beneficial for patients who had no exercise prefer-
(IGF)-1, and symptoms and side effects [9]. For many outcomes, ence, were unmarried, aged <50 years and had more advanced
there have been too few studies to draw conclusions. Similar disease stage [42]. Resistance exercise was most beneficial for pa-
small-to-moderate effect sizes were reported in other meta- tients who preferred it, were unmarried, received nontaxane-based
analyses on this topic [12–15,38,39]. chemotherapy and had more advanced disease stage [42]. Prostate
Explanations for small-to-moderate effects of exercise in cancer cancer patients with lower psychosocial function at baseline
survivors include problems with program participation and adher- showed higher improvements in QoL after completing a lifestyle
ence, and the use of one-size-fits-all approaches in a heterogeneous PA program [43]. These studies indicate that demographic, clinical
group of cancer survivors. Thus, similar to developments in personal- and personal factors may help us to understand differences in exer-
ized primary cancer treatment, exercise prescription for cancer survi- cise responses. Insight into moderators of interventions is needed
vors should be optimally tailored to the individual characteristics, to maximize benefits of interventions for individual patients.
states, needs, capabilities and preferences of a patient. To be able to Future studies should focus on identifying relevant moderators of
shift towards personalized PA and exercise programs, it is essential exercise intervention effects.
to know which mode of exercise, at which frequency, intensity and
duration is best for whom, and under what circumstances, for exam-
Exercise mode, frequency, intensity and duration
ple the timing with respect to the treatment phase (i.e. treatment
RCTs directly comparing different exercise modes, frequencies,
moderators). To further optimize exercise interventions, it is essential
intensities and durations in cancer survivors are necessary to iden-
to gain insight into the working mechanisms and to identify effective
tify optimal exercise prescriptions. Unfortunately, few RCTs have
intervention components (i.e. treatment mediators).
been published (Table 3). Regarding exercise mode, Courneya
et al. [44] found peak oxygen uptake (peakVO2) to be superior after
Moderators of exercise intervention effects aerobic exercise training (AET) compared with usual care (UC) and
resistance exercise training (RET), and lower and upper body
Specific treatment moderators can help determine which pa- strength to be superior after RET compared with AET and UC in wo-
tients should receive a particular treatment [40]. Few previous men with breast cancer during chemotherapy. Neither exercise
studies have examined moderators of exercise intervention effects interventions prevented weight gain, but they both altered body
L.M. Buffart et al. / Cancer Treatment Reviews 40 (2014) 327–340 331

Table 3
Overview of studies directly comparing different exercise modes, frequencies, intensities, durations, and timing.

Author Population Study design Main findings


Trial identification Diagnosis, n
Exercise mode
Courneya et al. [44]Supervised Breast cancer during 3-armed RCT:  PeakVO2 (ml/kg/min):
Trial of Aerobic versus chemotherapy; n = 242 AET > RET (4 1.4, 95% CI = 0.1; 2.7, p = 0.031)
 AET
Resistance Training (START) RET = UC (4 0.5, 95% CI = -0.8; 1.8, N.S.)
 RET
trial AET > UC (4 2.0, 95% CI = 0.6; 3.3, p = 0.004)
 UC
 Lower body strength (1 RM in kg):
RET > AET (4 5.2, 95% CI = 2.5; 7.9, p = 0.001)
RET > UC (4 6.8, 95% CI = 4.2; 9.5, p = 0.001)
AET = UC (4 1.6, 95% CI = 1.1; 0.12, N.S.)
 Upper body strength (1 RM in kg):
RET > AET (4 6.8, 95% CI = 4.5; 9.0, p = 0.001)
RET > UC (4 7.7, 95% CI = 5.5; 9.9, p = 0.001)
AET = UC (4 1.0, 95% CI = 1.3; 3.2, N.S.)
 Body weight (kg):
AET = RET (4 -0.7, 95% CI = 0.4; 0.9, N.S.)
RET = UC (4 0.5, 95% CI = 1.6; 0.6, N.S.)
AET = UC (4 0.2, 95% CI = 0.9; 1.4, N.S.)
 Body fat (%):
AET = RET (4 0.0, 95% CI = 1.0; 1.0, N.S.)
RET = UC (4 0.9, 95% CI = 0.1; 1.9, N.S.)
AET > UC (4 0.9, 95% CI = 0.1; 1.9, p = 0.076)
 Lean mass (kg):
RET = AET (4 0.6, 95% CI = 0.1; 1.3, N.S.)
RET > UC (4 1.0, 95% CI = 0.3; 1.6, p = 0.004)
AET = UC (4 0.3, 95% CI = 0.3; 1.0, N.S.)
 Chemotherapy completion rate:
RET = AET (4 3.3, 95% CI = 2.5; 9.2, p = 0.266)
RET > UC (4 5.7, 95% CI = 0.4; 11.0, p = 0.033)
AET = UC (4 not reported, N.S.)
Self esteem:
RET = AET (4 0.0, 95% CI = 1.1; 1.1, N.S.)
RET > UC (4 1.2, 95% CI = 0.2; 2.3, p = 0.025)
AET > UC (4 1.2, 95% CI = 0.1; 2.3, p = 0.026)
Segal et al. [45] Men with prostate cancer 3-armed RCT:  QoL*:
during radiation therapy; RET > UC (4 4.3, 95% CI = 0.9; 7.8, p = 0.015)
 AET
n = 121 AET > UC (4 2.5, 95% CI = -0.9; 5.9, p = 0.141)
 RET
Fatigue*:
 UC
RET > UC (4 4.8, 95% CI = 1.8; 7.8, p = 0.002)
AET > UC (4 2.7, 95% CI = 0.3; 5.6, p = 0.08)
 PeakVO2 (ml/kg/min)
RET > UC (4 1.6, 95% CI = 1.0; 3.1, p = 0.037)
AET > UC (4 1.4, 95% CI = 0.1; 2.8, p = 0.063)
Lower body strength (8 RM in kg):
RET > UC (4 25.1, 95% CI = 15.3; 34.9, p < 0.001)
AET = UC (4 3.7, 95% CI = 6.0; 13.4 p = 0.45)
 Upper body strength (8 RM in kg):
RET > UC (4 13.7, 95% CI = 10.7; 16.6, p < 0.001)
AET > UC (4 4.0, 95% CI = 1.2; 6.9, p = 0.006)
 Body fat (%):
RET > UC (4 1.5, 95% CI = 3.1; 0.03, p = 0.055)
AET = UC (4 0.2, 95% CI = 1.7; 1.4, p = 0.847)
 Triglyceride (mmol/L):
RET > UC (0.1 (SD = 0.6) decrease in RET vs 0.3 (SD = 0.7)
increase in
UC, p = 0.036)
AET = UC (0.3 (SD = 0.7) increase in AET and UC, N.S.)
Newton et al. Men with prostate cancer 3-armed RCT: Study is ongoing
[48](ACTRN12609000200280) during ADT; n = 195
 RET + impact loading
 RET + AET
 UC
Jones et al. [46]The Lung Cancer Histologically confirmed 4-armed RCT: Study is ongoing
Exercise Training (LUNGEVITY) stage I-IIIA post-operative
 RET
study (NCT00018255) NSCLC; n = 160
 AET
 RET + AET
 Attention-control
(progressive stretching)
Winters-Stone Older (>65 years) female 3-armed RCT: Study is ongoing
[50](NCT00662103) breast cancer after
 AET
treatment. n = 141
 RET
 Flexibility and relaxation (control)

(continued on next page)


332 L.M. Buffart et al. / Cancer Treatment Reviews 40 (2014) 327–340

Table 3 (continued)

Author Population Study design Main findings


Trial identification Diagnosis, n
Young-McCaughan Sedentary adults after 2-armed RCT: Study is ongoing
[51](NCT00237926) treatment with
 AET
chemotherapy or radiation
 RET
therapy n = 58
National Institutes of Health Adult survivors of solid 3-armed RCT: Study is ongoing
Clinical Center tumours. n = 76
 Tai Chi
[47](NCT00246818)
 AET
 UC
Siew Yim Breast cancer after 3-armed RCT: Study is ongoing
[49](ACTRN12611000093987) treatment n = 114
 QuiGong
 AET (line dancing)
 UC
Exercise frequency

Exercise intensity
Burnham and Wilcox [54] Survivors of breast and 3-armed RCT: Differences at post-test between moderate (n = 6) and low
colon cancer n = 18 AET (n = 6):
 Moderate AET
 Low AET  PeakVO2 (ml/kg/min): p = 0.824
 UC  Body fat (%): p = 0.512
 Sit and reach (cm): p = 0.722
Exercise (n = 12) vs UC (n = 6)
 " PeakVO2 (ml/kg/min): p < 0.001
 ; Body fat (%): p < 0.001
 " Sit and reach (cm): p = 0.027
 " QoL: p < 0.001
 " Energy: p = 0.038
Gibbs et al. Breast cancer patients 3-armed RCT:  Lymphatic relative volume (%):
[55](ACTRN12612000256875) with lymphedema after High vs Low: N.S.
 High intensity RET
cancer treatment; n = 73 High vs UC: 4 8.6; p = 0.001
 Low intensity RET
Low vs UC: 4 7.8; p = 0.001
 UC
 Chest press (kg):
High vs Low: N.S.
High vs UC: 4 4.7; p = 0.001
Low vs UC: 4 3.8; p = 0.001
 Seated row (kg):
High vs Low: N.S.
High vs UC: 4 9.7; p = 0.001
Low vs UC: 4 4.9; p = 0.001
 Leg extension (kg):
High vs Low: N.S.
High vs UC: 4 5.6; p = 0.001
Low vs UC: 4 3.9; p = 0.001
 400 m walk (sec):
High vs Low: 4 16.3; p = 0.020
High vs UC: 4 36.2; p = 0.025
Low vs UC: 4 18.9; p = 0.004
 General fatigue (%)
High vs Low: N.S.
High vs UC: 4 28.2; p = 0.001
Low vs UC: 4 25.3; p = 0.001
 Body image (%)
High vs Low: N.S.
High vs UC: 4 20.8; p < 0.05
Low vs UC: 4 23.4; p < 0.05
 Breast symptoms (%)
High vs Low: N.S.
High vs UC: 4 9.5; p < 0.05
Low vs UC: 4 10.8; p < 0.05
 Arm symptoms (%)
High vs Low: N.S.
High vs UC: 4 15.2; p < 0.05
Low vs UC: 4 13.6; p < 0.05
Cormie et al. Women with breast cancer 3-armed RCT: Study is ongoing
[56](ACTRN12610000788077) related lymphedema.
 High intensity RET
n = 60
 Low intensity RET
 UC
Kampshoff et al. [57]Resistance Mixed group after 3-armed RCT: Study is ongoing
and Endurance exercise After chemotherapy; n = 400
 High intensity RET + AET
ChemoTherapy (REACT)
 Low-moderate intensity RET + AET
study.(NTR2153)
 Wait-list
L.M. Buffart et al. / Cancer Treatment Reviews 40 (2014) 327–340 333

Table 3 (continued)

Author Population Study design Main findings


Trial identification Diagnosis, n
Jones et al. [58]Exercise Intensity Postmenopausal, operable 3-armed RCT: Study is ongoing
trial (EXCITE)(NCT01186367) breast cancer after
 High intensity aerobic
treatment; n = 174
 Moderate-high intensity aerobic
 Attention control
Exercise duration
Sprod et al. [59] Breast cancer survivors; 3-armed RCT:  Cardiovascular endurance (treadmill time):
n = 114 6M=3M
 3 months exercise
6 M > UC (p < 0.05)
 6 months exercise
3 M > UC (p < 0.05)
 UC
 Forced Vital Capacity:
6 M > 3 M (p < 0.05)
6 M > UC (p < 0.05)
3 M = UC
 Upper body muscle endurance (bench press) :
6 M > 3 M (p < 0.05)
6 M = UC
3 M = UC
 Crunches:
6M=3M
6 M > UC (p < 0.05)
3 M > UC (p < 0.05)
 Affective fatigue:
6 M > 3 M (p < 0.05)
6 M = UC
3 M = UC
Timing of exercise
Gibbs et al. Breast cancer survivors 3-armed RCT: 12 weeks/3 months follow-up
[60](ACTRN1212000646842) scheduled for
 RET + AET during treatment  MFI, general fatigue (%)
radiotherapy; n = 63.
 RET + AET post treatment During vs Post: N.S./N.S.
 UC During vs UC: 4 17.2; p = 0.042/4 10.8; p = 0.012
Post vs UC: 4 14.4; p = 0.031/4 9.2; p = 0.031
 MFI, physical fatigue (%)
During vs Post: 4 5.5; p = 0.045/N.S.
During vs UC: 4 18.7; p = 0.034/4 11.1; p = 0.034
Post vs UC: 4 13.0; p = 0.038/4 10.3; p = 0.035
 Chest press (1-RM in kg)
During vs Post: N.S./4 3.2; p = 0.026
During vs UC: 4 6.5; p = 0.001/4 2.3; p = 0.002
Post vs UC: 4 5.3; p = 0.002/N.S.
 Seated row (1-RM in kg)
During vs Post: N.S./4 3.5; p = 0.002
During vs UC: 4 7.3; p = 0.002/4 1.5; p = 0.045
Post vs UC: 4 4.4; p = 0.001/N.S.
 Leg extension (1-RM in kg)
During vs Post: N.S./N.S.
During vs UC: 4 5.8; p = 0.006/4 1.4; p = 0.042
Post vs UC: N.S./N.S.
 Upper body endurance (rep)
During vs Post: N.S./N.S.
During vs UC: 4 6.3; p = 0.011/4 2.9; p = 0.038
Post vs UC: 4 4.5; p = 0.025/N.S.
 Lower body endurance (rep)
During vs Post: N.S./N.S.
During vs UC: 4 7.1; p = 0.022/4 3.9; p = 0.032
Post vs UC: 4 6.9; p = 0.021/N.S.
 Global health (%):
During vs Post: N.S./N.S.
During vs UC: 4 23.5; p = 0.006/N.S.
Post vs UC: N.S./N.S.
 Physical functioning (%)
During vs Post: N.S./N.S.
During vs UC: 4 12.0; p = 0.010/N.S.
Post vs UC: N.S./N.S.
 Role functioning (%)
During vs Post: N.S./N.S.
During vs UC: 4 13.3; p = 0.010/N.S.
Post vs UC: N.S./N.S.
 Emotional functioning (%)
During vs Post: N.S./N.S.
During vs UC: 4 20.1; p = 0.002/N.S.
Post vs UC: N.S./N.S.

(continued on next page)


334 L.M. Buffart et al. / Cancer Treatment Reviews 40 (2014) 327–340

Table 3 (continued)

Author Population Study design Main findings


Trial identification Diagnosis, n
Newton et al. Prostate cancer during 2-armed RCT Study is ongoing
[61](ACTRN12612000097842) ADT; n = 124
 Commence exercise simultaneously
with ADT
 start after 6 months
Walenkamp & Gietema [62] Testicular, colon and 2-armed RCT Study is ongoing
Optimal timing of PA in Cancer breast cancer n = 206
 PA program during (3 months) and
Treatment (ACT)
after chemotherapy (3 months)
study(NCT01642680)
 PA after chemotherapy (6 months)

ACTRN = Australian New Zealand Clinical Trials Registry Number; ADT = androgen deprivation therapy; AET = aerobic exercise training; NCT = ClinicalTrials.gov Identifier;
N.S. = not significant; NSCLC = non-small cell lung cancer; RCT = randomized controlled trial; RET = resistance exercise training; RM = repetition maximum; UC = usual care;
VO2 = oxygen uptake.
*
Unadjusted for covariates. > = more benefits.

composition compared to UC: AET prevented fat gain, and RET Regarding exercise duration, physiological and psychological
added lean body mass. Further, compared to AET (87.4%) and UC outcomes following 3 or 6 months of prescriptive, individualized
(84.1%), chemotherapy completion rate was higher during RET exercise were compared with UC in a non-randomized controlled
(89.8%) [44]. Both RET and AET improved self-esteem compared trial among 114 breast cancer survivors [59] (Table 3). Exercising
to UC, but no differences were found between AET and RET. Also for 6 months showed additional improvements in pulmonary func-
cancer-specific QoL, fatigue, anxiety, depression and arm volumes tion (forced vital capacity) and upper body muscle endurance
did not differ between groups. QoL and lower body strength were (bench press) compared with 3 months. No significant differences
higher, and body fat and triglycerides lower after 24 weeks of RET between the exercise groups were found in cardiovascular endur-
compared to UC in men receiving radiotherapy for prostate cancer ance (treadmill time) and abdominal muscle endurance (crunches),
[45]. No significant differences were found between AET and UC but both showed improvements compared to UC. No differences
(Table 3) [45]. Compared with UC, both RET and AET improved between groups were found for forced expiratory volume in one
upper body strength and peakVO2, and reduced fatigue. No signif- second, leg press, lateral pull-down, shoulder press, fatigue and
icant differences between groups were found for prostate-specific depression, except for affective fatigue, which improved more after
symptoms, body weight, body fat, hemoglobin, testosterone, and 6 months than after 3 months, but showed no differences com-
prostate-specific antigen. Although both RET and AET were com- pared to UC [59].
pared with UC, with RET showing longer-term improvements and
additional benefits for QoL, strength, triglycerides and body fat, di- Optimal timing of interventions
rect comparisons between RET and AET were not reported. To be Although studies have shown beneficial effects of exercise both
able to tailor exercise interventions to individual cancer-related se- during and after cancer treatment, little is known about the opti-
quelae, more studies evaluating the effects of different exercise mal timing of starting exercise interventions. A three-armed RCT
modes on specific outcomes are needed. Few studies are currently in 63 survivors of breast cancer scheduled for radiotherapy com-
underway [46–51] (Table 3). In addition to RET and AET, specific paring RET + AET during treatment with RET + AET post treatment
clinical exercises may be recommended for specific cancer diagno- and UC showed that both exercise groups reduced general and
ses. For example, for survivors of prostate cancer, pelvic floor train- physical fatigue, and improved muscle strength and endurance
ing before surgery and during radiation and ADT may be important compared to UC at 12 weeks and 3 months follow-up [60] (Table 3).
to reduce incontinence and to improve QoL [52,53]. In patients who exercised during treatment, improvements in glo-
RCTs directly comparing different exercise frequencies among bal health, physical functioning, role functioning and emotional
cancer survivors have not been published. The majority (59%) of functioning compared to UC were found at 12 weeks, but not at
studies evaluating effects of PA in cancer survivors focused on a 3 months follow-up. Improvements in physical fatigue at
frequency of 3–5 times per week, [9] corresponding with general 12 weeks, and improvements in upper body muscle strength at
PA guidelines. Interventions focusing on structured supervised 3 months follow-up were superior after exercising during treat-
exercise generally offered two sessions per week with or without ment compared to exercising post treatment, [60] indicating that
additional PA counseling. it may be more beneficial to start exercising during treatment.
Two studies compared responses to different exercise intensi- Two RCTs comparing early versus delayed onset of exercise are
ties [54,55] (Table 3). Comparisons of 10 weeks of moderate and currently being conducted [61,62] (Table 3). Although it may be
low intensity AET with UC in a small group (n = 18) of breast and expected that early initiation of exercise is better, patients may
colon cancer survivors after completion of treatment showed no experience specific barriers to exercise at different time points in
differences in peakVO2, lower-body flexibility and body fat be- the cancer trajectory, limiting them to comply with specific exer-
tween the two exercise groups [54]. Both exercise groups com- cise programs early in the trajectory. Starting later would then
bined showed increased peakVO2, lower-body flexibility, QoL, be more effective and efficient. Therefore, more insight into the
and energy, and decreased body fat compared to UC [54]. A optimal timing of interventions is warranted.
three-armed RCT among 73 women with breast cancer-related
lymphedema showed that 12 weeks of both high and low intensity Mediators of exercise effects
RET are safe and effective in reducing lymphatic relative volume,
improving upper and lower body muscle strength, 400 m walk test, Insight into mediators of exercise programs is important for
general fatigue, body image, and breast and arm symptoms [55]. identifying and subsequently targeting critical intervention com-
High intensity RET was superior to low intensity RET on the 400- ponents to improve effectiveness and efficiency and to reduce
m walk [55]. Three [56–58] RCTs comparing effects of different the costs [40,63,64]. Few studies examined mediators of exercise
exercise intensities are currently being conducted (Table 3). interventions in cancer survivors. In uncontrolled trials and a
L.M. Buffart et al. / Cancer Treatment Reviews 40 (2014) 327–340 335

Table 4
Cancer survivors’ interests and counseling and program preferences regarding PA and exercise.

Reference number [134] [135] [136] [137] [138] [139] [140] [141] [142] [143] [144] [145] [146] [147] Total
Characteristics
n 588 1284 307 106 386 397 90 50 192 90 483 345 703 431 5452
Country Can Nor Can USA Can Can USA Can USA USA USA Can Can Can
Diagnosis Mixed Mixed Mixed Brain Endometrial Bladder Breast Palliative Breast Head Breast Ovarian Kidney NHL Mean 
Neck (rural)
Months since diagnosis, mean (SD) 73.6 42.9 28; 6– 28; 6– 52 (32) 74 14.9 62.5 39.0 73.6 69.0 62 50
(52.1) (15.5) 18 178 (14.7) (25.9) (21.5) (52.6) (55.5) (25)
Age in years, mean (SD) 38.2 56.6 60.8 44.8 64.5 (10.6) 30% < 65 57.1 61.5 64 65% < 65 63 (12) 60.2 65.0 61 57
(5.6) (13.8) (11.5) (12.0) (12.0) (13.1) (11.5) (12.6) (11.1) (13)
Gender, % male 30 44 41.1 49.1 0 74.3 0 40 100 78 0 0 63 52 39

COUNSELLING
b c
Interest
Yes 51.7 55 60.3 29.2 55.7 47.5 44 51.7 51
No 19.5 24 15.9 37.7 22.6 29.6 24.8 22.9 23
Maybe 28.8 21 23.7 16 14.2 22.9 31.2 25.4 25
Source
Physician 17.2 12 9.8 18.7 20.5 4 5 11 9 21.9 16
Nurse 2.2 7 4.6 12.7 12.7 3 2 6 0.6 12.2 7
Exercise specialist from cancer 49.6 53 76.8 40.9 40.1 71 25 11 18 55.7 47
center
Exercise specialist from health 13.8 27 4.9 17.3 15.9 12 26 6 22 19.1 19
club or community center
Cancer patient/survivor 6.5 2 3.6 10.4 11 9 2 0 2 19.6 7
Delivery
Face-to-face 47.2 95 85.3 82.8 77.7 55 61 40 47 34 68
Telephone 11.2 0 1.7 3.4 3.5 3 0 0 0.8 6.1 3
(self-help) video 15.1 2 6.5 7 9 6 13.5 6
Print material 63.5 2 6.5 11.8 17.7 7 5 3 11 50 21
Internet 38.5 1 0 6 0 1 3 13.9 8
By e-mail 45.9 19.5 9
Other 1 2 30 3 3 2

PROGRAM
Interested
Yes 48 67 29 56 41.5 44.5 78 38 33 33 53.8 34.2 55.4 49
No 21.7 9 38 23 18.3 18.8 8 26 25 26 13.3 28.5 18.9 19
Maybe 30.3 24 16 14 35.3 36.6 14 36 42 41 32.9 37.3 25.7 31
Able to
Yes 62.2 74 30.2 65.1 46.4 47 58 64 51 55 64.6 48 52.9 59
No 12.5 6 32.1 8.5 18.3 15.7 8 14 17 15 6.6 18.9 15.5 13
Maybe 25.3 20 17 18.9 35.3 37.3 34 22 32 30 28.9 33.1 31.6 28
Preferred timing
At the time of diagnosis, before 25.8 10 32 18.1 21.4 17.9 17 27.7 19
treatment
During treatment 10.1 14 17 6 10.6 12.6 4.7 16.6 11
Immediately after treatment 16.6 46 24 26.8 39.1 25.5 22.4 29
At 3–6 months after treatment 33.5 26 23 39.3 21.4 25.8 36.5 38.2 30
At least 1 year after treatment 14.1 5 5 9.7 7.5 18.2 20.9 18.1 12
Company
Alone 16.7 17 44 23.8 35.8 21 54 41 50 41 29 39.1 30.8 29
With other cancer patients/ 16.9 26 34 4.7 4.7 11.7 6 25 0 15.9 20.6 12.7 16
survivors
Family/ friends/ spouse/ coworker 31.6 26 4 26.4 51.9 31.8 22 30 16 35 30 38 30.5 22.5 24
Location
Outside around my neighborhood 46.3 16 29 47 25 46.8 16
At home 43.5 20 40 25.5 43.4 32.7 53.7 37 84 36 33 38 48.9 52 42.6 38
At a community/local fitness 53.5 35 13 9.4 22.6 24.3 11.4 16 0 15 8 19 20.9 32.5 17.3 27
center
At a cancer center 11.2 23 19 10.2 7.8 33 0 6 0 3 6.9 7.3 12.6 12
Time of the day
Morning 34 48 51.3 36.6 40 52 47 53 48.9 58.3 41.3 45
Afternoon 32 23 12.3 15.6 32 13 17 11 15 25.9 17.4 22
Evening 16 5 12.9 11.4 4 20 15 20 17.4 30.3 18.7 18
* * * *
Mode
Walking 45.7 33 81 51 53 68.6 81.1 61 72 55.5 45.5 48.5 62.7 69.4 55
Cycling 16.6 14 4 19 16.1 24 7.5 4.5 8.5 3.3 23.8 12
Swimming 20.8 14 4 15 7.5 8.5 4.4 16.9 10
Resistance training 23 4 44 36 26.9 20.5 24 12 3 9.5 5 12
Intensity

(continued on next page)


336 L.M. Buffart et al. / Cancer Treatment Reviews 40 (2014) 327–340

Table 4 (continued)

Reference number [134] [135] [136] [137] [138] [139] [140] [141] [142] [143] [144] [145] [146] [147] Total
Light 11 35 26.3 26.1 45 22 26 26 35.3 23.1 23
Moderate 77 56 61.1 61.7 47 64 50 65 58.4 62 64
Vigorous 9 4 7.7 6.7 3 7 6 4 6.3 9.7 7
Variability
Same each time 34 51 40.9 49.1 22 27 35 28
Different each time 66 49 59.1 50.9 78 21 23 64.7 38.9 54
Supervision
Supervised 64 43 53.1 29.4 66 25 12 22 40.8 41.5 45
Unsupervised 36 57 46.9 70.6 34 49 55 47 59.2 58.5 49
Structure
Spontaneous/Flexible 21 47 35.5 56.9 46 66 62 49.2 45 41
Structured/scheduled 79 53 64.5 43.1 54 10 24 50.8 55 57

NHL = Non-Hodgkin lymphoma. aBefore diagnosis.


b
During treatment.
c
After treatment.
*
Summer and winter averaged.
 
weighted according to sample size. Proportions do not add up to 100 due to missing categories (e.g. no preference, other).

cross-sectional study, exercise effects on QoL were found to be Endocrine function


mediated by fatigue [65–67]. As another example, the effects of Insulin, IGF, and IGF binding proteins (IGFBP) are important reg-
an 8-week mixed modality community-based group exercise and ulators of energy metabolism and growth, and may also be in-
information support intervention on QoL of patients with breast volved in tumor development and progression. High insulin
cancer was mediated by exercise familiarity and self-efficacy but levels have been associated with increased risk of tumor recur-
not by social support [68]. rence or death in survivors of breast [95,96] and colon cancer
In addition to personal and psychosocial working mechanisms, [97,98]. In addition, increasing evidence suggests that insulin, IGF
biological factors (e.g. function of immune and endocrine systems) and IGFBP may have an important mediating role in the effect of
and physiological factors (e.g. neuromuscular function) may medi- exercise on cancer risk and prognosis.
ate the effect of PA and exercise on relevant health outcomes, Recently, Ballard-Barbash et al. [16] systematically reviewed
including fatigue, QoL and survival. studies among cancer survivors examining associations between
PA and biomarkers. One observational study [77] and four RCTs
[99–102] evaluated effects of PA on insulin, IGF and IGFBP among
Immune system function breast cancer survivors [16]. Higher levels of PA were found to be
Cancer and its treatments may alter immune system function associated with lower levels of C-peptide (marker of insulin secre-
[69]. C-reactive protein (CRP) is a sensitive marker of low-grade tion) and leptin, and higher levels of IGF-1, but no association was
systemic inflammation [70]. In cancer survivors, elevated concen- found for IGFBP-III [77]. Results from RCTs provide preliminary evi-
trations of CRP have been found to be associated with fatigue dence that PA may result in beneficial changes in the circulating
[71] and reduced overall and disease-free survival [72]. Exercise levels of insulin and insulin-related pathways, which may be more
can have a beneficial effect on the immune system, [73–76] includ- pronounced for obese or sedentary women who generally have
ing cancer survivors [16,69]. Reductions in CRP after exercise [16] higher serum insulin levels at baseline, and for women who are
were found in an observational study among breast cancer survi- not taking Tamoxifen [16]. Weighted-mean effect size of the effect
vors [77] and RCTs among survivors of breast, [78] lung, [79] and of post-treatment PA on IGF-1 was found to be significant but
prostate cancer [80]. small-to-moderate [9]. No changes were reported for insulin and
Pro-inflammatory cytokines (e.g. tumor necrosis factor-alpha IGFBP-III, and evidence was insufficient for IGF-II and IGFBP-I [9].
(TNF-a) and Interleukin (IL)-6) may be released in response to The number of studies on the effects of PA on insulin, IGF and
the tumor or cancer treatment, and may promote tumor growth IGFBPs during treatment was too small to draw conclusions [9].
and angiogenesis [81–83]. TNF-a and IL-6 have been associated Three RCTs in prostate cancer survivors on androgen depriva-
with cancer-related anemia, [83] cachexia, [84] loss of muscle tion therapy (ADT) [45,80,103] showed consistent evidence that
mass, [84,85] and cancer-related fatigue [86,87]. PA and exercise exercise does not alter prostate-specific antigen (PSA) or testoster-
may have an anti-inflammatory effect in cancer survivors, [88] as one levels, [16] however, the effect on survivors not on ADT is un-
cytokines may be expressed and released by muscle fibers [89– clear. Studies on the effect of PA on biomarkers, such as serotonin,
92]. However, no change in IL-6 levels after exercise was found cortisol, bilirubin [93] or others are limited or nonexistent [16].
in 20 patients with breast cancer [93]. Comparably, a study in 10 Future studies among cancer survivors should further explore
patients with prostate cancer did not find changes in resting serum the effects of PA on biomarkers of endocrine function and their
concentrations of IL-6 and TNF-a after a 20-week resistance exer- mediating role in the association between PA and health outcomes.
cise program, but increased IL-6 levels were found after acute
resistance exercise [94]. Exercise improved Natural Killer cell cyto- Neuromuscular function
toxity among cancer survivors, [16,69] indicating improved im- Cancer and its treatment may affect neuromuscular function.
mune function. Other beneficial effects included increased Peripheral neuropathy has emerged as a side effect of several che-
lymphocyte proliferation and granulocyte cell counts. Although motherapeutic agents including thalidomide, [104,105] paclitaxel
evidence is still preliminary, PA and exercise may be associated [106,107], and oxaliplatin [108–110]. Cancer treatment may also
with beneficial changes in inflammation and immunity [16]. Fu- damage the muscle itself. Several case reports describe
ture studies among cancer survivors should examine immune sys- chemotherapy-induced myopathy or myopathy-related disorders
tem responses to exercise and their mediating role on reducing among cancer survivors [111–113]. Deconditioning, neuropathy
fatigue, and improving QoL and survival. and myopathy may lead to PA avoidance and consequently to
L.M. Buffart et al. / Cancer Treatment Reviews 40 (2014) 327–340 337

muscle atrophy. Sarcopenia (age-related loss of skeletal muscle preferences depend on age, current PA level, employment status,
mass and strength) is present in many cancer survivors, and may education, income, the presence of overweight or obesity, and
be associated with poor outcomes including increased fatigue, re- medical variables including the type and stage of cancer, time since
duced QoL, and higher mortality risk. Sarcopenia has been associ- diagnosis, type of treatment, and the presence of comorbidities
ated with time to tumor progression and overall survival after [134–139,141–147]. Also self-efficacy, enjoyment, social support,
breast cancer [114–115]. Also, androgen deprivation therapy perceived barriers, and the presence of depressive symptoms and
(ADT), which is used as adjuvant therapy for patients with prostate fatigue determine exercise preferences [142,144]. For optimal par-
cancer, may result in toxicity-related musculoskeletal deficits, ticipation in and adherence to PA and exercise programs, it is nec-
including loss of skeletal muscle and strength, reducing physical essary that they fit with patients’ interests, abilities, opportunities,
function and QoL[116–119]. and preferences while being aware of the therapy intent of the
Exercise may improve muscle strength [9] and reverse muscle exercise prescription.
loss, [116] thereby reducing the acceleration of sarcopenia. Com-
bined resistance and aerobic exercise has been demonstrated to Conclusions
improve muscle mass and strength in patients undergoing ADT
for prostate cancer [80,120]. Further knowledge on the detrimental Current PA guidelines for cancer survivors are generic. It is nec-
effects of various types of cancer treatment on muscle mass and essary to move from a one-size fits all approach to specific PA
neuromuscular function, and potential counteractive effects of PA guidelines tailored to the characteristics, needs, capabilities and
and exercise is necessary. The use of voluntary and electrically preferences of individual patients. More research is needed to be
stimulated contractions may be helpful in identifying these able to develop specific guidelines for a given exercise prescription
changes in contractile muscle properties (e.g., force production (e.g. mode, frequency, intensity, duration), for a given cancer site at
and rate of force development and relaxation) [121]. a particular phase of the cancer trajectory, disease impact and
treatment side-effects, and for specific outcomes. These studies
How to motivate cancer survivors to exercise? should focus on identifying clinical, personal, physical, psychoso-
cial, and intervention moderators explaining ‘which’ interventions
Traditionally, exercise trials were designed as efficacy trials to work ‘for whom’ and ‘under what circumstances.’ It is time to
examine whether the intervention produced the intended effect move towards second generation studies that compare different
on health outcomes under ideal circumstances [122]. The exercise modes, frequencies, intensities, duration and timing.
effectiveness, which refers to whether an intervention works in Further, more insight into the working mechanisms of exercise
‘‘real-world’’ conditions, largely depends on the motivation and interventions on health outcomes in cancer survivors is needed to
adherence of patients [122]. To improve the effectiveness of exer- improve the efficacy and efficiency of interventions. Future studies
cise, it is essential to improve the uptake, dissemination and main- should identify the mediating role of personal, psychosocial,
tenance of PA and exercise interventions. Common barriers to PA immunological, endocrine, and physiological factors in the exercise
among cancer survivors include physical discomfort, feeling sick, intervention effects on health outcomes among cancer survivors.
having low mood, feelings of self-consciousness relating to physi- For optimal uptake of PA and exercise interventions, it is necessary
cal appearance, fatigue, fear of overdoing it, and being too busy that existing programs embrace interests and preferences of indi-
[123–128]. Also, socio-demographic and clinical variables, smok- vidual patients or patient groups.
ing, alcohol consumption, obesity as well as attitude, self-efficacy,
and intention may play a role [129–131]. Specific PA barriers may
Conflict of interest statement
vary between patient groups [128] and exercise modes. To be able
to increase adoption and maintenance of a physically active life-
All authors have declared no conflicts of interest.
style, we need to obtain insight into barriers to specific modes of
exercise, to develop strategies that help patients overcome these
barriers, and to identify the most effective formats of delivering Authorship
PA and exercise interventions (e.g. home-based, supervised, hospi-
tal-based, internet-based) and whether or not this should be com- All authors have made a substantial contribution to the concep-
bined with nutrition and/or psychosocial support interventions. tion and design of the study, drafting or critically revising the man-
Incorporation of behavior change techniques may further assist uscript, and approved the version to be submitted.
with PA adoption and maintenance [122]. Development of inter-
ventions to improve PA behavior – i.e. behavior change trials-, Acknowledgements
should be theory-based incorporating a clearly defined set of
behavioral change techniques [132,133]. The contribution of LMB was funded by a ‘Bas Mulder’ grant of
PA participation and adherence can also be influenced by pa- the Alpe d’HuZes foundation, which is part of the Dutch Cancer
tients’ preferences. Previous studies have investigated patients’ Society, a fellowship granted by the EMGO Institute for Health
interests in PA and exercise and their preferences regarding exer- and Care Research, and a travel grant funded by the Edith Cowan
cise programs in various groups of cancer survivors [134,147] (Ta- University.
ble 4). A summary of these studies showed that most patients were The contribution of DAG was funded by a Movember New
interested in receiving PA information, preferably by an exercise Directions Development Award obtained through Prostate Cancer
specialist from a cancer center, and face-to-face (Table 4). Most pa- Foundation of Australia’s Research Program. All sponsors had no
tients were interested in an exercise program, feel able to partici- involvement in the content of the manuscript and in the decision
pate, prefer to start exercising after completion of therapy, at to submit the manuscript for publication.
moderate intensity, alone, at home, and in the morning. Walking
is the preferred type of exercise for most patients, both in summer References
and in winter. Some patients prefer a different activity each ses-
sion, whereas others prefer the same activity each time. Most pa- [1] Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer
statistics. CA Cancer J Clin 2011;61:69–90.
tients prefer unsupervised exercise with a flexible structure [2] Australian Institute of Health and Welfare. Cancer survival and prevalence in
(Table 4). However, patients’ interests, counseling and program Australia. Cat no CAN 38, 2008, Canberra.
338 L.M. Buffart et al. / Cancer Treatment Reviews 40 (2014) 327–340

[3] IKCnet. Dutch Cancer Registration. Survival. <http://www.ikcnet.nl>; 2012. [32] Steins Bisschop CN, Velthuis MJ, Wittink HM, Kuiper K, Takken T, van der
[4] Courneya KS. Exercise in cancer survivors: an overview of research. Med Sci Meulen WJTM, et al. Cardiopulmonary exercise testing in cancer
Sports Exerc 2003;35:1846–52. rehabilitation. A systematic review. Sports Med 2012;42:367–79.
[5] Curt GA, Breitbart W, Cella D, Groopman JE, Horning SJ, Itri LM, et al. Impact of [33] De Backer I, Schep G, Hoogeveen A, Vreugdenhil G, Kester AD, van Breda E.
cancer-related fatigue on the lives of patients: new findings from the Fatigue Exercise testing and training in a cancer rehabilitation program: the
Coalition. Oncologist 2000;5:353–60. advantage of the steep ramp test. Arch Phys Med Rehabil 2007;88:610–6.
[6] Centers for Disease Control and Prevention (CDC). Cancer survivors – United [34] Rock CL, Doyle C, Demark-Wahnefried W, Meyerhardt J, Courneya KS,
States, 2007. MMWR Morb Mortal Wkly Rep 2011;60:269–72. Schwartz AL, et al. Nutrition and physical activity guidelines for cancer
[7] Caspersen CJ, Powell KE, Christenson GM. Physical activity, exercise, and survivors. CA Cancer J Clin 2012;62:242–74.
physical fitness: definitions and distinctions for health-related research. [35] Campbell A, Stevinson C, Crank H. The BASES expert statement on exercise
Public Health Rep 1985;100:126–31. and cancer survivorship. J Sports Sci 2012;30:949–52.
[8] Fong JYT, Ho JWC, Hui BPH, Lee AM, Macfarlane DJ, Leung SSK, et al. Physical [36] Bull FC, Expert Working Groups. Physical activity guidelines in the U.K.:
activity for cancer survivors: a meta-analysis of randomised controlled trials. review and recommendations. Loughborough: School of Sport, Exercise and
BMJ 2012;344:e70. Health Sciences; 2010.
[9] Speck RM, Courneya KS, Masse LC, Duval S, Schmitz KH. An update of [37] Courneya KS, Friedenreich CM. Physical activity and cancer control. Semin
controlled physical activity trials in cancer survivors: a systematic review and Oncol Nurs 2007;23:242–52.
meta-analysis. J Cancer Surviv 2010;4:87–100. [38] Cramp F, Daniel J. Exercise for the management of cancer-related fatigue in
[10] Jones LW, Liang Y, Pituskin EN, Battaglini CL, Scott JM, Hornsby WE, et al. adults. Cochr Database Syst Rev 2008:CD006145.
Effect of exercise training on peak oxygen consumption in patients with [39] Duijts SF, Faber MM, Oldenburg HS, van Beurden M, Aaronson NK.
cancer: a meta-analysis. Oncologist 2011;16:112–20. Effectiveness of behavioral techniques and physical exercise on
[11] Cramp F, Byron-Daniel J. Exercise for the management of cancer-related psychosocial functioning and health-related quality of life in breast cancer
fatigue in adults. Cochr Database Syst Rev 2012;11:CD006145. patients and survivors – a meta-analysis. Psychooncology 2011;20:115–26.
[12] Velthuis MJ, Gasi-Idenburg SC, Aufdemkampe G, Wittink HM. The effect of [40] Kraemer HC, Wilson GT, Fairburn CG, Agras WS. Mediators and moderators of
physical exercise on cancer-related fatigue during cancer treatment: a meta- treatment effects in randomized clinical trials. Arch Gen Psychiatry
analysis of randomised controlled trials. Clin Oncol (R Coll Radiol) 2002;59:877–83.
2010;22:208–21. [41] Courneya KS, Sellar CM, Stevinson C, McNeely ML, Friedenreich CM, Peddle CJ,
[13] Craft LL, Vaniterson EH, Helenowski IB, Rademaker AW, Courneya KS. et al. Moderator effects in a randomized controlled trial of exercise training in
Exercise effects on depressive symptoms in cancer survivors: a systematic lymphoma patients. Cancer Epidemiol Biomarkers Prev 2009;18:2600–7.
review and meta-analysis. Cancer Epidemiol Biomarkers Prev 2012;21:3–19. [42] Courneya KS, McKenzie DC, Mackey JR, Gelmon K, Reid RD, Friedenreich CM,
[14] Mishra SI, Schrerer RW, Snyder C, Geigle PM, Berlanstein DR, Topaloglu O. et al. Moderators of the effects of exercise training in breast cancer patients
Exercise interventions on health-related quality of life for people with cancer receiving chemotherapy: a randomized controlled trial. Cancer
during active treatment. Cochr Database Syst Rev 2012:CD008465. 2008;112:1845–53.
[15] Mishra SI, Schrerer RW, Geigle PM, Berlanstein DR, Topaloglu O, Gotay CC, [43] Carmack Taylor CL, de Moor C, Basen-Engquist K, Smith MA, Dunn AL, Badr H,
et al. Exercise interventions on health-related quality of life for cancer et al. Moderator analyses of participants in the active for life after cancer trial:
survivors. Cochr Database Syst Rev 2012:CD007566. implications for physical activity group intervention studies. Ann Behav Med
[16] Ballard-Barbash R, Friedenreich C, Courneya KS, Siddiqi SM, McTiernan A, 2007;30:115–29.
Alfano CM. Physical activity, biomarkers, and disease outcomes in cancer [44] Courneya KS, Segal RJ, Mackey JR, Gelmon K, Reid RD, Friedenreich CM, et al.
survivors: a systematic review. J Natl Cancer Inst 2012;104:815–40. Effects of aerobic and resistance exercise in breast cancer patients receiving
[17] Holmes MD, Chen WY, Feskanich D, Kroenke CH, Colditz GA. Physical activity adjuvant chemotherapy: a multicenter randomized controlled trial. J Clin
and survival after breast cancer diagnosis. JAMA 2005;293:2479–86. Oncol 2007;25:4396–404.
[18] Irwin ML, McTiernan A, Manson JE, Thomson CA, Sternfeld B, Stefanick ML, [45] Segal RJ, Reid RD, Courneya KS, Sigal RJ, Kenny GP, Prud’Homme DG, et al.
et al. Physical activity and survival in postmenopausal women with breast Randomized controlled trial of resistance or aerobic exercise in men receiving
cancer: results from the Women’s Health Initiative. Cancer Prev Res radiation therapy for prostate cancer. J Clin Oncol 2009;27:344–51.
2011;4:522–9. [46] Jones LW, Eves ND, Kraus WE, Potti A, Crawford J, Blumenthal JA, et al. The
[19] Schmitz KH. Exercise for secondary prevention of breast cancer: moving from lung cancer exercise training study: a randomized trial of aerobic training,
evidence to changing clinical practice. Cancer Prev Res 2011;4:476–80. resistance training, or both in postsurgical lung cancer patients: rationale and
[20] Meyerhardt JA, Giovannucci EL, Holmes MD, Chan AT, Chan JA, Colditz GA, design. BMC Cancer 2010;10:155.
et al. Physical activity and survival after colorectal cancer diagnosis. J Clin [47] National Institutes of Health Clinical Center. Effect of Tai Chi vs. structured
Oncol 2006;24:3527–34. exercise on physical fitness and stress in cancer survivors. ClinicalTrials.gov.
[21] Meyerhardt JA, Giovannucci EL, Ogino S, Kirkner GJ, Chan AT, Willett W, et al. NCT00246818. <www.clinicaltrials.gov>; 2011.
Physical activity and male colorectal cancer survival. Arch Intern Med [48] Newton RU, Taaffe DR, Spry N, Gardener RA, Levin G, Wall B, et al. A phase III
2009;169:2102–8. clinical trial of exercise modalities on treatment side-effects in men receiving
[22] Kenfield SA, Stampfer MJ, Giovannucci EL, Chan JM. Physical activity and therapy for prostate cancer. BMC Cancer 2009;9:210.
survival after prostate cancer diagnosis in the health professionals follow-up [49] Siew Yim L. Randomised controlled trial of physical activity with breast
study. J Clin Oncol 2011;29:726–32. cancer survivors. Australian New Zealand Clinical Trials Registry.
[23] Richman EL, Kenfield SA, Stampfer MJ, Paciorek A, Carroll PR, Chan JM. ACTRN12611000093987. <www.anzctr.org>; 2011.
Physical activity after diagnosis and risk of prostate cancer progression: data [50] Winters-Stone K. Comparison of aerobic and resistance exercise in older
from the cancer of the prostate strategic urologic research endeavor. Cancer breast cancer survivors. ClinicalTrials.gov. NCT00662103. <http://
Res 2011;71:3889–95. www.clinicaltrials.gov>; 2012.
[24] Courneya KS, Booth CM, Gill S, O’Brien P, Vardy J, Friedenreich CM, et al. The [51] Young-McCaughan S. Comparing aerobic to resistance training in recovery
colon health and life-long exercise change trial: a randomized trial of the from cancer. ClinicalTrials.gov. NCT00237926. <www.clinicaltrials.gov>;
national cancer institute of Canada clinical trials group. Curr Oncol 2008.
2008;15:271–8. [52] Baumann FT, Zopf EM, Bloch W. Clinical exercise interventions in prostate
[25] Brown JK, Byers T, Doyle C, Courneya KS, Demark-Wahnefried W, Kushi LH, cancer patients – a systematic review of randomized controlled trials.
et al. Nutrition and physical activity during and after cancer treatment: an Support Care Cancer 2012;20:221–33.
American cancer society guide for informed choices. CA Cancer J Clin [53] Van Kampen M, De Weerdt W, Van Poppel H, De Ridder D, Feys H, Baert L.
2003;53:268–91. Effect of pelvic-floor re-education on duration and degree of incontinence
[26] Doyle C, Kushi LH, Byers T, Courneya KS, Mark-Wahnefried W, Grant B, et al. after radical prostatectomy: a randomised controlled trial. Lancet
Nutrition and physical activity during and after cancer treatment: an 2000;355:98–102.
American cancer society guide for informed choices. CA Cancer J Clin [54] Burnham TR, Wilcox A. Effects of exercise on physiological and psychological
2006;56:323–53. variables in cancer survivors. Med Sci Sports Exerc 2002;34:1863–7.
[27] Hayes SC, Spence RR, Galvao DA, Newton RU. Australian association for [55] Gibbs Z. Exercise for breast cancer patietns with lymphedema. Australian
exercise and sport science position stand: optimising cancer outcomes New Zealand Clinical Trials Registry. ACTRN12612000256875.
through exercise. J Sci Med Sport 2009;12:428–34. <www.anzctr.org.au>; 2012.
[28] Schmitz KH, Courneya KS, Matthews C, Mark-Wahnefried W, Galvao DA, [56] Cormie P. Exercise intervention trial for women with breast cancer related
Pinto BM, et al. American college of sports medicine roundtable on exercise lymphoedema. Australian New Zealand Clinical Trials Registry.
guidelines for cancer survivors. Med Sci Sports Exerc 2010;42:1409–26. ACTRN12610000788077. <www.anzctr.org.au>; 2012.
[29] Physical Activities Guidelines Advisory Committee. Physical activity [57] Kampshoff CS, Buffart LM, Schep G, van Mechelen W, Brug J, Chinapaw MJ.
guidelines advisory committee report. Washington (DC): US Department of Design of the Resistance and Endurance exercise After ChemoTherapy
Health and Human Services; 2008. (REACT) study: a randomized controlled trial to evaluate the effectiveness
[30] Comprehensive Cancer Center the Netherlands. Guideline cancer and cost-effectiveness of exercise interventions after chemotherapy on
rehabilitation. <http://www.oncoline.nl/oncologische-revalidatie>; 2011. physical fitness and fatigue. BMC Cancer 2010;10:658.
[31] van den Berg JP, Velthuis MJ, Gijssen BCM, Lindeman E, van der Pol MA, Hillen [58] Jones LW, Douglas PS, Eves ND, Marcom K, Kraus WE, Herndon JE, et al.
HFP. Richtlijn ‘Oncologische revalidatie’ [guideline cancer rehabilitation]. Ned Rationale and design of the Exercise Intensity Trial (EXCITE): a randomized
Tijdschr Geneeskd 2011;155. trial comparing the effects of moderate versus moderate to high-intensity
L.M. Buffart et al. / Cancer Treatment Reviews 40 (2014) 327–340 339

aerobic training in women with operable breast cancer. BMC Cancer [85] van Hall G, Steensberg A, Fischer C, Keller C, Moller K, Moseley P, et al.
2010;10:531. Interleukin-6 markedly decreases skeletal muscle protein turnover and
[59] Sprod LK, Hsieh CC, Hayward R, Schneider CM. Three versus six months of increases nonmuscle amino acid utilization in healthy individuals. J Clin
exercise training in breast cancer survivors. Breast Cancer Res Treat Endocrinol Metab 2008;93:2851–8.
2010;121:413–9. [86] Bower JE. Cancer-related fatigue: links with inflammation in cancer patients
[60] Gibbs Z. Fatigue and changes to muscle strength and physical function in and survivors. Brain Behav Immun 2007;21:863–71.
delayed versus immediate exercise for breast cancer survivors scheduled for [87] Schubert C, Hong S, Natarajan L, Mills PJ, Dimsdale JE. The association
radiation therapy: a randomised controlled trial. Australian New Zealand between fatigue and inflammatory marker levels in cancer patients: a
Clinical Trials Registry. ACTRN12612000646842. <www.anzctr.org.au>; 2012. quantitative review. Brain Behav Immun 2007;21:413–27.
[61] Newton RU, Taaffe DR, Spry N, Cormie P, Chambers S, Gardener RA, et al. Can [88] Seruga B, Zhang H, Bernstein LJ, Tannock IF. Cytokines and their relationship
exercise ameliorate treatment toxicity during the initial phase of testosterone to the symptoms and outcome of cancer. Nat Rev Cancer 2008;8:887–99.
deprivation in prostate cancer patients? Is this more effective than delayed [89] Pedersen BK, Akerstrom TC, Nielsen AR, Fischer CP. Role of myokines in
rehabilitation? BMC Cancer 2012;12:432. exercise and metabolism. J Appl Physiol 2007;103:1093–8.
[62] Walenkamp A, Gietema JA. Optimal timing of physical activity in cancer [90] Pedersen BK, Febbraio M. Muscles, exercise and obesity: skeletal muscle
treatment (ACT). ClinicalTrials.gov. NCT01642680. <www.clinicaltrials.gov>; organ as a secretary organ. Nat Rev Endocrinol 2012;8:457–65.
2012. [91] Petersen AM, Pedersen BK. The role of IL-6 in mediating the anti-
[63] Hafeman DM, Schwartz S. Opening the Black Box: a motivation for the inflammatory effects of exercise. J Physiol Pharmacol 2006;57(Suppl.
assessment of mediation. Int J Epidemiol 2009;38:838–45. 10):43–51.
[64] MacKinnon DP, Luecken LJ. How and for whom? Mediation and moderation [92] Pedersen BK. Exercise-induced myokines and their role in chronic diseases.
in health psychology. Health Psychol 2008;27:S99–S100. Brain Behav Immun 2011;25:811–6.
[65] Schwartz AL. Fatigue mediates the effects of exercise on quality of life. Qual [93] Payne JK, Held J, Thorpe J, Shaw H. Effect of exercise on biomarkers, fatigue,
Life Res 1999;8:529–38. sleep disturbances, and depressive symptoms in older women with breast
[66] Buffart LM, De Backer IC, Schep G, Vreugdenhil A, Brug J, Chinapaw MJ. cancer receiving hormonal therapy. Oncol Nurs Forum 2008;35:635–42.
Fatigue mediates the relationship between physical fitness and quality of life [94] Galvao DA, Nosaka K, Taaffe DR, Peake J, Spry N, Suzuki K, et al. Endocrine and
in cancer survivors. J Sci Med Sport 2013;16:99–104. immune responses to resistance training in prostate cancer patients. Prostate
[67] Buffart LM, Thong MS, Schep G, Chinapaw MJ, Brug J, van de Poll-Franse LV. Cancer Prostatic Dis 2008;11:160–5.
Self-reported physical activity: its correlates and relationship with health- [95] Goodwin P, Ennis M, Pritchard KI, Trudeau ME, Koo J, Madarnas Y, et al.
related quality of life in a large cohort of colorectal cancer survivors. PLoS One Fasting insulin and outcome in early-stage breast cancer: results of a
2012;7:e36164. prospective cohort study. J Clin Oncol 2002;20:42–51.
[68] Sherman KA, Heard G, Cavanagh KL. Psychological effects and mediators of a [96] Irwin ML, Duggan C, Wang CY, Smith AW, McTiernan A, Baumgartner RN,
group multi-component program for breast cancer survivors. J Behav Med et al. Fasting C-peptide levels and death resulting from all causes and breast
2010;33:378–91. cancer: the health, eating, activity and lifestyle study. J Clin Oncol
[69] Fairey AS, Courneya KS, Field CJ, Mackey JR. Physical exercise and immune 2011;29:47–53.
system function in cancer survivors: a comprehensive review and future [97] Haydon AMM, MacInnis RJ, English DR, Morris H, Giles GG. Physical activity,
directions. Cancer 2002;94:539–51. insulin-like growth factor 1, insulin-like growth factor binding protein 3, and
[70] Pearson TA, Mensah GA, Alexander RW, Anderson JL, Cannon III RO, Criqui M, survival from colorectal cancer. Gut 2006;55:689–94.
et al. Markers of inflammation and cardiovascular disease: application to [98] Wolpin BM, Meyerhardt JA, Chan AT, Ng K, Chan JA, Wu K, et al. Insulin, the
clinical and public health practice: a statement for healthcare professionals insulin-like growth factor axis, and mortality in patients with nonmetastatic
from the centers for disease control and prevention and the American heart colorectal cancer. J Clin Oncol 2009;27:176–85.
association. Circulation 2003;107:499–511. [99] Fairey AS, Courneya KS, Field CJ, Bell GJ, Jones LW, Mackey JR. Effects of
[71] Wratten C, Kilmurray J, Nash S, Seldon M, Hamilton CS, O’Brien PC, et al. exercise training on fasting insulin, insulin resistance, insulin-like growth
Fatigue during breast radiotherapy and its relationship to biological factors. factors, and insulin-like growth factor binding proteins in postmenopausal
Int J Radiat Oncol Biol Phys 2004;59:160–7. breast cancer survivors: a randomized controlled trial. Cancer Epidemiol
[72] Pierce BL, Ballard-Barbash R, Bernstein L, Baumgartner RN, Neuhouser ML, Biomarkers Prev 2003;12:721–7.
Wener MH, et al. Elevated biomarkers of inflammation are associated with [100] Irwin ML, Varma K, varez-Reeves M, Cadmus L, Wiley A, Chung GG, et al.
reduced survival among breast cancer patients. J Clin Oncol Randomized controlled trial of aerobic exercise on insulin and insulin-like
2009;27:3437–44. growth factors in breast cancer survivors: the Yale exercise and survivorship
[73] Chinapaw MJ, de JN, Pallast EG, Kloek GC, Schouten EG, Kok FJ. Immunity in study. Cancer Epidemiol Biomarkers Prev 2009;18:306–13.
frail elderly: a randomized controlled trial of exercise and enriched foods. [101] Ligibel JA, Campbell N, Patridge A, Chen WY, Salinardi T, Chen H, et al. Impact
Med Sci Sports Exerc 2000;32:2005–11. of a mixed strength and endurance exercise intervention on insulin levels in
[74] Mathur N, Pedersen BK. Exercise as a mean to control low-grade systemic breast cancer survivors. J Clin Oncol 2008;26:907–12.
inflammation. Mediators Inflamm 2008;2008:109502. [102] Schmitz KH, Ahmed RL, Hannan PJ, Yee D. Safety and efficacy of weight
[75] Kadoglou NP, Iliadis F, Angelopoulou N, Perrea D, Ampatzidis G, Liapis CD, training in recent breast cancer survivors to alter body composition, insulin
et al. The anti-inflammatory effects of exercise training in patients with type and insulin-like growth factor axis proteins. Cancer Epidemiol Biomarkers
2 diabetes mellitus. Eur J Cardiovasc Prev Rehabil 2007;14:837–43. Prev 2005;14:1672–80.
[76] Lakka TA, Lakka HM, Rankinen T, Leon AS, Rao DC, Skinner JS, et al. Effect of [103] Segal RJ, Reid RD, Courneya KS, Malone SC, Parliament MB, Scott CG, et al.
exercise training on plasma levels of C-reactive protein in healthy adults: the Resistance exercise in men receiving androgen deprivation therapy for
HERITAGE family study. Eur Heart J 2005;26:2018–25. prostate cancer. J Clin Oncol 2003;21:1653–9.
[77] Irwin ML, McTiernan A, Bernstein L, Gilliland FD, Baumgartner R, [104] Kocer B, Sucak G, Kuruoglu R, Aki Z, Haznedar R, Erdogmus NI. Clinical and
Baumgartner K, et al. Relationship of obesity and physical activity with C- electrophysiological evaluation of patients with thalidomide-induced
peptide, leptin, and insulin-like growth factors in breast cancer survivors. neuropathy. Acta Neurol Belg 2009;109:120–6.
Cancer Epidemiol Biomarkers Prev 2005;14:2881–8. [105] Molloy FM, Floeter MK, Syed NA, Sandbrink F, Culcea E, Steinberg SM, et al.
[78] Fairey AS, Courneya KS, Field CJ, Bell GJ, Jones LW, Martin BS, et al. Effect of Thalidomide neuropathy in patients treated for metastatic prostate cancer.
exercise training on C-reactive protein in postmenopausal breast cancer Muscle Nerve 2001;24:1050–7.
survivors: a randomized controlled trial. Brain Behav Immun 2005;19: [106] Openshaw H, Beamon K, Synold TW, Longmate J, Slatkin NE, Doroshow JH,
381–8. et al. Neurophysiological study of peripheral neuropathy after high-dose
[79] Jones LW, Eves ND, Peddle CJ, Courneya KS, Haykowsky M, Kumar V, et al. paclitaxel: lack of neuroprotective effect of amifostine. Clin Cancer Res
Effects of presurgical exercise training on systemic inflammatory markers 2004;10:461–7.
among patients with malignant lung lesions. Appl Physiol Nutr Metab [107] Openshaw H, Beamon K, Longmate J, Synold T, Slatkin NE, Somlo G. The effect
2009;34:197–202. of height on paclitaxel nerve damage. J Neurooncol 2005;74:207–10.
[80] Galvao DA, Taaffe DR, Spry N, Joseph D, Newton RU. Combined resistance and [108] Lehky TJ, Leonard GD, Wilson RH, Grem JL, Floeter MK. Oxaliplatin-induced
aerobic exercise program reverses muscle loss in men undergoing androgen neurotoxicity: acute hyperexcitability and chronic neuropathy. Muscle Nerve
suppression therapy for prostate cancer without bone metastases: a 2004;29:387–92.
randomized controlled trial. J Clin Oncol 2010;28:340–7. [109] Leonard GD, Wright MA, Quinn MG, Fioravanti S, Harold N, Schuler B, et al.
[81] Bower JE, Ganz PA, Tao ML, Hu W, Belin TR, Sepah S, et al. Inflammatory Survey of oxaliplatin-associated neurotoxicity using an interview-based
biomarkers and fatigue during radiation therapy for breast and prostate questionnaire in patients with metastatic colorectal cancer. BMC Cancer
cancer. Clin Cancer Res 2009;15:5534–40. 2005;5:116.
[82] Lee CC, Liu KJ, Wu YC, Huang TS. Tumor necrosis factor-a Interleukin-8 and [110] Wilson RH, Lehky T, Thomas RR, Quinn MG, Floeter MK, Grem JL. Acute
Interleukin-6 are involved in vascular endothelial cell capillary tube and oxaliplatin-induced peripheral nerve hyperexcitability. J Clin Oncol
network formation induced by tumor-associated macrophages. J Cancer Mol 2002;20:1767–74.
2006;2:155–60. [111] Anderlini P, Buzaid AC, Legha SS. Acute rhabdomyolysis after concurrent
[83] Buck I, Morceau F, Grigorakaki C, Dicato M, Diederich M. Linking anemia to administration of interleukin-2, interferon-alfa, and chemotherapy for
inflammation and cancer: the crucial role of TNFalpha. Biochem Pharmacol metastatic melanoma. Cancer 1995;76:678–9.
2009;77:1572–9. [112] Rini BI, Gajewski TF. Polymyositis with respiratory muscle weakness
[84] Hong DS, Angelo LS, Kurzrock R. Interleukin-6 and its receptor in cancer: requiring mechanical ventilation in a patient with metastatic thymoma
implications for translational therapeutics. Cancer 2007;110:1911–28. treated with octreotide. Ann Oncol 1999;10:973–9.
340 L.M. Buffart et al. / Cancer Treatment Reviews 40 (2014) 327–340

[113] Scruggs ER, Rks Naylor AJ. Mechanisms of zidovudine-induced mitochondrial [131] Trinh L, Plotnikoff RC, Rhodes RE, North S, Courneya KS. Correlates of physical
toxicity and myopathy. Pharmacology 2008;82:83–8. activity in population-bases sample of kidney cancer survivors: an
[114] Prado CM, Baracos VE, McCargar LJ, Reiman T, Mourtzakis M, Tonkin K, et al. application of the theory of planned behavior. Int J Behav Nutr Phys Act
Sarcopenia as a determinant of chemotherapy toxicity and time to tumor 2012;9:96.
progression in metastatic breast cancer patients receiving capecitabine [132] Abraham C, Michie S. A taxonomy of behavior change techniques used in
treatment. Clin Cancer Res 2009;15:2920–6. interventions. Health Psychol 2008;27:379–87.
[115] Villaseñor A, Ballard-Barbash R, Baumgartner K, Baumgartner R, Bernstein L, [133] Michie S, Ashford S, Sniehotta FF, Dombrowski SU, Bishop A, French DP. A
McTiernan A, et al. Prevalence and prognostic effect of sarcopenia in breast refined taxonomy of behaviour change techniques to help people change
cancer survivors: the HEAL study. J Cancer Surviv 2012;6:398–406. their physical activity and healthy eating behaviours: the CALO-RE
[116] Galvao DA, Taaffe DR, Spry N, Newton RU. Exercise can prevent and even taxonomy. Psychol Health 2011;26:1479–98.
reverse adverse effects of androgen suppression treatment in men with [134] Belanger LJ, Plotnikoff RC, Clark A, Courneya KS. A survey of physical activity
prostate cancer. Prostate Cancer Prostatic Dis 2007;10:340–6. programming and counseling preferences in young-adult cancer survivors.
[117] Galvao DA, Spry NA, Taaffe DR, Newton RU, Stanley J, Shannon T, et al. Cancer Nurs 2012;35:48–54.
Changes in muscle, fat and bone mass after 36 weeks of maximal androgen [135] Gjerset GM, Fossa SD, Courneya KS, Skovlund E, Jacobsen AB, Thorsen L.
blockade for prostate cancer. BJU Int 2008;102:44–7. Interest and preferences for exercise counselling and progamming among
[118] Galvao DA, Taaffe DR, Spry N, Joseph D, Turner D, Newton RU. Reduced Norwegian cancer survivors. Eur J Cancer Care (Engl) 2011;20:96–105.
muscle-strength and functional performance in men with prostate cancer [136] Jones LW, Courneya KS. Exercise counseling and programming preferences of
undergoing androgen suppression: a comprehensive cross-sectional cancer survivors. Cancer Pract 2002;10:208–15.
investigation. Prostate Cancer Prostatic Dis 2009;12:198–203. [137] Jones LW, Guill B, Keir ST, Carter K, Friedman HS, Bigner DD, et al. Exercise
[119] Spry N, Kristjanson L, Hooton B, Hayden L, Neerhut G, Gurney H, et al. interest and preferences among patients diagnosed with primary brain
Adverse effects to quality of life arising from treatment can recover with cancer. Support Care Cancer 2007;15:47–55.
intermittent androgen suppression in men with prostate cancer. Eur J Cancer [138] Karvinen KH, Courneya KS, Campbell KL, Pearcey RG, Dundas G, Capstick V,
2006;42:1083–92. et al. Exercise preferences of endometrial cancer survivors. Cancer Nurs
[120] Galvao DA, Taaffe DR, Spry N, Joseph D, Newton RU. Cardiovascular and 2006;29:259–65.
metabolic complications during androgen deprivation: exercise as a potential [139] Karvinen KH, Courneya KS, Venner P, North S. Exercise programming and
countermeasure. Prostate Cancer Prostatic Dis 2009;12:233–40. counseling preferences in bladder cancer survivors: a population-based
[121] Jones D, Round J, de Haan A. Skeletal muscle from molecutes to study. J Cancer Surviv 2007;1:27–34.
movement. London: Churchill Livingstone; 2004. [140] Karvinen KH, Raedeke TD, Arastu H, Allison RR. Exercise programming and
[122] Courneya KS. Efficacy, effectiveness, and behavior change trials in exercise counseling preferences of breast cancer survivors during or after radiation
research. Int J Behav Nutr Phys Act 2010;7:81. treatment. Oncol Nurs Forum 2011;38:E326–34.
[123] Courneya KS, Friedenreich CM, Quinney HA, Fields AL, Jones LW, Fairey AS. [141] Lowe SS, Watanabe SM, Baracos VE, Courneya KS. Physical activity interests
Predictors of adherence and contamination in a randomized trial of exercise and preferences in palliative cancer patients. Support Care Cancer
in colorectal cancer survivors. Psychooncology 2004;13:857–66. 2010;18:1469–75.
[124] Courneya KS, Friedenreich CM, Quinney HA, Fields AL, Jones LW, Vallance JK, [142] Rogers LQ, Courneya KS, Verhulst S, Markwell SJ, McAuley E. Factors
et al. A longitudinal study of exercise barriers in colorectal cancer survivors associated with exercise counseling and program preferences among breast
participating in a randomized controlled trial. Ann Behav Med cancer survivors. J Phys Act Health 2008;5:688–705.
2005;29:147–53. [143] Rogers LQ, Malone J, Rao K, Courneya KS, Fogleman A, Tippey A, et al. Exercise
[125] Courneya KS, McKenzie DC, Reid RD, Mackey JR, Gelmon K, Friedenreich CM, preferences among patients with head and neck cancer: prevalence and
et al. Barriers to supervised exercise training in a randomized controlled trial associations with quality of life, symptom severity, depression and rural
of breast cancer patients receiving chemotherapy. Ann Behav Med residence. Head Neck 2009;31:994–1005.
2008;35:116–22. [144] Rogers LQ, Markwell SJ, Verhulst S, McAuley E, Courneya KS. Rural breast
[126] Perna FM, Craft L, Carver CS, Antoni MH. Negative affect and barriers to exercise cancer survivors: exercise preferences and their determinants.
among early stage breast cancer patients. Health Psychol 2008;27:275–9. Psychooncology 2009;18:412–21.
[127] Whitehead S, Lavelle K. Older breast cancer survivors’ views and preferences [145] Stevinson C, Capstick V, Schepansky A, Tonkin K, Vallance JK, Ladha AB, et al.
for physical activity. Qual Health Res 2009;19:894–906. Physical activity preferences of ovarian cancer survivors. Psychooncology
[128] Ottenbacher AJ, Day RS, Taylor WC, Sharma SV, Sloane R, Snyder DC, et al. 2009;18:422–8.
Exercise among breast and prostate cancer survivors – what are their [146] Trinh L, Plotnikoff RC, Rhodes RE, North S, Courneya KS. Physical activity
barriers? J Cancer Surviv 2011;5:413–9. preferences in a population-based sample of kidney cancer survivors.
[129] Speed-Andrews AE, McGowan EL, Rhodes RE, Blanchard CM, Culos-Reed SN, Support Care Cancer 2012;20:1709–17.
Friedenreich CM, et al. Correlates of strength exercise in colorectal cancer [147] Vallance JKH, Courneya KS, Jones LW, Reiman T. Exercise preferences among
survivors. Am J Health Behav 2013;37:162–70. a population-based sample of non-Hodgkin’s lymphoma survivors. Eur J
[130] Lowe SS, Watanabe SM, Barclay SI, Courneya KS. Determinants of physical Cancer Care (Engl) 2006;15:34–43.
activity in palliative cancer patients: an application of the theory of planned
behavior. J Support Oncol 2012;10:30–6.

You might also like