Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest types of cancer, and the increasing incidence of PDAC may be related to the prevalence of obesity. Physical activity (PA), a method known to mitigate obesity by increasing total energy expenditure, also modifies multiple cellular pathways associated with cancer hallmarks. Epidemiologic evidence has shown that PA can lower the risk of developing a variety of cancers, reduce some of the detrimental side effects of treatments, and improve patient’s quality of life during cancer treatment. However, little is known about the pathways underlying the correlations observed between PA interventions and PDAC. Moreover, there is no standard dose of PA intervention that is ideal for PDAC prevention or as an adjuvant of cancer treatments. In this review, we summarize relevant literature showing how PDAC patients can benefit from PA, the potential of PA as an adjuvant treatment for PDAC, the studies using preclinical models of PDAC to study PA, and the clinical trials to date assessing the effects of PA in PDAC.
Keywords: Physical Activity, Exercise, Pancreatic Ductal Adenocarcinoma, Pancreatic Cancer, Translational Research
Introduction
Pancreatic ductal adenocarcinoma (PDAC) is a fatal disease that became the third leading cause of cancer-related death in the US in 2020, and has an average five-year survival rate of less than 10% 1. PDAC is projected to become the second leading cause of cancer-related death by 20302. Low survival rates of PDAC patients are often attributed to a combination of factors, including the absence of preventive measures, late stage diagnosis, and the lack of effective treatment options. Currently, no interventions to prevent PDAC are available. Therefore, understanding and preventing known risk factors is critical to the prevention of this deadly disease.
Obesity is a major risk factor for many cancers including PDAC 3–6. A case-control study showed that the death rate from all cancers increased by 52% in men and 62% in women with a body mass index (BMI) greater than 40 kg/m2 compared to normal subjects 7. Moreover, the relative risk of PDAC increased 1.12-fold for each 5 kg/m2 increase in BMI 8 and the survival rate decreased in subjects whose BMI was over 25 kg/m2 from the ages of 30 to 79 9. These studies suggest that maintaining a healthy weight may reduce the risk of developing cancers associated with obesity, such as PDAC.
Increasing physical activity (PA) could be a practical approach for weight loss with the goal of modulating molecular mechanisms associated with decreasing tumor development and growth10–13 to reduce cancer risk 12, 14, 15. The intensity of PA in humans is expressed in metabolic equivalents of task (METs), defined as the energy it takes for an adult to sit quietly and is roughly equivalent to the expenditure of one kilocalorie per kilogram of body weight per hour 16. Activities that expend three to six METs, such as brisk walking, are considered “moderate activities,” and those that expend more than six METs, like running are considered “vigorous activities” 17. The World Health Organization guidelines recommend that adults (18–64 years) engaged in 150 minutes of moderate activity or 75 minutes of vigorous activity per week 17. On the other hand, cancer patients or survivors are recommended to exercise three times per week for about 30 minutes and perform resistance training twice a week for a minimum of 120 minutes of activity 18. However, for some cancer patients, achieving these recommendations can be difficult especially for those that develop of a secondary wasting disorder called cachexia.
Cachexia is a metabolic syndrome commonly found in PDAC patients that lowers their survival 19, 20. Symptoms of cachexia include muscle and adipose tissue wasting, involuntary weight loss, reduced appetite, insulin resistance, and edema 21. Cachexia-related tissue muscle wasting, or atrophy, reduces the ability to perform physical activity or exercise 22. This atrophy is driven by a loss of muscle mass, myofiber size, muscle oxidative capacity and protein synthesis with a concurrent increase in protein breakdown 22. In preclinical studies, physical activity or exercise training counteracts muscle atrophy23, 24. Studies in humans are small and less clear about the effectiveness of physical activity with or without nutritional supplementation on muscle wasting in cachexia25–27. However, larger clinical trials, like the Norwegian MENAC study on exercise, nutrition, and anti-inflammatory medications, are under way in cachectic pancreatic cancer patients to examine the effects of a multimodal therapy on cachexia (EUCTR2013–002282-19). These trials aim to better understand PA outcomes, like improved muscle atrophy or increased patient survival 22, 28.
Despite the recommended PA guidelines, it remains poorly understood whether PA: can benefit patients at risk of developing PDAC, be beneficial as an adjuvant treatment of PDAC, and be accurately modeled using preclinical models of PDAC. To investigate these knowledge gaps, we performed a literature search on Google Scholar and Pubmed using the following keywords: “physical activity”, “pancreatic ductal adenocarcinoma”, “pancreatic cancer”, “sports”, “leisure time physical activity” and “physical exercise” from 1995 to April, 2021. Moreover, to summarize the clinical trials in these setting to date, we searched the clinicaltrials.gov website, the World Health Organization (WHO) database, and the European Society of Medical Oncology (ESMO) using the following criteria: “physical activity” OR “exercise” AND “pancreatic cancer”. We summarize and discuss the studies that resulted from these searches in this review. We also highlight the most commonly utilized methods to study the effects of PA in the preclinical setting and the ongoing clinical trials conducted to explore the effects of PA on multiple aspects of PDAC. Additionally, we summarize the general findings of the pre-clinical and clinical studies in PDAC (Figure 1), bringing attention to the overlaps and gaps in research between them.
Figure 1:
Overview of how PA affects pre-clinical and clinical models. Created with Biorender.com
Pancreatic Cancer Risk Factors and their Relationship with Physical Activity
There are multiple risk factors of PDAC that are either modifiable or non-modifiable 29, 30. Among the modifiable risk factors, PA may be beneficial at mitigating their risk. Below, we detail some of the risk factors of PDAC and the evidence for PA as a countermeasure.
Smoking:
Tobacco smoking is a modifiable and independent risk factor of PDAC 31–33 for which some of the mechanisms have been recently summarized 34. One of the side effect of smoking is the development of bronchial asthma. A prospective study observed a positive association between bronchial asthma and the risk of pancreatic cancer (HR=2.16, 95% CI 1.17–3.98). Moderate/heavy PA in these patients suggested a decreased risk of pancreatic cancer (HR=0.42, 95% CI 0.22–0.83) however it was not directly correlated with improvements in the bronchial asthma 35.
Alcohol:
Epidemiological human studies 32, 36–42 and preclinical studies in mice using models expressing oncogenic Kras 43 have linked alcohol consumption to an increased risk of pancreatic cancer. In addition, alcohol consumption is a risk factor for chronic pancreatitis (CP) development 44, which promotes chronic inflammation in the pancreas and also increases the risk of pancreatic cancer. The pooled analysis conducted by Perreault et al. showed that people who met the PA recommendations can nearly attenuated the associations between alcohol intake and mortality risk of cancers 45, however the study did not report on pancreatic cancer specifically.
Obesity:
Obesity is another modifiable risk factor of pancreatic cancer 30, 46. The connection between pancreatic cancer and obesity has been well established 47–50. PA can reduce obesity by increasing the bodies energy expenditure and decreasing general adiposity 51, which may be contributing to reduce pancreatic cancer risk. It is proposed that the expansion of the adipose tissue promotes tumor development through the deposition of fibrosis, connective tissue, and the activation of inflammatory pathways among others 47, 52. However, whether increased PA could decrease the risk of pancreatic cancer by blocking these pathways induced by increased adiposity is unclear. Therefore, these topics and associations merit further investigation since weight loss via bariatric surgery has been associated with a decreased cancer incidence, particularly obesity-associated cancers such as pancreatic cancer53.
Non-genetic chronic pancreatitis:
The risk of CP is associated with smoking and heavy alcohol consumption 54, 55. The inflammatory state that partially characterizes CP can promote metaplasia and neoplastic transformation, leading to pancreatic cancer development 56, 57. A meta-analysis showed that CP patients have almost an eight-fold increased risk of PDAC five year after diagnosis 58. However, limited evidence was found in our search regarding the contribution of increased PA in reducing the risk of CP-associated with PDAC development.
Diabetes:
Diabetes mellitus is another known risk factor for pancreatic cancer 59. High glucose levels increased O-linked N-acetylglucosaminylation and promoted de novo KRAS mutations 60, which is present in more than 95% of PDAC 61. Diabetes or impaired glucose tolerance could be found in 50–80% of pancreatic cancer patients when diagnosed 59. Pannala et al. showed that in pancreatic cancer patients, 74% of diabetes were new-onset diabetes mellitus, meaning that diabetes developed no more than two years before the cancer diagnosis 62. High diabetes prevalence, especially new-onset diabetes mellitus is unique to pancreatic cancer patients 63. Researchers are currently identifying novel biomarkers to screen pancreatic cancer in patients with new-onset diabetes mellitus 64, 65. PA may delay or prevent diabetes development 66 and improve blood glucose control 67 in diabetes patients. In a large cohort study from Japan, Iwasaki et al. suggested that fast walking was preventive for new-onset diabetes (OR=0.93, 95% CI 0.88–0.98) 68. Not many studies describe the association between type 1 diabetes and pancreatic cancer. One cohort study using data from five countries showed that the HR associated with type I diabetes with pancreatic cancer was 1.53 in men (95% CI 1.30–1.79) and 1.25 in women (95% CI 1.02–1.53) 69. However, overall it is still unclear whether PA can reduce the risk of pancreatic cancer after the onset of diabetes and further studies are needed.
Mitigating Pancreatic Cancer Risk with Physical Activity
Moderate- or vigorous- intensity activities as defined by the 2008 physical activity guidelines for americans 70 have been associated with longevity in cancer patients 71. Moreover, several large cohorts or case control studies have demonstrated an inverse relationship between PA and pancreatic cancer (including PDAC) incidence and/or mortality in men and women. A meta-analysis of 26 independent studies concluded that, although there was significant heterogeneity between studies (due to study design, the median age of the population, and confounding variables used for adjustment), the overall protective association between leisure time PA and pancreatic cancer was 11% 72. Interestingly, low or moderate intensity PA was more beneficial than vigorous activity in lowering the risk of pancreatic cancer 73. A cohort study that included 43,479 men indicated that a moderate range of MET hours (42–62.9 MET hrs/week) was more beneficial (hazard ratio [HR] = 0.59) than a low range (< 20.9 MET hrs/week; HR = 1) or a high range (> 63 MET hrs/wk; HR = 0.90) of MET hours. The range of 42–62.9 MET hours/week provided an optimal inverse correlation risk for pancreatic cancer 74. Song and Giovannucci further divided participants in the two nationwide cohorts into a low-risk group (nonsmokers, with no or moderate alcohol consumption, a BMI between 18.5–27.5, and 150 minutes of moderate or 75 minutes of vigorous PA weekly) and a high-risk group (those who failed to meet all four of the low-risk-group criteria). According to the analysis, both the incidence rate and mortality rate of pancreatic cancer were lower in the low-risk group 75. Moreover, the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort study showed that healthy life style habits including increased PA were inversely associated with pancreatic cancer risk in European adults 76. Together, these studies illustrate that increased PA may lower the risk of developing pancreatic cancer. However, several limiting factors made it hard to understand the true protective effect of PA, which included study design, age of the patient population, measurement of PA, and confounding variables used for analysis adjustments. In some studies, it was also hard to discern whether other well-known pancreatic cancer risk factors discussed above were confounding factors in interpreting the effects of PA in PDAC. Future research should consider all risk variables to improve the interpretation of the studies on the effects of PA in pancreatic cancer.
Effects of Physical Activity on PDAC Tumor Growth
Although preventing PDAC is ideal, the cancer diagnosis may still occur before a PA intervention could begin. Therefore, many researchers have focused on studying whether PA can be used as an adjuvant therapy for PDAC in combination with standard of care to slow or reverse its growth in both pre-clinical models and clinical trials.
One of the first pre-clinical animal studies to demonstrate the benefits of exercise in reducing pancreas tumors evaluated the protective effects of voluntary wheel running in male Lewis and female F344 rats injected with azaserine, a pancreatic carcinogen. Voluntary wheel running in these rats reduced the growth rate of azaserine-induced pancreatic foci for four months after carcinogen initiation 77. A study using voluntary wheel running prior to subcutaneously implanting PANC-1 tumor cells into mice showed that PA inhibited tumor growth 78. In this study, voluntary wheel running was started one week before tumor cell implantation and lasted 63 days. After tumor cell injection, the Panc-1 tumors in the mice with access to running wheels grew at a slower rate (0.00476cm2 per day) compared to the control mice (0.00654cm2 per day) 78. These results suggest that voluntary wheel running could delay PDAC tumor growth in pre-clinical mouse models; however, these results have not been validated using genetically engineered mouse models of PDAC and/or in combination with PDAC treatment regimens. Moreover, these results are only modest and tested only one human pancreatic cancer cell line.
Aerobic exercise can also play a role in remodeling the tumor microenvironment. Florez Bedoya et al. reported that exercise remodels the tumor vasculature in a patient-derived xenograft mouse model of PDAC and in PDAC patients 79. The increased shear stress via aerobic exercise can cause tumor vessel remodeling through calcineurin-NFAT-TSP1 signaling in endothelial cells and improve the delivery of gemcitabine in a PDAC tumor-bearing mouse model 80. The evidence suggests that PDAC patients could benefit from PA, clinically.
Although several studies have demonstrated some of the positive effects of exercise in delaying PDAC growth, the underlying mechanisms of these effects are unclear. Several molecular mechanisms had linked PA to cancer prevention and treatment 81, 82. Among them are molecular mechanisms related to changes in metabolism, systemic immune function 10, 13, tumor microenvironment 11, and some other hallmarks of cancer 12. Metabolically, PA increased a tumoricidal immune response by reducing glucose consumption 83 and tumor proliferation by altering gene expression of fatty acid metabolism pathways 84. However, many of these mechanisms have not been verified in PDAC as a result of PA. Therefore, further studies on these phenomena are needed to explore the molecular mechanisms of how PA could reduce the risk of PDAC or delay tumor growth in particular using genetically engineered mouse models of PDAC that recapitulate various stages of the disease.
Effects of Physical Activity in Combination with PDAC Treatments
Although PA may have direct anti-tumor effects for some cancer types, PA may also help improve some of the side effects cancer patients experience as a result of cancer treatments. PA interventions can mitigate common chemotherapy side effects like fatigue and tissue wasting, improve better outcomes, treatment completion rates, and quality of life during and after treatment 85–96. The safety and efficacy of PA in PDAC patients that undergo adjuvant treatment were first reported by Cormie et al. 97. In the study, a 49-year-old male PDAC patient whose tumor was surgically removed three months before the PA intervention and who received adjuvant chemotherapy and radiotherapy tolerated the exercise program well. The exercise program also had a positive impact on several outcomes, including physical capacity and functional ability 97. A single-arm prospective trial showed that PDAC patients who were able to adhere to a home-based exercise program during preoperative therapy increased their capacity for PA and the exercise program had no related adverse effects 98 (Table 1, NCT02295956). Moreover, PDAC patients receiving preoperative chemotherapy and/or chemoradiation showed that light PA increased weekly was correlated with increased health-related quality of life (HRQOL) (β = 0.03, p = 0.02), while sedentary activities were associated with decreased HRQOL ((β = −0.02, p = 0.01) 99 (Table 1, NCT02295956). A study by Niels et al. reported that a well-tolerated and feasible PA program in advanced PDAC patients could improve medical treatment outcomes, improve oxygen uptake, and quality of life (QoL) in pre-operative patients before liver metastasis resection100 (Table 1, NCT02940067). Even less strenuous activity, like progressive mobilization after pancreatic cancer surgery, reported benefits by increasing oxygenation when performed on the same day of the surgery, compared to patients mobilized a day after the surgery101 (Table 1, NCT03466593). Moreover a post-operative randomized control trial (RCT) suggested that three months of resistance training improve patients’ sleep problems and fatigue102 (Table 2, NCT01977066). They also showed improved muscle strength but no significant effects on body composition after six months of resistance training. In this trial, loss of muscle mass was a predictor of poor overall survival, suggesting that training to maintain muscle mass and avoid muscle wasting from malnutrition or cachexia benefited patients103 (Table 2, NCT01977066). A recent systematic review conducted by Luo et al. 104 also suggested that exercise is a safe and feasible treatment for PDAC patients and may positively affect several physical and psychological outcomes. So far the improvements to patient outcomes as a result of PA may be due to several aspects such as, tumor vasculature remodeling, lowered inflammation, and metabolic improvements, however there might be many undiscovered mechanisms that require further research. While most PA intervention studies in PDAC are associated with improved quality of life, fatigue, and functional capacity, most of the conclusions are drawn from studies with low levels of evidence like case series and case reports. However, another systematic review of exercise efficacy in PDAC also suggested a significant lack of high-quality evidence from RCTs, which severely limits the interpretation of the results from many of the studies conducted on this topic 105. In addition, even the few RCTs that investigate the role of PA in pancreatic cancer patients have significant variations in the type of intervention modality, including the type and duration of the physical activity (aerobic and resistance, 1–5X per week, etc.), outcomes measured (VO2 peak, VO2 max, 6 min walk test, body composition, fatigue, etc.) and the frequency of these measurements. The heterogeneity of the studies and the lack of high-quality evidence delays reaching a consensus on the most appropriate PA intervention for these patients. Therefore, well-designed clinical trials are needed to address the limitations of current studies and determine the efficacy and prescription of PA for pancreatic cancer patients 105. We recorded the primary outcome measures in the studies we found from our search in Tables 1–5. Our findings highlight the heterogeneity in study design of these trials in the different patient settings.
Table 1.
Clinical Trials of PDAC and Physical Activity for Pre-Operative Patients (N/A: Not applicable)
| Study Title | Conditions & Diseases | Interventions | Study Population | Primary Outcome Measure | Phase | ID Number |
|---|---|---|---|---|---|---|
|
| ||||||
| Pilot Study of a Multimodal Prehabilitation Pancreatic Cancer Program | Pancreatic Ductal Adenocarcinoma | Exercise Program, Nutrition Program | 40 | Number of participants to complete prehabilitation program | N/A | NCT03865875 |
| Exercise and Nutrition to Improve Pancreatic Outcomes | Pancreatic Cancer | Nutritional Counseling, Standard Exercise, Enhanced Exercise | 60 | Quality of Life (QOL), change in body weight, physical performance, fatigue | N/A | NCT03256201 |
| PancFit: Multimodal Exercise During Preoperative Therapy for Pancreatic Cancer | Pancreatic Cancer | Questionnaires, Nutritional Counseling, Physical Assessments | 128 | Change in 6 Minute Walk Test (6MWT) | N/A | NCT03187951 |
| Improving Outcomes in Cancer Patients With a Nutritional and Physical Conditioning Prehabilitation Program | Pancreatic Cancer, Liver Cancer, Bile Duct Cancer, Hepatobiliary Cancer, Surgery | Exercise, Nutrition, Relaxation Techniques | 60 | 6MWT | N/A | NCT03475966 |
| Monitoring Heart Rate Variability for the Early Detection of Pancreatic Cancer | Pancreatic Ductal Adenocarcinoma | Activity monitor, Quality-of-Life Assessment, Questionnaire Administration | 750 | Magnitude of heart rate variability (HRV) decline (Stage I) Compliance statistics for wristband use (Stage II) |
N/A | NCT04400903 |
| Re-Defining Frailty and Improving Outcomes Through Prehabilitation in Patients With Pancreatic, Liver, or Gastric Cancer, The RIOT Trial | Adult Liver Carcinoma, Gastric Carcinoma, Malignant Solid Neoplasm, Pancreatic Carcinoma | Best Practice Behavioral, Exercise Intervention, Physical Therapy, Questionnaire Administration | 50 | Frailty assessment | N/A | NCT04602026 |
| Walking for Recovery From Surgery in Improving Quality of Life in Older Adults With Lung or Gastrointestinal Cancer and Their Family Caregivers | Adult Liver Carcinoma, Caregiver, Colorectal Carcinoma, Lung Carcinoma, Malignant Digestive System Neoplasm, Pancreatic Carcinoma | Exercise Intervention, Quality-of-Life Assessment, Survey Administration | 50 | 6MWT, daily steps, timed-up and go | N/A | NCT03267524 144 |
| HOPE - A Study to Evaluate the Effect of a Prehabilitation Program on GI Cancer Patients Planning to Undergo Surgery | Pancreatic Cancer, Esophageal Cancer, Gastric Cancer | Nutritional Intervention, Physical Activity Intervention | 60 | Frailty Measurement weight loss, grip strength, and prealbumin serum level | 4 | NCT03642093 |
| Preoperative Exercise in Pancreatic Cancer | Pancreatic Cancer | Exercise Program, Standard Care | 3 | Change in 400-meter walk time | N/A | NCT02648880 |
| Preoperative Rehabilitation During Neoadjuvant Therapy for Pancreatic Cancer | Pancreatic Cancer | Questionnaires, Exercise, Nutrition Counseling, Phone Calls, Booklet | 75 | Feasibility of Prehabilitation Program Among Pancreatic Patients | 1 | NCT02295956 98, 99, 145 |
| Enhancing Fitness Before Pancreatic Surgery | Pancreatic Cancer Chronic Pancreatitis | MedEx: A Combination of Dietary and Exercise Interventions over Four Weeks Before Scheduled Pancreatic Surgery | 20 | Peak power - measured using cardiopulmonary exercise testing | N/A | NCT02940067 100, 146, 147 |
| Effects of Prehabilitation and Early Mobilization for Patients Undergoing Pancreas Surgery. | Pancreas Cancer | Prehabilitation, Routine Care, Extra Early Mobilization, Standard Mobilization | 245 | Postoperative complications, Partial Pressure of Oxygen (PaO2) | N/A | NCT03466593 101 |
| Exercise medicine as chemotherapy adjunct: the feasibility and efficacy of a multicomponent program in pancreatic cancer patients receiving neoadjuvant therapy (The EXPAN Project) | Pancreatic Cancer | Resistance Training, Aerobic Exercise or Adapted Sport-Related Drills | 40 | Feasibility, Safety | 1 | ACTRN12620001081909148 |
| A phase II study about the efficacy of an intervention of exercise therapy on the adjuvant chemotherapy for pancreatic cancer: Exercise-PC Study - Exercise impact on adjuvant chemotherapy for pancreatic cancer | Invasive Ductal Adenocarcinoma of Pancreas | Exercise Therapy | 44 | The completion rate of four courses of S-1 adjuvant chemotherapy. | 2 | JPRN-UMIN000030124 |
Table 2.
Clinical Trials of PDAC and Physical Activity for Post-Operative Patients (N/A: Not applicable)
| Study Title | Conditions & Diseases | Interventions | Study Population | Primary Outcome Measure | Phase | ID Number |
|---|---|---|---|---|---|---|
|
| ||||||
| PRECISE: Pancreatic Cancer and Exercise | Pancreas Adenocarcinoma | Exercise | 10 | Participants adhering, participants acceptability | N/A | NCT04305067 |
| Diet and Exercise After Pancreatic Cancer | Pancreatic Cancer | Diet + Exercise | 50 | Adherence, adverse events | N/A | NCT03187028 |
| Adapted Physical Activity in Patients With Resected Pancreatic Cancer (APACaPOp PRODIGE-56 Study): a National Multicenter Randomized Controlled Phase II Trial | Pancreas Cancer | Unsupervised Adapted Physical Activity (APA) program, Supervised APA program | 252 | Month 6 (M6), Health-Related quality of life (HRQoL) | N/A | NCT03400072 |
| Improving Outcomes in Cancer Patients With a Nutritional and Physical Conditioning Prehabilitation Program | Pancreatic Cancer, Liver Cancer, Bile Duct Cancer, Hepatobiliary Cancer | Exercise, Nutrition Relaxation techniques | 60 | 6MWT | N/A | NCT03475966 |
| Association Between Health Care Provider (HCP)-Assessed ECOG Performance Status (PS) and Overall Survival, and Objectively Measure of Physical Activity (PA) Levels in Advance-cancer Patients” | Pancreatic Adenocarcinoma, Pancreatic Neuroendocrine Carcinoma and 45 more | Exercise Intervention, Health Telemonitoring, Quality-of Life Assessment, Questionnaire Administration | 590 | Study Completion, Physical Activity (PA) Assessment, ECOG (Eastern Cooperative Oncology Group) Performance Status | N/A | NCT01365169 |
| Resistance Training Intervention to Improve Physical Function in Patients With Pancreatic Cancer Receiving Combination Chemotherapy or Have Undergone Surgery, PancStrength Study | Advanced Pancreatic Adenocarcinoma, Pancreatic Adenocarcinoma, Stage III Pancreatic Cancer AJCC v8, Stage IV Pancreatic Cancer AJCC v8 | Educational Intervention Quality-of Life Assessment, Questionnaire Administration, Resistance Training | 75 | Safety of the tele-resistance training (RT) program, Post-exercise muscle soreness, Body composition, Aerobic fitness | N/A | NCT04837118 |
| Walking for Recovery From Surgery in Improving Quality of Life in Older Adults With Lung or Gastrointestinal Cancer and Their Family Caregivers | Adult Liver Carcinoma, Caregiver, Colorectal Carcinoma Lung Carcinoma, Malignant Digestive System Neoplasm, Pancreatic Carcinoma | Exercise Intervention, Quality-of Life Assessment, Survey Administration | 80 | 6MWT, daily steps, timed-up and go | N/A | NCT03267524 144 |
| Exercise Intervention Study for Pancreatic Cancer Patients | Pancreatic Cancer | Six months Supervised Resistance Training, Six months Home-Based Exercise Training | 65 | Physical functioning score, as assessed by the European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ) EORTC QLQ-C30 | N/A | NCT01977066 102, 103 |
| A validation study of biomarkers to predict responders to non-pharmacological multimodal program for advanced cancers - NEXTAC-Three | Non-Small-Cell Lung and Pancreatic Cancer | Nutritional Counseling, Home-based Exercise Intervention, Physical Activity Intervention | 100 | Positive predictive value | N/A | JPRN-UMIN000033574 |
| A Randomized Phase II study of the nutritional and exercise treatment for the elderly patients with advanced non-small-cell lung and pancreatic cancer - NEXTAC-TWO study | Non-Small-Cell Lung and Pancreatic Cancer | Nutritional Counseling, Home-based Exercise Intervention, Physical Activity Intervention, Usual Care | 130 | Disability-free survival | 2 | JPRN-UMIN000028801149 |
| A Feasibility study of the nutritional and exercise treatment for the elderly patients with advanced non-small-cell lung and pancreatic cancer - NEXTAC-ONE study | Non-Small-Cell Lung and Pancreatic Cancer | Early Multimodal Intervention (Nutritional Intervention, Home-Based Muscle Training, and Lifestyle Intervention to Promote Physical Activity) | 30 | Attendance rate for exercise and nutritional sessions in 8 weeks | N/A | JPRN-UMIN000023207150 |
| Post-operative high intensity interval training in patients undergoing major foregut cancer surgery, investigating the effect on the 6 minute walk test: A randomised controlled trial | Liver Cancer, Gastric Cancer, Pancreatic Cancer, Oesophageal Cancer, Oesophago-gastric Junction Cancer | Exercise, Usual Care | 60 | 6 Minute Walk Test Grip Strength Test. | N/A | ACTRN12620000315910 |
| The influence of exercise on blood glucose control and the anti-oxidative ability of the blood in patients after pancreatectomy: a randomized controlled trial | Pancreatic Disease, mainly Neoplastic Tumors of the Pancreas | Pancreatectomy, Followed by Exercise | 40 | Measurement of blood glucose levels, HBA1c, albumin and uric acid, 12 weeks after the initiation of exercise (once every 4 weeks interval) | N/A | ISRCTN10827174 |
Table 5.
Clinical Trials of PDAC and Physical Activity: Without Specified Patient Setting (N/A: Not applicable)
| Study Title | Conditions & Diseases | Interventions | Study Population | Primary Outcome Measure | Phase | ID Number |
|---|---|---|---|---|---|---|
|
| ||||||
| Early Palliative Care for Patients With Advanced Pancreatic Cancer. | Pancreatic Cancer | Early Palliative Care | 250 | Adjusted mean change in global health status/QOL score at 12 weeks | N/A | NCT04632303 |
| Brown Adipose Tissue Activity and Energy Metabolism in Cachexia | Cachexia, Neoplasms Pulmonary Disease, Chronic Obstructive Pancreatic Neoplasms | Radiation, Abdominal Subcutaneous Adipose Tissue Biopsy, Blood Sampling, Indirect Calorimetry, Device: Accelerometry, Double-Labeled Water | 16 | Brown adipose tissue (BAT) activity measured by PET(-MRI) (Positron Emission Tomography – Magnetic Resonance Imaging) | N/A | NCT02500004 153 |
| Effect of exercise on sarcopenia in patients with pancreatic cancer | Pancreatic Cancer | Physical Exercise and Standard Therapy | 40 | Evaluation for sarcopenia using bioimpedance analysis | N/A | JPRN-UMIN000029271154 |
| Effect of exercise on sarcopenia in patients with pancreatic cancer | Pancreatic Cancer | Physical Exercise and Standard Therapy | 40 | Evaluation for sarcopenia using bioimpedance analysis | N/A | JPRN-UMIN000029272155 |
| A feasibility study of Multimodal Exercise/Nutrition/Anti-inflammatory treatment for Cachexia – the pre-MENAC study - Pre-MENAC Study | Non-operable Stage III/IV Non-Small Cell Lung Cancer, Pancreatic Cancer | Celecoxib CAS Number: 194044-54-7, 100mg | 40 | Is a multimodal intervention for cancer cachexia feasible? | 2 | EUCTR2010-022897-14-GB |
| PreMENAC: Multimodal Exercise/Nutrition/Anti-inflammatory treatment for Cachexia: A feasibility study (phase II) - Pre - MENAC | Non-operable Stage III/IV Non-Small Cell lung Cancer, Pancreatic Cancer | Celecoxib CAS Number: 194044-54-7, 100mg | 40 | The overall objectives are the feasibility of patient recruitment, compliance to the multimodal intervention as well as the assessment of possible contamination of the control group. | 2 | EUCTR2010-022897-14-NO |
| A Feasibility Study of Multimodal Exercise/Nutrition/Anti-inflammatory Treatment for Cachexia - the Pre-MENAC Study | Non-Operable Stage III/IV Non-Small Cell Lung Cancer, Pancreatic Cancer | Multimodal intervention Other Standard of care | 46 | Feasibility of recruitment and retention | N/A | EUCTR2013-002282-19/NCT01419145156–158 |
Benefits of Physical Activity in Pancreatic Cancer Survivors
Cancer survivors tend to experience physical and psychological deconditioning after cancer therapy 106, 107. PA can potentially help patients recover from deconditioning due to its potential to improve common cancer-related disorders, such as fatigue, anxiety, and reduced physical functioning 108, 109. Cancer survivors are encouraged to do moderate-intensity aerobic training at least three times per week, for at least 30 minutes for 8 to 12 weeks to address health-related outcomes after cancer treatment 18. A telephone survey found that over 70 % of pancreatic cancer survivors are willing to participate in exercise and diet interventions 110. One study pointed out that most cancer survivors are insufficiently active, especially those who are obese 111. Moreover, Parker et al. showed that less than one-quarter of patients after PDAC resection met the U.S. exercise guidelines 112. Barriers that keep survivors from regular PA include past exercise experience, motivation, environmental barriers, and logistical barriers 112. Therefore, ways to break exercise barriers and elicit patients’ motivation to increase PA are needed for cancer survivors to experience the benefits of increased PA.
Commonly Used Preclinical Models to Study Physical Activity
Mouse models have been used for decades to study the impact of structured exercise training on a variety of cancers 113, 114. Several techniques are routinely used to study the effects of exercise in mice, such as either voluntary or involuntary running systems including wheel running (voluntary), treadmill running (involuntary), swimming (involuntary), and resistance training (involuntary) (Figure 2). Unfortunately, there are inconsistencies in study design related to how studies used voluntary or involuntary methods of exercise when using preclinical models of cancers to assess the effects of exercise. The exercise methods utilized depends on the investigator’s preferences of the experimental design. However, there are some differences between voluntary and involuntary exercise that influence the interpretation of results and the biological relevance of each model. Kim et al. showed that mice running on wheels had higher average grip strength and a different expression pattern of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1 α, compared to those mice running on a treadmill 115. However, Conner et al. showed that using electrical shock to train mouse to performed involuntary exercise increased the levels of stress hormones and cytokines after exercise 116. These elevated stress hormones and cytokines might lead to confounding results in the context of cancer research as some of these stress hormones 117 and cytokines 118 are known to affect cancer development.
Figure 2:
Summary of the common methods of pre-clinical PA assessment. Created with Biorender.com
In addition to the variety of exercise techniques to choose from, it is also important to consider the time of day at which the mice exercise. Mice are nocturnal animals and are most active during the night. Therefore, to study PA/exercise using mouse models, some researchers have altered the mouse housing environments to provide a reversed light-dark cycle so that the exercise/PA interventions can occur during the rodent’s night phase when mice are naturally more active 119.
Wheel Running (Voluntary):
Wheel running is one of the two most common forms of exercise training used in preclinical models because it is less labor intensive and reduces the amount of psychological stress experienced by the mice by allowing them to choose the duration and intensity of running. The lack of control over exercise intensity is one of the limitations of using wheel running instead of treadmills; wheel running often prevents the mice from running at their maximal intensity. Moreover, the mice run voluntarily and therefore must be motivated and willing to run when given access to the wheels.
Treadmill Running (Involuntary):
Treadmill running is the other most common form of exercise used in both human clinical studies and murine preclinical studies. One of the primary advantages of using treadmills is the uniformity with which the level of exertion of each experimental group is measured. Treadmill running enables factors such as running duration, intensity, and frequency to be controlled so subjects exercise at maximal or submaximal levels. Unlike voluntary wheel running, treadmill exercise sometimes requires methods like tail tapping, shock grid, air burst, or cold-water jets to encourage the animal to run. These types of methods can aggravate an already stressful experience in these animals, and they have been associated with tumor development and growth 120. To avoid stress induced by forced exercise, treadmill running requires the mice to gradually become familiarized with the apparatus. Often, treadmill performance parameters are evaluated based on the duration of running at fixed speeds and/or the gradual increase in speed until each mouse reaches their maximal workload 121. Among the disadvantages of treadmill exercise are frequent animal handling, the difficulty of motivating mice to exercise, lack of access to food and water during the training periods, and the need for constant investigator vigilance to ensure that the animals run for the entire duration of the exercise regimen. Moreover, researchers have identified strain-related differences in how mice handle treadmill exercises. Lerman et al. showed that C57BL6J mice had the least maximum speed of all the mice strains they tested. 121.
Swimming (Involuntary):
Swimming is another model used to study PA. Swimming has several advantages over other types of exercise modalities and has been shown to suppress tumor growth in mice 122. Swim training is typically performed in a swimming pool at a controlled temperature for less than one hour per day. The initial swim training consists of a gradual increase in workload until a maximal load is obtained. The procedure requires inexpensive equipment and relies on the natural swimming ability of laboratory rodents. Swimming behaviors are affected by diverse factors, such as the diving reflex, mental stress, and episodes of hypoxia associated with diving 123.
Resistance Training (Involuntary):
Resistance training is a form of exercise that can strengthen your skeletal muscles by providing progressive overload 124. Exercise interventions can benefit cancer patients by improving muscular strength, cardiovascular function, and quality of life.125. However, few studies have translated the use of exercise regimes in mice to identify the protective mechanisms of resistance training. The failure to identify protective mechanisms due to resistance training is in part due to the inability to relate exercise prescriptions used in humans to mice and vice versa. For example, a typical form of resistance training for mice is a “squat-like” movement proposed by Tamaki et al. 126. The major limitation of this method is the need for electrical stimulation, and this setting cannot be directly used in humans 127. Another form of resistance training for mice is climbing a rope with a small weight on their tail to provide a resistance-training stimulus 128 which does not have an equivalent exercise in humans.
Although mouse models are an essential and commonly used tool in preclinical studies, their ability to mimic human physiologic changes is still limited. Researchers still face important challenges, such as determining how to design a novel approach to bridge the mouse–human gap and determining how to use the results from current mouse models of exercise to design future human clinical trials to test these interventions.
Translating Preclinical Exercise Findings to the Clinic
Using mouse models to study disease mechanisms is often insightful; however, exercise interventions currently used in human clinical trials for patients with pancreatic cancer are not based on preclinical exercise studies or mechanisms identified in mice. There is a disconnect between translating important findings in mouse exercise studies and designing exercise programs for clinical trials based on these studies. This disconnect is due, in part, to the lack of linear correlation between mouse aerobic or resistance exercise intensity and the human equivalent, i.e. the lack of understanding of what 30 minutes of moderate intensity aerobic exercise in a mouse is equivalent to in a human.
Very few clinical trials have been conducted with patients to determine the effectiveness of incorporating exercise or PA for pancreatic cancer patients 97, 129. A limited number of studies have used animal models to investigate how exercise, or increased PA, affects or prevents pancreatic cancer 77, 78. Thus, despite the epidemiological evidence suggesting that PA decreases the risk of pancreatic cancer, a lack of translation to the clinic has affected the progress of investigating how PA/exercise could provide anti-pancreatic-cancer effects. Zheng et al. showed that exercise was sufficient to suppress tumor growth, increase food consumption, and decrease fat pad size in mice 78. This study demonstrated that exercise induced a positive effect by increasing active caspase-3 expression and decreasing the ratio of mitotic to apoptotic cells that lead to inhibition of tumor-growth. Similar studies are needed to further pinpoint the mechanisms by which exercise could inhibit pancreatic cancer growth. Given the limited research on links between the effects of PA and PDAC prevention or PDAC-risk reduction, it is beneficial to review studies of other cancers that have used exercise and PA as means of preventing cancer and reducing tumorigenesis. Recent studies have demonstrated that PA, alone or in combination with pharmacological treatments, has an effect on cancer prevention and prognosis 23, 125, 128, 130–132.
Several mouse models of PDAC also develop cachexia 133. Research on cancer cachexia has demonstrated that appropriate exercise recommendations play an important role in identifying how exercise affects cancer biology and disease progression. For example, voluntary wheel running counteracts cachexia-mediated overload of autophagic flux, which results in rescued muscle mass and function in tumor-bearing mice. Treating with 5-aAminoimidazole-4-carboxamide ribonucleotide or rapamycin trigger autophagic flux and replicate the effects of exercise on muscle mass 23. Another study has shown that exercise-induced inflammation, via the inflammatory regulator nuclear factor-κB, mediates suppression of the transcription factor paired box 7, and improves muscle mass in cancer cachexia 130. These findings indicate the benefits of using exercise to activate the immune system. Unfortunately, many studies on cancer cachexia have noted no changes in muscle size in rodents with cachexia, but exercise before tumor inoculation resulted in improved outcome134. This further strengthens the rationale for instituting physical activity or exercise regimens prior to the development of cachexia to reduce its severity.
To better translate these findings, it is vital that the exercise prescribed to mice be purposeful and that there is compatibility between mouse and human responses to exercise and adaptation. Both exercise and tumors are stressors, and, when adapting to exercise, an organism can handle only so much stress before adaptations become unfavorable. It is therefore important to standardize animal-training protocols to evaluate dosages of activity needed for beneficial results without adding too many additional stressors to the cancer patients. Additionally, PA and exercise are different by definition 135. Physical activity refers to any movement that results in energy expenditure; whereas, exercise is a subset of physical activity that aims to maintain physical fitness. It is critical to keep this in mind so that findings concluded in mice can be appropriately interpreted and translated to PA or exercise therapies in humans. Although challenging, by closing this gap, this field can move toward recommendations for optimal exercise prescription for cancer patients, and even toward identifying pharmacological exercise mimetics for cancer patients who cannot or will not exercise.
Clinical Trials Investigating Physical Activity Regimens for Pancreatic Cancer Patients
Epidemiological data indicates a protective role of PA for patients with cancer 136–139. Nonetheless, only a few clinical trials have incorporated PA interventions for patients diagnosed with PDAC which are summarized in Tables 1–5 depending on the patient setting. Among all available clinical trials that met the search criteria, 14 evaluate PA in pre-operative patients, 13 evaluate PA in post-operative patients, 3 evaluate PA in patients with advanced cancer, 7 evaluate PA in patients undergoing chemotherapy or radiotherapy, and 7 did not specified the patient setting (Figure 3A). Different types of PA intervention are used in these trials, 4 use aerobic exercise, 3 use resistance training, and 10 use both aerobic exercise and resistance training (Figure 3B). Unfortunately, the vast majority (23 of 40) do not specify the type of PA intervention which may make it difficult late to make comparisons among trials.
Figure 3:
Summary of the clinical trials. (A) Times of intervention and (B) Types of exercise using in clinical trials. Created with Graphpad Prism 9
Of all the clinical trials, only one trial that involves exercise and pancreatic cancer is a phase 4 trial (Table 1, NCT03642093). The goal of the trial is to evaluate whether a four-week nutritional and PA (walking and inspiratory muscle training) intervention program can alter frailty in upper gastrointestinal surgical oncology patients. Based on the results of the preclinical studies in mice and humans in addition to this phase 4 trial, we expect more interventions like this to become part of approved cancer treatment in the near future. Moreover, we expect to see more unified experimental design among clinical trials to determine the efficacy of PA on PDAC patients.
Conclusions
Although there are pieces of evidence showing that PA improves quality of life in PDAC patients and survivors and that PA potentially protects against PDAC growth, the relationship between PA and PDAC-risk reduction remains unexplored 140. The lack of long-term epidemiologic evidence that supports the beneficial role of PA in PDAC has been hindered by the limited investigations and the low percentage of the population that could be recruited for studies in this area.
Well-designed preclinical studies need to be developed to generate stronger evidence of the role of PA in PDAC. This will be important for justifying the need to increase the number of clinical trials that target PA and the need to recommend PA as a prevention strategy or adjuvant intervention in PDAC. These studies should implement relevant disease mouse models that recapitulate the human disease and PA regimens that can be translated into patients. Moreover, the majority of current epidemiological studies of exercise are undertaken in higher-income regions, and research with more diverse ethnicity and genetic ancestry is needed for evidence based guidelines 140.
PA may not be feasible for some elder or weaker patients. Hence, finding alternative strategies to mimic the effects of PA are needed. Electrical stimulation of individual or collective muscle groups can improve general muscle strength 141, 142 and may be a valuable substitution for exercise in more frail populations 143. However, future studies related to the safety and efficacy of electrical stimulation in PDAC patients with or without cachexia are needed before it can become a viable treatment regimen.
Overall, there is abundant evidence that PA promotes health, and it is generally advised for cancer patients 140. Since PA is usually combined with other supportive cares such as nutritional interventions in cancer patients, these can create confounding factors that make it difficult for us to understand the sole effects of PA on PDAC patients that can then translate the information and specific recommendations for PDAC patients and at-risk individuals. However, because PA is potentially a cost-effective intervention, further research in this area should be encouraged.
Table 3.
Clinical Trials of PDAC and Physical Activity for Patients with Advanced Cancer (N/A: Not applicable)
| Study Title | Conditions & Diseases | Interventions | Study Population | Primary Outcome Measure | Phase | ID Number |
|---|---|---|---|---|---|---|
|
| ||||||
| Evaluation of an Adapted Physical Activity Program in Patients With Unresectable Pancreatic Cancer | Unresectable Locally Advanced Cancer, Metastatic Pancreatic Cancer | Adapted Physical Activity program | 317 | Fatigue, QOL | N/A | NCT02184663 151 |
| Ambulatory Measurement of Physical Activity in Pancreatic Cancer Patients | Unresectable Pancreatic Adenocarcinoma | Wrist-worn Accelerometer, Auto-questionnaires | 30 | Average time wearing the accelerometer, in hours | N/A | NCT03490604 |
| Patient Activation Through Counseling, Exercise and Mobilization | Pancreas Cancer, Biliary Tract Cancer, Non Small Cell Lung Cancer, Advanced Cancer | Multimodal and Exercise-based Intervention | 99 | Lower body strength measured with the 30-second chair stand test | N/A | NCT03411200 152 |
Table 4.
Clinical Trials of PDAC and Physical Activity for Patients Undergoing Chemotherapy or Radiotherapy (N/A: Not applicable)
| Study Title | Conditions & Diseases | Interventions | Study Population | Primary Outcome Measure | Phase | ID Number |
|---|---|---|---|---|---|---|
|
| ||||||
| Cardiopulmonary Exercise Testing: An Assessment of Patients Fitness for Palliative Chemotherapy for Pancreatic Cancer | Pancreatic Cancer | Cardiopulmonary Exercise Test | 100 | Survival | NCT03215459 | |
| Ambulatory Measurement of Physical Activity in Pancreatic Cancer Patients | Unresectable Pancreatic Adenocarcinoma | Wrist-worn Accelerometer Auto-questionnaires | 30 | Average time wearing the accelerometer, in hours | N/A | NCT03490604 |
| Randomised Trial Evaluating the Benefit of a Fitness Tracker Based Workout During Radiotherapy | Bronchial Carcinoma, Esophageal Carcinoma, Tumor of the Brain, Head and Neck Cancer, Pancreas Cancer, Sarcoma, Cervix Uteri Cancer | Fitness Tracker Based Activity Training, Booklet “physical training, exercise and cancer” and an Introduction About Physical Activity During Cancer Therapy | 201 | Evaluation of the impact of an Activity tracker based Fitness programme on the Qualitiy of Life after oncological Therapy | N/A | NCT04517019 |
| Resistance Training Intervention to Improve Physical Function in Patients With Pancreatic Cancer Receiving Combination Chemotherapy or Have Undergone Surgery, PancStrength Study | Advanced Pancreatic Adenocarcinoma, Pancreatic Adenocarcinoma, Stage III Pancreatic Cancer AJCC v8, Stage IV Pancreatic Cancer AJCC v8 | Educational Intervention Quality-of-Life Assessment, Questionnaire Administration, Resistance Training | 75 | Safety of the tele-resistance training (RT) program, Post-exercise muscle soreness, Body composition, Aerobic fitness | N/A | NCT04837118 |
| Exercise to Reduce Chemotherapy-Induced Peripheral Neuropathy | Gastrointestinal Cancer, Colorectal Cancer, Pancreatic Cancer, Gastric Cancer, Stomach Cancer, Esophageal Cancer | MI-Walk Intervention Physical Activity Education Pamphlet | 54 | Sensory neuropathy at 8 weeks | N/A | NCT03515356 |
| Resilience and Exercise in Advanced Cancer Treatment | Pancreatic Adenocarcinoma, Gastric Adenocarcinoma, Adenocarcinoma of the Gastroesophageal Junction | Band Together, Exercise Education | 14 | Feasibility of Large-Scale Trial: Adherence & Contamination | N/A | NCT02680990 |
| A pilot study to explore efficacy of outpatient cancer rehabilitation for advanced lung or pancreatic cancer under palliative chemotherapy | Advanced or Recurrenced Lung and Pancreatic Cancer | Nutritional Counseling, Home-based Exercise Intervention, Physical Activity Intervention, Usual Care | 30 | SPPB(Short Physical Performance Battery) | N/A | JPRN-UMIN000036771 |
Funding sources:
Research reported in this publication was supported by The National Center for Advancing Translational Sciences under award number TL1TR002735 (KG), and the Pelotonia Fellowship Program (to M. C-T, AL, and VP). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Center For Advancing Translational Sciences or the National Institutes of Health. Any opinions, findings, and conclusions expressed in this material are those of the author(s) and do not necessarily reflect those of the Pelotonia Fellowship Program or The Ohio State University.
Footnotes
Conflict of interest/disclosures: None
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Siegel RL, Miller KD, Fuchs HE, Jemal A: Cancer statistics, 2021. CA: a cancer journal for clinicians 2021; 71: 7–33. [DOI] [PubMed] [Google Scholar]
- 2.Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM: Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the united states. Cancer Res 2014; 74: 2913–2921. [DOI] [PubMed] [Google Scholar]
- 3.Sengupta R, Honey K: Aacr cancer progress report 2018: Harnessing research discoveries for patient benefit, AACR, 2018 [DOI] [PubMed] [Google Scholar]
- 4.Michaud DS, Giovannucci E, Willett WC, Colditz GA, Stampfer MJ, Fuchs CS: Physical activity, obesity, height, and the risk of pancreatic cancer. Jama 2001; 286: 921–929. [DOI] [PubMed] [Google Scholar]
- 5.Patel AV, Rodriguez C, Bernstein L, Chao A, Thun MJ, Calle EE: Obesity, recreational physical activity, and risk of pancreatic cancer in a large u.S. Cohort. Cancer Epidemiol Biomarkers Prev 2005; 14: 459–466. [DOI] [PubMed] [Google Scholar]
- 6.Berrington de Gonzalez A, Sweetland S, Spencer E: A meta-analysis of obesity and the risk of pancreatic cancer. Br J Cancer 2003; 89: 519–523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Frankel PH: Obesity and cancer. N Engl J Med 2003; 349: 502–504; author reply 502–504. [PubMed] [Google Scholar]
- 8.Larsson SC, Orsini N, Wolk A: Body mass index and pancreatic cancer risk: A meta-analysis of prospective studies. Int J Cancer 2007; 120: 1993–1998. [DOI] [PubMed] [Google Scholar]
- 9.Stocks T, Rapp K, Bjorge T, Manjer J, Ulmer H, Selmer R et al. : Blood glucose and risk of incident and fatal cancer in the metabolic syndrome and cancer project (me-can): Analysis of six prospective cohorts. PLoS Med 2009; 6: e1000201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Wang Q, Zhou W: Roles and molecular mechanisms of physical exercise in cancer prevention and treatment. J Sport Health Sci 2021; 10: 201–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Koelwyn GJ, Quail DF, Zhang X, White RM, Jones LW: Exercise-dependent regulation of the tumour microenvironment. Nat Rev Cancer 2017; 17: 620–632. [DOI] [PubMed] [Google Scholar]
- 12.Ruiz-Casado A, Martin-Ruiz A, Perez LM, Provencio M, Fiuza-Luces C, Lucia A: Exercise and the hallmarks of cancer. Trends Cancer 2017; 3: 423–441. [DOI] [PubMed] [Google Scholar]
- 13.Pedersen L, Idorn M, Olofsson GH, Lauenborg B, Nookaew I, Hansen RH et al. : Voluntary running suppresses tumor growth through epinephrine- and il-6-dependent nk cell mobilization and redistribution. Cell metabolism 2016; 23: 554–562. [DOI] [PubMed] [Google Scholar]
- 14.Matthews CE, Moore SC, Arem H, Cook MB, Trabert B, Hakansson N et al. : Amount and intensity of leisure-time physical activity and lower cancer risk. J Clin Oncol 2020; 38: 686–697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Moore SC, Lee IM, Weiderpass E, Campbell PT, Sampson JN, Kitahara CM et al. : Association of leisure-time physical activity with risk of 26 types of cancer in 1.44 million adults. JAMA Intern Med 2016; 176: 816–825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Ainsworth BE, Haskell WL, Herrmann SD, Meckes N, Bassett DR Jr., Tudor-Locke C et al. : 2011. compendium of physical activities: A second update of codes and met values. Med Sci Sports Exerc 2011; 43: 1575–1581. [DOI] [PubMed] [Google Scholar]
- 17.Organization WH: Global recommendations on physical activity for health. Geneva, 2010. ISBN: 9789241599979 http://wwwwhoint/dietphysicalactivity/publications/9789241599979/en/ Accessed 2017; 1: 60. [Google Scholar]
- 18.Campbell KL, Winters-Stone KM, Wiskemann J, May AM, Schwartz AL, Courneya KS et al. : Exercise guidelines for cancer survivors: Consensus statement from international multidisciplinary roundtable. Med Sci Sports Exerc 2019; 51: 2375–2390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.von Haehling S, Anker SD: Cachexia as a major underestimated and unmet medical need: Facts and numbers. J Cachexia Sarcopenia Muscle 2010; 1: 1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hendifar AE, Chang JI, Huang BZ, Tuli R, Wu BU: Cachexia, and not obesity, prior to pancreatic cancer diagnosis worsens survival and is negated by chemotherapy. J Gastrointest Oncol 2018; 9: 17–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Mattox TW: Cancer cachexia: Cause, diagnosis, and treatment. Nutr Clin Pract 2017; 32: 599–606. [DOI] [PubMed] [Google Scholar]
- 22.Hardee JP, Counts BR, Carson JA: Understanding the role of exercise in cancer cachexia therapy. Am J Lifestyle Med 2019; 13: 46–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Pigna E, Berardi E, Aulino P, Rizzuto E, Zampieri S, Carraro U et al. : Aerobic exercise and pharmacological treatments counteract cachexia by modulating autophagy in colon cancer. Sci Rep 2016; 6: 26991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Theilen NT, Jeremic N, Weber GJ, Tyagi SC: Exercise preconditioning diminishes skeletal muscle atrophy after hindlimb suspension in mice. J Appl Physiol (1985) 2018; 125: 999–1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Uster A, Ruehlin M, Mey S, Gisi D, Knols R, Imoberdorf R et al. : Effects of nutrition and physical exercise intervention in palliative cancer patients: A randomized controlled trial. Clin Nutr 2018; 37: 1202–1209. [DOI] [PubMed] [Google Scholar]
- 26.Tobberup R, Carus A, Rasmussen HH, Falkmer UG, Jorgensen MG, Schmidt EB et al. : Feasibility of a multimodal intervention on malnutrition in patients with lung cancer during primary anti-neoplastic treatment. Clin Nutr 2021; 40: 525–533. [DOI] [PubMed] [Google Scholar]
- 27.Grande AJ, Silva V, Sawaris Neto L, Teixeira Basmage JP, Peccin MS, Maddocks M: Exercise for cancer cachexia in adults. Cochrane Database Syst Rev 2021; 3: CD010804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Leal LG, Lopes MA, Peres SB, Batista ML Jr.: Exercise training as therapeutic approach in cancer cachexia: A review of potential anti-inflammatory effect on muscle wasting. Front Physiol 2020; 11: 570170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Midha S, Chawla S, Garg PK: Modifiable and non-modifiable risk factors for pancreatic cancer: A review. Cancer Lett 2016; 381: 269–277. [DOI] [PubMed] [Google Scholar]
- 30.Klein AP: Pancreatic cancer epidemiology: Understanding the role of lifestyle and inherited risk factors. Nat Rev Gastroenterol Hepatol 2021; 18: 493–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Alsamarrai A, Das SL, Windsor JA, Petrov MS: Factors that affect risk for pancreatic disease in the general population: A systematic review and meta-analysis of prospective cohort studies. Clin Gastroenterol Hepatol 2014; 12: 1635–1644 e1635; quiz e1103. [DOI] [PubMed] [Google Scholar]
- 32.Yadav D, Whitcomb DC: The role of alcohol and smoking in pancreatitis. Nat Rev Gastroenterol Hepatol 2010; 7: 131–145. [DOI] [PubMed] [Google Scholar]
- 33.Yadav D, Lowenfels AB: The epidemiology of pancreatitis and pancreatic cancer. Gastroenterology 2013; 144: 1252–1261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Weissman S, Takakura K, Eibl G, Pandol SJ, Saruta M: The diverse involvement of cigarette smoking in pancreatic cancer development and prognosis. Pancreas 2020; 49: 612–620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Stolzenberg-Solomon RZ, Pietinen P, Taylor PR, Virtamo J, Albanes D: A prospective study of medical conditions, anthropometry, physical activity, and pancreatic cancer in male smokers (finland). Cancer Causes Control 2002; 13: 417–426. [DOI] [PubMed] [Google Scholar]
- 36.Soler M, Chatenoud L, La Vecchia C, Franceschi S, Negri E: Diet, alcohol, coffee and pancreatic cancer: Final results from an italian study. Eur J Cancer Prev 1998; 7: 455–460. [DOI] [PubMed] [Google Scholar]
- 37.Ye W, Lagergren J, Weiderpass E, Nyren O, Adami HO, Ekbom A: Alcohol abuse and the risk of pancreatic cancer. Gut 2002; 51: 236–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Genkinger JM, Spiegelman D, Anderson KE, Bergkvist L, Bernstein L, van den Brandt PA et al. : Alcohol intake and pancreatic cancer risk: A pooled analysis of fourteen cohort studies. Cancer Epidemiol Biomarkers Prev 2009; 18: 765–776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Jiao L, Silverman DT, Schairer C, Thiebaut AC, Hollenbeck AR, Leitzmann MF et al. : Alcohol use and risk of pancreatic cancer: The nih-aarp diet and health study. Am J Epidemiol 2009; 169: 1043–1051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Gapstur SM, Jacobs EJ, Deka A, McCullough ML, Patel AV, Thun MJ: Association of alcohol intake with pancreatic cancer mortality in never smokers. Arch Intern Med 2011; 171: 444–451. [DOI] [PubMed] [Google Scholar]
- 41.Lucenteforte E, La Vecchia C, Silverman D, Petersen GM, Bracci PM, Ji BT et al. : Alcohol consumption and pancreatic cancer: A pooled analysis in the international pancreatic cancer case-control consortium (panc4). Ann Oncol 2012; 23: 374–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Bagnardi V, Rota M, Botteri E, Tramacere I, Islami F, Fedirko V et al. : Alcohol consumption and site-specific cancer risk: A comprehensive dose-response meta-analysis. Br J Cancer 2015; 112: 580–593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Asahina K, Balog S, Hwang E, Moon E, Wan E, Skrypek K et al. : Moderate alcohol intake promotes pancreatic ductal adenocarcinoma development in mice expressing oncogenic kras. Am J Physiol Gastrointest Liver Physiol 2020; 318: G265–G276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Apte MV, Pirola RC, Wilson JS: Molecular mechanisms of alcoholic pancreatitis. Dig Dis 2005; 23: 232–240. [DOI] [PubMed] [Google Scholar]
- 45.Perreault K, Bauman A, Johnson N, Britton A, Rangul V, Stamatakis E: Does physical activity moderate the association between alcohol drinking and all-cause, cancer and cardiovascular diseases mortality? A pooled analysis of eight british population cohorts. Br J Sports Med 2017; 51: 651–657. [DOI] [PubMed] [Google Scholar]
- 46.Zanini S, Renzi S, Limongi AR, Bellavite P, Giovinazzo F, Bermano G: A review of lifestyle and environment risk factors for pancreatic cancer. Eur J Cancer 2021; 145: 53–70. [DOI] [PubMed] [Google Scholar]
- 47.Rawla P, Thandra KC, Sunkara T: Pancreatic cancer and obesity: Epidemiology, mechanism, and preventive strategies. Clin J Gastroenterol 2019; 12: 285–291. [DOI] [PubMed] [Google Scholar]
- 48.Abbruzzese JL, Andersen DK, Borrebaeck CAK, Chari ST, Costello E, Cruz-Monserrate Z et al. : The interface of pancreatic cancer with diabetes, obesity, and inflammation: Research gaps and opportunities: Summary of a national institute of diabetes and digestive and kidney diseases workshop. Pancreas 2018; 47: 516–525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Eibl G, Cruz-Monserrate Z, Korc M, Petrov MS, Goodarzi MO, Fisher WE et al. : Diabetes mellitus and obesity as risk factors for pancreatic cancer. J Acad Nutr Diet 2018; 118: 555–567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Philip B, Roland CL, Daniluk J, Liu Y, Chatterjee D, Gomez SB et al. : A high-fat diet activates oncogenic kras and cox2 to induce development of pancreatic ductal adenocarcinoma in mice. Gastroenterology 2013; 145: 1449–1458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Wiklund P: The role of physical activity and exercise in obesity and weight management: Time for critical appraisal. J Sport Health Sci 2016; 5: 151–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Incio J, Liu H, Suboj P, Chin SM, Chen IX, Pinter M et al. : Obesity-induced inflammation and desmoplasia promote pancreatic cancer progression and resistance to chemotherapy. Cancer Discov 2016; 6: 852–869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Schauer DP, Feigelson HS, Koebnick C, Caan B, Weinmann S, Leonard AC et al. : Bariatric surgery and the risk of cancer in a large multisite cohort. Ann Surg 2019; 269: 95–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Yadav D, Slivka A, Sherman S, Hawes RH, Anderson MA, Burton FR et al. : Smoking is underrecognized as a risk factor for chronic pancreatitis. Pancreatology 2010; 10: 713–719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Samokhvalov AV, Rehm J, Roerecke M: Alcohol consumption as a risk factor for acute and chronic pancreatitis: A systematic review and a series of meta-analyses. EBioMedicine 2015; 2: 1996–2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Greer JB, Whitcomb DC: Inflammation and pancreatic cancer: An evidence-based review. Curr Opin Pharmacol 2009; 9: 411–418. [DOI] [PubMed] [Google Scholar]
- 57.Kolodecik T, Shugrue C, Ashat M, Thrower EC: Risk factors for pancreatic cancer: Underlying mechanisms and potential targets. Front Physiol 2013; 4: 415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kirkegard J, Mortensen FV, Cronin-Fenton D: Chronic pancreatitis and pancreatic cancer risk: A systematic review and meta-analysis. Am J Gastroenterol 2017; 112: 1366–1372. [DOI] [PubMed] [Google Scholar]
- 59.Li D: Diabetes and pancreatic cancer. Mol Carcinog 2012; 51: 64–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hu CM, Tien SC, Hsieh PK, Jeng YM, Chang MC, Chang YT et al. : High glucose triggers nucleotide imbalance through o-glcnacylation of key enzymes and induces kras mutation in pancreatic cells. Cell metabolism 2019; 29: 1334–1349 e1310. [DOI] [PubMed] [Google Scholar]
- 61.Bryant KL, Mancias JD, Kimmelman AC, Der CJ: Kras: Feeding pancreatic cancer proliferation. Trends Biochem Sci 2014; 39: 91–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Pannala R, Leirness JB, Bamlet WR, Basu A, Petersen GM, Chari ST: Prevalence and clinical profile of pancreatic cancer-associated diabetes mellitus. Gastroenterology 2008; 134: 981–987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Aggarwal G, Kamada P, Chari ST: Prevalence of diabetes mellitus in pancreatic cancer compared to common cancers. Pancreas 2013; 42: 198–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Zhang Z, Qin W, Sun Y: Contribution of biomarkers for pancreatic cancer-associated new-onset diabetes to pancreatic cancer screening. Pathol Res Pract 2018; 214: 1923–1928. [DOI] [PubMed] [Google Scholar]
- 65.Maitra A, Sharma A, Brand RE, Van Den Eeden SK, Fisher WE, Hart PA et al. : A prospective study to establish a new-onset diabetes cohort: From the consortium for the study of chronic pancreatitis, diabetes, and pancreatic cancer. Pancreas 2018; 47: 1244–1248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Schellenberg ES, Dryden DM, Vandermeer B, Ha C, Korownyk C: Lifestyle interventions for patients with and at risk for type 2 diabetes: A systematic review and meta-analysis. Ann Intern Med 2013; 159: 543–551. [DOI] [PubMed] [Google Scholar]
- 67.Chen L, Pei JH, Kuang J, Chen HM, Chen Z, Li ZW et al. : Effect of lifestyle intervention in patients with type 2 diabetes: A meta-analysis. Metabolism 2015; 64: 338–347. [DOI] [PubMed] [Google Scholar]
- 68.Iwasaki M, Kudo A, Asahi K, Machii N, Iseki K, Satoh H et al. : Fast walking is a preventive factor against new-onset diabetes mellitus in a large cohort from a japanese general population. Sci Rep 2021; 11: 716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Carstensen B, Read SH, Friis S, Sund R, Keskimaki I, Svensson AM et al. : Cancer incidence in persons with type 1 diabetes: A five-country study of 9,000 cancers in type 1 diabetic individuals. Diabetologia 2016; 59: 980–988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Physical activity guidelines advisory committee report, 2008. To the secretary of health and human services. Part a: Executive summary. Nutr Rev 2009; 67: 114–120. [DOI] [PubMed] [Google Scholar]
- 71.Arem H, Moore SC, Patel A, Hartge P, Berrington de Gonzalez A, Visvanathan K et al. : Leisure time physical activity and mortality: A detailed pooled analysis of the dose-response relationship. JAMA Intern Med 2015; 175: 959–967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Farris MS, Mosli MH, McFadden AA, Friedenreich CM, Brenner DR: The association between leisure time physical activity and pancreatic cancer risk in adults: A systematic review and meta-analysis. Cancer Epidemiol Biomarkers Prev 2015; 24: 1462–1473. [DOI] [PubMed] [Google Scholar]
- 73.Zhang J, Dhakal IB, Gross MD, Lang NP, Kadlubar FF, Harnack LJ et al. : Physical activity, diet, and pancreatic cancer: A population-based, case-control study in minnesota. Nutr Cancer 2009; 61: 457–465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Keum N, Bao Y, Smith-Warner SA, Orav J, Wu K, Fuchs CS et al. : Association of physical activity by type and intensity with digestive system cancer risk. JAMA Oncol 2016; 2: 1146–1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Song M, Giovannucci E: Preventable incidence and mortality of carcinoma associated with lifestyle factors among white adults in the united states. JAMA Oncol 2016; 2: 1154–1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Naudin S, Viallon V, Hashim D, Freisling H, Jenab M, Weiderpass E et al. : Healthy lifestyle and the risk of pancreatic cancer in the epic study. Eur J Epidemiol 2020; 35: 975–986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Roebuck BD, McCaffrey J, Baumgartner KJ: Protective effects of voluntary exercise during the postinitiation phase of pancreatic carcinogenesis in the rat. Cancer Res 1990; 50: 6811–6816. [PubMed] [Google Scholar]
- 78.Zheng X, Cui XX, Huang MT, Liu Y, Shih WJ, Lin Y et al. : Inhibitory effect of voluntary running wheel exercise on the growth of human pancreatic panc-1 and prostate pc-3 xenograft tumors in immunodeficient mice. Oncol Rep 2008; 19: 1583–1588. [PMC free article] [PubMed] [Google Scholar]
- 79.Florez Bedoya CA, Cardoso ACF, Parker N, Ngo-Huang A, Petzel MQ, Kim MP et al. : Exercise during preoperative therapy increases tumor vascularity in pancreatic tumor patients. Sci Rep 2019; 9: 13966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Schadler KL, Thomas NJ, Galie PA, Bhang DH, Roby KC, Addai P et al. : Tumor vessel normalization after aerobic exercise enhances chemotherapeutic efficacy. Oncotarget 2016; 7: 65429–65440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Ashcraft KA, Peace RM, Betof AS, Dewhirst MW, Jones LW: Efficacy and mechanisms of aerobic exercise on cancer initiation, progression, and metastasis: A critical systematic review of in vivo preclinical data. Cancer Res 2016; 76: 4032–4050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Hojman P, Gehl J, Christensen JF, Pedersen BK: Molecular mechanisms linking exercise to cancer prevention and treatment. Cell metabolism 2018; 27: 10–21. [DOI] [PubMed] [Google Scholar]
- 83.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. Exp Biol Med (Maywood) 2007; 232: 1289–1299. [DOI] [PubMed] [Google Scholar]
- 84.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. J Hepatol 2015; 62: 1296–1303. [DOI] [PubMed] [Google Scholar]
- 85.Dimeo FC, Stieglitz RD, Novelli-Fischer U, Fetscher S, Keul J: Effects of physical activity on the fatigue and psychologic status of cancer patients during chemotherapy. Cancer 1999; 85: 2273–2277. [PubMed] [Google Scholar]
- 86.Stevinson C, Lawlor DA, Fox KR: Exercise interventions for cancer patients: Systematic review of controlled trials. Cancer Causes Control 2004; 15: 1035–1056. [DOI] [PubMed] [Google Scholar]
- 87.Galvao DA, Newton RU: Review of exercise intervention studies in cancer patients. J Clin Oncol 2005; 23: 899–909. [DOI] [PubMed] [Google Scholar]
- 88.Knols R, Aaronson NK, Uebelhart D, Fransen J, Aufdemkampe G: Physical exercise in cancer patients during and after medical treatment: A systematic review of randomized and controlled clinical trials. J Clin Oncol 2005; 23: 3830–3842. [DOI] [PubMed] [Google Scholar]
- 89.Mustian KM, Morrow GR, Carroll JK, Figueroa-Moseley CD, Jean-Pierre P, Williams GC: Integrative nonpharmacologic behavioral interventions for the management of cancer-related fatigue. Oncologist 2007; 12 Suppl 1: 52–67. [DOI] [PubMed] [Google Scholar]
- 90.Mustian KM, Peppone LJ, Palesh OG, Janelsins MC, Mohile SG, Purnell JQ et al. : Exercise and cancer-related fatigue. US Oncol 2009; 5: 20–23. [PMC free article] [PubMed] [Google Scholar]
- 91.Mishra SI, Scherer RW, Snyder C, Geigle P, Gotay C: Are exercise programs effective for improving health-related quality of life among cancer survivors? A systematic review and meta-analysis. Oncol Nurs Forum 2014; 41: E326–342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Witlox L, Hiensch AE, Velthuis MJ, Steins Bisschop CN, Los M, Erdkamp FLG et al. : Four-year effects of exercise on fatigue and physical activity in patients with cancer. BMC Med 2018; 16: 86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Scott JM, Zabor EC, Schwitzer E, Koelwyn GJ, Adams SC, Nilsen TS et al. : Efficacy of exercise therapy on cardiorespiratory fitness in patients with cancer: A systematic review and meta-analysis. J Clin Oncol 2018; 36: 2297–2305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Grimmett C, Corbett T, Brunet J, Shepherd J, Pinto BM, May CR et al. : Systematic review and meta-analysis of maintenance of physical activity behaviour change in cancer survivors. Int J Behav Nutr Phys Act 2019; 16: 37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Vedie AL, Neuzillet C: Pancreatic cancer: Best supportive care. Presse Med 2019; 48: e175–e185. [DOI] [PubMed] [Google Scholar]
- 96.van Waart H, Stuiver MM, van Harten WH, Geleijn E, Kieffer JM, Buffart LM et al. : Effect of low-intensity physical activity and moderate- to high-intensity physical exercise during adjuvant chemotherapy on physical fitness, fatigue, and chemotherapy completion rates: Results of the paces randomized clinical trial. J Clin Oncol 2015; 33: 1918–1927. [DOI] [PubMed] [Google Scholar]
- 97.Cormie P, Spry N, Jasas K, Johansson M, Yusoff IF, Newton RU et al. : Exercise as medicine in the management of pancreatic cancer: A case study. Med Sci Sports Exerc 2014; 46: 664–670. [DOI] [PubMed] [Google Scholar]
- 98.Ngo-Huang A, Parker NH, Wang X, Petzel MQB, Fogelman D, Schadler KL et al. : Home-based exercise during preoperative therapy for pancreatic cancer. Langenbecks Arch Surg 2017; 402: 1175–1185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Ngo-Huang A, Parker NH, Bruera E, Lee RE, Simpson R, O’Connor DP et al. : Home-based exercise prehabilitation during preoperative treatment for pancreatic cancer is associated with improvement in physical function and quality of life. Integr Cancer Ther 2019; 18: 1534735419894061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.T NAT, L SIH, M H Ft B: Exercise improves patient outcomes in advanced pancreatic cancer patient during medical treatment. Pancreatic Disorders & Therapy 2018; 08. [Google Scholar]
- 101.Fagevik Olsen M, Becovic S, Dean E: Short-term effects of mobilization on oxygenation in patients after open surgery for pancreatic cancer: A randomized controlled trial. BMC Surg 2021; 21: 185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Steindorf K, Clauss D, Tjaden C, Hackert T, Herbolsheimer F, Bruckner T et al. : Quality of life, fatigue, and sleep problems in pancreatic cancer patients-a randomized trial on the effects of exercise. Dtsch Arztebl Int 2019; 116: 471–478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Wiskemann J, Clauss D, Tjaden C, Hackert T, Schneider L, Ulrich CM et al. : Progressive resistance training to impact physical fitness and body weight in pancreatic cancer patients: A randomized controlled trial. Pancreas 2019; 48: 257–266. [DOI] [PubMed] [Google Scholar]
- 104.Luo H, Galvao DA, Newton RU, Lopez P, Tang C, Fairman CM et al. : Exercise medicine in the management of pancreatic cancer: A systematic review. Pancreas 2021; 50: 280–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.O’Connor D, Brown M, Eatock M, Turkington RC, Prue G: Exercise efficacy and prescription during treatment for pancreatic ductal adenocarcinoma: A systematic review. BMC Cancer 2021; 21: 43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Harrington CB, Hansen JA, Moskowitz M, Todd BL, Feuerstein M: It’s not over when it’s over: Long-term symptoms in cancer survivors--a systematic review. Int J Psychiatry Med 2010; 40: 163–181. [DOI] [PubMed] [Google Scholar]
- 107.Packel L, Claghorn KV, Dekerlegand J: Cancer-related fatigue and deconditioning: A program evaluation. Rehabilitation Oncology 2006; 24: 3–8. [Google Scholar]
- 108.Rock CL, Doyle C, Demark-Wahnefried W, Meyerhardt J, Courneya KS, Schwartz AL et al. : Nutrition and physical activity guidelines for cancer survivors. CA: a cancer journal for clinicians 2012; 62: 243–274. [DOI] [PubMed] [Google Scholar]
- 109.Jones LW, Eves ND, Haykowsky M, Freedland SJ, Mackey JR: Exercise intolerance in cancer and the role of exercise therapy to reverse dysfunction. Lancet Oncol 2009; 10: 598–605. [DOI] [PubMed] [Google Scholar]
- 110.Arthur AE, Delk A, Demark-Wahnefried W, Christein JD, Contreras C, Posey JA 3rd et al. : Pancreatic cancer survivors’ preferences, barriers, and facilitators related to physical activity and diet interventions. J Cancer Surviv 2016; 10: 981–989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Loprinzi PD, Lee H, Cardinal BJ: Objectively measured physical activity among us cancer survivors: Considerations by weight status. J Cancer Surviv 2013; 7: 493–499. [DOI] [PubMed] [Google Scholar]
- 112.Parker NH, Basen-Engquist K, Rubin ML, Li Y, Prakash L, Ngo-Huang A et al. : Factors influencing exercise following pancreatic tumor resection. Ann Surg Oncol 2021; 28: 2299–2309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Woods JA, Davis JM, Kohut ML, Ghaffar A, Mayer EP, Pate RR: Effects of exercise on the immune response to cancer. Med Sci Sports Exerc 1994; 26: 1109–1115. [PubMed] [Google Scholar]
- 114.Fernandes G, Venkatraman JT: Possible mechanisms through which dietary lipids, calorie restriction, and exercise modulate breast cancer. Adv Exp Med Biol 1992; 322: 185–201. [DOI] [PubMed] [Google Scholar]
- 115.Kim YJ, Kim HJ, Lee WJ, Seong JK: A comparison of the metabolic effects of treadmill and wheel running exercise in mouse model. Lab Anim Res 2020; 36: 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Conner JD, Wolden-Hanson T, Quinn LS: Assessment of murine exercise endurance without the use of a shock grid: An alternative to forced exercise. J Vis Exp 2014: e51846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Renz BW, Takahashi R, Tanaka T, Macchini M, Hayakawa Y, Dantes Z et al. : Beta2 adrenergic-neurotrophin feedforward loop promotes pancreatic cancer. Cancer Cell 2018; 33: 75–90 e77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Landskron G, De la Fuente M, Thuwajit P, Thuwajit C, Hermoso MA: Chronic inflammation and cytokines in the tumor microenvironment. J Immunol Res 2014; 2014: 149185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Jud C, Schmutz I, Hampp G, Oster H, Albrecht U: A guideline for analyzing circadian wheel-running behavior in rodents under different lighting conditions. Biol Proced Online 2005; 7: 101–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.McDonald PG, Antoni MH, Lutgendorf SK, Cole SW, Dhabhar FS, Sephton SE et al. : A biobehavioral perspective of tumor biology. Discov Med 2005; 5: 520–526. [PMC free article] [PubMed] [Google Scholar]
- 121.Lerman I, Harrison BC, Freeman K, Hewett TE, Allen DL, Robbins J et al. : Genetic variability in forced and voluntary endurance exercise performance in seven inbred mouse strains. J Appl Physiol (1985) 2002; 92: 2245–2255. [DOI] [PubMed] [Google Scholar]
- 122.Almeida PW, Gomes-Filho A, Ferreira AJ, Rodrigues CE, Dias-Peixoto MF, Russo RC et al. : Swim training suppresses tumor growth in mice. J Appl Physiol (1985) 2009; 107: 261–265. [DOI] [PubMed] [Google Scholar]
- 123.Bernstein D: Exercise assessment of transgenic models of human cardiovascular disease. Physiol Genomics 2003; 13: 217–226. [DOI] [PubMed] [Google Scholar]
- 124.Phillips SM, Winett RA: Uncomplicated resistance training and health-related outcomes: Evidence for a public health mandate. Curr Sports Med Rep 2010; 9: 208–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Fairman CM, Focht BC, Lucas AR, Lustberg MB: Effects of exercise interventions during different treatments in breast cancer. J Community Support Oncol 2016; 14: 200–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Tamaki T, Uchiyama S, Nakano S: A weight-lifting exercise model for inducing hypertrophy in the hindlimb muscles of rats. Med Sci Sports Exerc 1992; 24: 881–886. [PubMed] [Google Scholar]
- 127.Barauna VG, Batista ML Jr., Costa Rosa LF, Casarini DE, Krieger JE, Oliveira EM: Cardiovascular adaptations in rats submitted to a resistance-training model. Clin Exp Pharmacol Physiol 2005; 32: 249–254. [DOI] [PubMed] [Google Scholar]
- 128.Khamoui AV, Park BS, Kim DH, Yeh MC, Oh SL, Elam ML et al. : Aerobic and resistance training dependent skeletal muscle plasticity in the colon-26 murine model of cancer cachexia. Metabolism 2016; 65: 685–698. [DOI] [PubMed] [Google Scholar]
- 129.Yeo TP, Burrell SA, Sauter PK, Kennedy EP, Lavu H, Leiby BE et al. : A progressive postresection walking program significantly improves fatigue and health-related quality of life in pancreas and periampullary cancer patients. J Am Coll Surg 2012; 214: 463–475; discussion 475–467. [DOI] [PubMed] [Google Scholar]
- 130.Coletti D, Aulino P, Pigna E, Barteri F, Moresi V, Annibali D et al. : Spontaneous physical activity downregulates pax7 in cancer cachexia. Stem Cells Int 2016; 2016: 6729268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Dethlefsen C, Hansen LS, Lillelund C, Andersen C, Gehl J, Christensen JF et al. : Exercise-induced catecholamines activate the hippo tumor suppressor pathway to reduce risks of breast cancer development. Cancer Res 2017; 77: 4894–4904. [DOI] [PubMed] [Google Scholar]
- 132.Thomas RJ, Kenfield SA, Jimenez A: Exercise-induced biochemical changes and their potential influence on cancer: A scientific review. Br J Sports Med 2017; 51: 640–644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Henderson SE, Makhijani N, Mace TA: Pancreatic cancer-induced cachexia and relevant mouse models. Pancreas 2018; 47: 937–945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Niels T, Tomanek A, Freitag N, Schumann M: Can exercise counteract cancer cachexia? A systematic literature review and meta-analysis. Integr Cancer Ther 2020; 19: 1534735420940414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Caspersen CJ, Powell KE, Christenson GM: Physical activity, exercise, and physical fitness: Definitions and distinctions for health-related research. Public Health Rep 1985; 100: 126–131. [PMC free article] [PubMed] [Google Scholar]
- 136.Friedenreich CM, Orenstein MR: Physical activity and cancer prevention: Etiologic evidence and biological mechanisms. J Nutr 2002; 132: 3456S–3464S. [DOI] [PubMed] [Google Scholar]
- 137.Kruk J, Aboul-Enein HY: Physical activity in the prevention of cancer. Asian Pac J Cancer Prev 2006; 7: 11–21. [PubMed] [Google Scholar]
- 138.Behrens G, Jochem C, Schmid D, Keimling M, Ricci C, Leitzmann MF: Physical activity and risk of pancreatic cancer: A systematic review and meta-analysis. Eur J Epidemiol 2015; 30: 279–298. [DOI] [PubMed] [Google Scholar]
- 139.McTiernan A, Friedenreich CM, Katzmarzyk PT, Powell KE, Macko R, Buchner D et al. : Physical activity in cancer prevention and survival: A systematic review. Med Sci Sports Exerc 2019; 51: 1252–1261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Clinton SK, Giovannucci EL, Hursting SD: The world cancer research fund/american institute for cancer research third expert report on diet, nutrition, physical activity, and cancer: Impact and future directions. J Nutr 2020; 150: 663–671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Martinez-Lopez EJ, Benito-Martinez E, Hita-Contreras F, Lara-Sanchez A, Martinez-Amat A: Effects of electrostimulation and plyometric training program combination on jump height in teenage athletes. J Sports Sci Med 2012; 11: 727–735. [PMC free article] [PubMed] [Google Scholar]
- 142.Kilinc M, Yildirim SA, Tan E: The effects of electrical stimulation and exercise therapy in patients with limb girdle muscular dystrophy. A controlled clinical trial. Neurosciences (Riyadh) 2015; 20: 259–266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Kemmler W, Teschler M, Weissenfels A, Bebenek M, Frohlich M, Kohl M et al. : Effects of whole-body electromyostimulation versus high-intensity resistance exercise on body composition and strength: A randomized controlled study. Evid Based Complement Alternat Med 2016; 2016: 9236809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Lafaro KJ, Raz DJ, Kim JY, Hite S, Ruel N, Varatkar G et al. : Pilot study of a telehealth perioperative physical activity intervention for older adults with cancer and their caregivers. Support Care Cancer 2020; 28: 3867–3876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Parker NH, Lee RE, O’Connor DP, Ngo-Huang A, Petzel MQB, Schadler K et al. : Supports and barriers to home-based physical activity during preoperative treatment of pancreatic cancer: A mixed-methods study. J Phys Act Health 2019; 16: 1113–1122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Sato H, Carvalho G, Sato T, Lattermann R, Matsukawa T, Schricker T: The association of preoperative glycemic control, intraoperative insulin sensitivity, and outcomes after cardiac surgery. J Clin Endocrinol Metab 2010; 95: 4338–4344. [DOI] [PubMed] [Google Scholar]
- 147.Snowden CP, Prentis JM, Anderson HL, Roberts DR, Randles D, Renton M et al. : Submaximal cardiopulmonary exercise testing predicts complications and hospital length of stay in patients undergoing major elective surgery. Ann Surg 2010; 251: 535–541. [DOI] [PubMed] [Google Scholar]
- 148.Luo H, Galvao DA, Newton RU, Tang C, Dean A, Jasas K et al. : Feasibility and efficacy of a multicomponent exercise medicine programme in patients with pancreatic cancer undergoing neoadjuvant therapy (the expan trial): Study protocol of a dual-centre, two-armed phase i randomised controlled trial. BMJ Open Gastroenterol 2021; 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Miura S, Naito T, Mitsunaga S, Omae K, Mori K, Inano T et al. : A randomized phase ii study of nutritional and exercise treatment for elderly patients with advanced non-small cell lung or pancreatic cancer: The nextac-two study protocol. BMC Cancer 2019; 19: 528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Naito T, Mitsunaga S, Miura S, Tatematsu N, Inano T, Mouri T et al. : Feasibility of early multimodal interventions for elderly patients with advanced pancreatic and non-small-cell lung cancer. J Cachexia Sarcopenia Muscle 2019; 10: 73–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Neuzillet C, Vergnault M, Bonnetain F, Hammel P: Rationale and design of the adapted physical activity in advanced pancreatic cancer patients (apacap) gercor (groupe cooperateur multidisciplinaire en oncologie) trial: Study protocol for a randomized controlled trial. Trials 2015; 16: 454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Mikkelsen MK, Lund CM, Vinther A, Tolver A, Ragle AM, Johansen JS et al. : Engaging the older cancer patient; patient activation through counseling, exercise and mobilization - pancreatic, biliary tract and lung cancer (pace-mobil-pbl) - study protocol of a randomized controlled trial. BMC Cancer 2018; 18: 934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Sanders K, Klooster K, Vanfleteren L, Plasqui G, Dingemans AM, Slebos DJ et al. : Effect of bronchoscopic lung volume reduction in advanced emphysema on energy balance regulation. Respiration 2021: 1–8. [DOI] [PubMed] [Google Scholar]
- 154.Yoh K, Nishikawa H, Enomoto H, Ishii N, Iwata Y, Ishii A et al. : Effect of exercise therapy on sarcopenia in pancreatic cancer: A study protocol for a randomised controlled trial. BMJ Open Gastroenterol 2018; 5: e000194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Yoh K, Nishikawa H, Enomoto H, Ishii N, Iwata Y, Ishii A et al. : Implication of exercise interventions on sleep disturbance in patients with pancreatic cancer: A study protocol for a randomised controlled trial. BMJ Open Gastroenterol 2018; 5: e000196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Solheim TS, Laird BJA, Balstad TR, Stene GB, Bye A, Johns N et al. : A randomized phase ii feasibility trial of a multimodal intervention for the management of cachexia in lung and pancreatic cancer. J Cachexia Sarcopenia Muscle 2017; 8: 778–788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Stene GB, Balstad TR, Leer ASM, Bye A, Kaasa S, Fallon M et al. : Deterioration in muscle mass and physical function differs according to weight loss history in cancer cachexia. Cancers (Basel) 2019; 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Balstad TR, Brunelli C, Pettersen CH, Schonberg SA, Skorpen F, Fallon M et al. : Power comparisons and clinical meaning of outcome measures in assessing treatment effect in cancer cachexia: Secondary analysis from a randomized pilot multimodal intervention trial. Front Nutr 2020; 7: 602775. [DOI] [PMC free article] [PubMed] [Google Scholar]



