ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

The roles of diet, physical activity, and body weight in cancer survivors

The roles of diet, physical activity, and body weight in cancer survivors
Literature review current through: Jan 2024.
This topic last updated: May 01, 2023.

INTRODUCTION — With advances in early detection and treatments for cancer, there are approximately 32.6 million people alive globally with a history of cancer (within five years of diagnosis), as of 2012 [1]. Novel methods to improve outcomes, especially those that are also associated with improvements in quality of life and other health benefits, may offer attractive additions to the currently available treatment options.

Obesity and other factors related to energy balance have been linked to outcomes in several cancers, including breast cancer, endometrial cancer, prostate cancer, and colorectal cancer. We will provide an overview of the many studies that have looked at the feasibility and potential benefits of energy balance interventions in survivors of these cancers. In addition, we provide consensus-based recommendations for cancer survivors.

Weight, diet, and physical activity, as they relate to survivors of other cancers, including lung cancer, ovarian cancer, and head and neck cancers, are discussed separately:

(See "Overview of approach to lung cancer survivors", section on 'Addressing risk behaviors'.)

(See "Approach to survivors of epithelial ovarian, fallopian tube, or peritoneal carcinoma", section on 'Promoting a healthy lifestyle'.)

(See "Overview of approach to long-term survivors of head and neck cancer", section on 'Diet and exercise'.)

An overview of cancer survivorship is also discussed separately. (See "Overview of cancer survivorship care for primary care and oncology providers".)

Topics addressing diet, physical activity, and weight in patients without a history of cancer are discussed separately:

(See "Healthy diet in adults" and "Obesity in adults: Dietary therapy".)

(See "Physical activity and exercise in older adults" and "The benefits and risks of aerobic exercise" and "Obesity in adults: Role of physical activity and exercise".)

(See "Overweight and obesity in adults: Health consequences" and "Obesity: Genetic contribution and pathophysiology" and "Obesity in adults: Prevalence, screening, and evaluation" and "Obesity in adults: Overview of management".)

(See "Overview of cancer prevention".)

(See "Factors that modify breast cancer risk in women", section on 'Alcohol use and smoking'.)

ENERGY BALANCE IN CANCER — Diet, physical activity, and weight are collectively considered energy balance factors because they describe the relationship between energy consumed (diet), energy expended (physical activity), and energy stored (adiposity). Obesity, inactivity, poor dietary quality, and the metabolic syndrome are linked to decreased overall and cancer-specific survival in individuals with cancer [2-6]. A positive energy balance results from excess of energy intake relative to the energy expended, which results in increases in the storage of energy and weight gain [7].

The definition of metabolic syndrome is discussed elsewhere. (See "Metabolic syndrome (insulin resistance syndrome or syndrome X)", section on 'Definition'.)

Studies have demonstrated that interventions targeting weight, diet, and physical activity are feasible, can improve quality of life (QOL), and may minimize disease and treatment-related side effects for cancer survivors [3,8,9]. Some preliminary evidence suggests that lifestyle change may also improve prognosis in individuals with early stage cancers [10], but much work still needs to be done to validate this and to determine which types of lifestyle change are most important.

Diet — Prospective studies of diet and chronic diseases have facilitated major advances in our understanding of the contribution of diet to the pathogenesis of disease [11]. They suggest that changes in dietary patterns might help reduce the risk of many common diseases in the United States, including some cancers. The potential impact of dietary patterns upon prognosis in patients with cancer has also been widely studied and is discussed in the sections below, according to disease type. (See 'Breast cancer' below and 'Prostate cancer' below and 'Colorectal cancer' below.)

In a meta-analysis of 117 cohort studies enrolling 209,597 cancer survivors, adherence to a high-quality diet and a prudent, healthy dietary pattern was inversely associated with overall mortality among cancer survivors, whereas a Western dietary pattern was positively associated with overall mortality in this population [12].

The role of specific dietary regimens as a complementary or alternative treatment for cancer is discussed separately. (See "Overview of complementary, alternative, and integrative medicine practices in oncology care, and potential risks and harm".)

Weight — Obesity is a well-established risk factor for many cancers including multiple myeloma, breast, gastrointestinal (eg, colorectal, gastric, liver, pancreatic, gallbladder), kidney, uterine, and ovarian cancers [13-19]. A number of studies have also linked obesity to an increased risk of cancer recurrence and mortality [7,20,21].

The morbidity and mortality associated with being overweight or obese have been known to the medical profession for more than 2000 years [22]. "Overweight" refers to a weight above the "normal" range, which is determined by calculating the body mass index (BMI, defined as the weight in kilograms divided by height in meters squared). Overweight is defined as a BMI of 25 to 29.9 kg/m2, and obesity as a BMI of >30 kg/m2. (See "Definition, epidemiology, and etiology of obesity in children and adolescents" and "Obesity in adults: Prevalence, screening, and evaluation".)

Given the increasing evidence linking obesity and cancer, the American Society of Clinical Oncology made Obesity and Cancer one of its core initiatives in 2013 to 2014 [23]. The Initiative focuses on building awareness of the links between obesity and cancer within the oncology community, building a robust research agenda in this area, and advocating for access to weight management programs for oncology patients.

Physical activity — Exercise is associated with improved survival and patient reported outcomes in most cancer survivors, and it is safe to perform in both the adjuvant and posttreatment settings.

Exercise and prognosis — Observational studies have reported a link between exercise and improved survival in cancer survivors, with most of the data coming from survivors with breast, colorectal, or prostate cancers [2,5,24-31].

Growing evidence also suggests that sedentary behavior, time spent engaged in activities such as TV-watching and computer use, may be an independent risk factor for poor prognosis in cancer survivors as well as for cancer risk in the general population [32,33]. For example, a meta-analysis of 14 studies including individuals with and without cancer demonstrated that sedentary behavior was associated with an increased risk of all-cause mortality (22 percent), cardiovascular mortality (15 percent), cancer mortality (14 percent), and incidence of type 2 diabetes (91 percent) [32]. Some cancer survivors may benefit from physical activity [34] and exercise rehabilitation to prevent or mitigate such risks. (See "Physical rehabilitation for cancer survivors", section on 'Cardiotoxicity'.)

Data for the association between increased physical activity and lower cancer risk in the general population are discussed separately. (See "Overview of cancer prevention", section on 'Physical activity'.)

Exercise and outcomes in cancer survivors — The American College of Sports Medicine (ACSM) has reviewed data from multiple randomized trials and concluded that exercise could safely be performed in both the adjuvant and posttreatment settings [24,29].

Additionally, the ACSM evaluated data on the impact of exercise interventions on patient-reported outcomes in cancer survivors [29]. They concluded that there was strong evidence that exercise (optimally combined aerobic exercise and resistance training) led to reductions in anxiety, depressive symptoms, and cancer-related fatigue, and improvements in health-related QOL and physical function [24,25,29,35-39]. While the exercise regimens from trials for each outcome differ slightly, general recommendations would be at least 30 minutes of aerobic exercise three times per week and resistance training twice a week. Additionally, supervised resistance training led to reductions in lymphedema symptoms in breast cancer survivors. Notably, the majority of the data were derived from studies in breast cancer survivors. (See "Cancer-related fatigue: Treatment", section on 'Exercise'.)

EVIDENCE RELATED TO SPECIFIC CANCERS

Breast cancer

Diet — Some evidence suggests that reducing dietary fat intake after a cancer diagnosis could improve breast cancer outcomes in cancer survivors, but the data have not been consistent, and dietary modification is not a standard part of adjuvant therapy for women with breast cancer at this time.

Studies have focused both upon intake of specific dietary nutrients as well as on broader dietary patterns (eg, ingestion of a fruit and vegetable-based diet versus a diet high in processed foods, red meat, and fat) [40]. However, any relationships between breast cancer outcomes and any dietary pattern or intake of a particular nutrient have been inconsistent, especially after adjustment for body weight and other related factors. Examples of meta-analyses and large-scale reviews are summarized below:

A 2016 meta-analysis of 41 observational studies found an association between ingestion of a prudent dietary pattern (a diet rich fruits, vegetables, and whole grains and low in red and processed meats) and decreased risk of overall mortality (relative risk [RR] 0.76, 95% CI 0.71-0.89) [12]. Conversely, the analysis showed ingestion of a Western-pattern diet (a diet rich in red meat, processed foods, and sugary snacks) was associated with a significantly higher risk of overall mortality (RR 1.44, 95% CI 1.17-1.77). Alcohol use after breast cancer diagnosis was also associated with an increased risk of breast cancer recurrence (RR 1.21, 95% CI 1.06-1.39) but not overall mortality (RR 0.94, 95% CI 0.85-1.04).

The World Cancer Research Fund (WCRF) reviewed the evidence and concluded that lower fat intake, particularly saturated fat, and higher intake of food containing fiber and soy are associated with lower breast cancer mortality. However, the group concluded that it was not possible to determine whether these effects were causative. WCRF therefore did not generate specific dietary recommendations for breast cancer survivors, instead suggesting that breast cancer survivors follow the WCRF dietary recommendations for cancer prevention [41].

Two large-scale randomized trials have examined the impact of dietary modification on disease outcomes in early stage breast cancer but with differing results:

The Women's Interventional Nutrition Study (WINS) randomized 2437 women with stages I to IIIa breast cancer to a low-fat dietary intervention or usual care control group [10]. After a median follow-up of 5.6 years, dietary intervention improved disease-free survival (DFS; hazard ratio [HR] 0.76, 95% CI 0.60-0.98), although with longer follow-up, the difference was no longer significant [10]. A survival analysis presented after 383 deaths demonstrated no benefit of the dietary intervention in the study population overall, but exploratory subgroup analysis suggested that patients with hormone receptor-negative cancers continued to experience benefits from the intervention (HR for mortality 0.46, 95% CI 0.27-0.78).

In the Women's Healthy Eating and Living (WHEL) study, women with stages I to IIIa breast cancer were randomly assigned to low-fat, high-fruit and vegetable diet or to a control group [42]. Those assigned to the dietary intervention increased intake of fruits and vegetables and decreased percentage of dietary calories from fat, but there was no difference in the rate of recurrence between the groups at a median follow-up for 5.6 years (16.7 versus 16.9 percent).

The reason for the differences in outcomes of these studies is not clear. Many experts have attributed the improvements in DFS seen in the WINS trial to the weight loss experienced by participants; this hypothesis is currently being tested in a number of randomized trials. Given the discrepancies in findings of WINS and WHEL, dietary change has not been widely adopted as a part of the adjuvant treatment of breast cancer patients. For a discussion of the effect of weight loss interventions on outcomes in breast cancer survivors, please refer to the separate section within this topic. (See 'Effects of weight loss on cancer outcomes' below.)

Low-fat dietary patterns in postmenopausal women (who may be at risk for breast cancer) and healthy diet in the general population (not specifically in cancer survivors) are discussed separately. (See "Healthy diet in adults" and "Factors that modify breast cancer risk in women", section on 'Low-fat dietary pattern in postmenopausal women'.)

Weight — Obesity at diagnosis appears to be a risk factor for worse outcomes after a diagnosis of breast cancer. However, the impact of weight gain following diagnosis and the influence of interventions aimed at weight loss on outcomes require further study.

Weight at diagnosis — Obesity at the time of breast cancer diagnosis has been linked to worsened outcomes, both in clinical trials as well as in observational studies.

As examples of these data:

A meta-analysis presented by the Early Breast Cancer Trialists' Collaborative Group (EBCTCG) including 80,000 women in 70 different adjuvant trials demonstrated a 5 percent absolute increase in mortality in obese versus normal-weight premenopausal women with hormone receptor-positive disease [43].

In the Anastrozole, Tamoxifen, Alone or in Combination (ATAC) trial, women who had a body mass index (BMI) ≥35 kg/m2 had a 60 percent higher risk of recurrence as compared with women with a BMI <25 kg/m2 [44]. A similar result was noted among obese premenopausal women treated with ovarian suppression and anastrozole in the Austrian Breast Cancer Study Group 12 (ABCSG-12) study [45]. (See "Adjuvant endocrine and targeted therapy for postmenopausal women with hormone receptor-positive breast cancer", section on 'Obese women'.)

Obese women with hormone receptor-positive tumors treated with chemotherapy (doxorubicin, cyclophosphamide, and a taxane) in the Eastern Cooperative Oncology Group E1199 trial had a 20 percent higher risk of recurrence as compared with leaner women [46]. (See "Selection and administration of adjuvant chemotherapy for HER2-negative breast cancer", section on 'Obese women'.)

Observational studies also consistently link obesity at the time of breast cancer diagnosis to higher rates of breast cancer recurrence, cancer-related mortality, and overall mortality [20,47,48]. Several meta-analyses of observational studies demonstrated that women who were obese at diagnosis had a higher risk of breast cancer mortality and overall mortality compared with leaner women [47,48]. This increased risk of mortality was seen in both pre- and postmenopausal women [47] and across various breast cancer subtypes [48].

The etiology of the poor outcomes seen in obese women with breast cancer is not well understood. Some older observational studies did not take treatment factors into consideration, introducing potential bias into these reports. Historically, many oncologists used ideal body weight or capped the absolute doses of chemotherapy administered to obese patients due to fears of increased toxicity. Studies subsequently demonstrated that these practices resulted in inferior outcomes [49,50], leading the American Society of Clinical Oncology to develop guidelines recommending the use of full, weight-based doses of chemotherapy for obese individuals. This topic is discussed separately. (See "Dosing of anticancer agents in adults", section on 'Dosing for overweight/obese patients'.)

Weight after diagnosis — Weight gain after a breast cancer diagnosis may be associated with an increased risk of recurrence, although the available data do not consistently report this association [51]. As examples:

In the Nurses' Health Study (NHS), nonsmoking women previously treated for breast cancer who gained 0.5 to 2 kg/m2 and women who gained more than 2 kg/m2 had an increased risk of breast cancer death compared with women who maintained a stable weight (RR 1.35, 95% CI 0.93-1.95 and RR 1.64, 95% CI 1.07-2.51, respectively) [52].

In a combined analysis of 3215 women from the Life After Cancer Epidemiology (LACE) cohort and the WHEL study control group, weight gain after diagnosis was not associated with an increased risk of recurrence, even in individuals who gained >10 percent of body weight [53].

Effects of weight loss on cancer outcomes — Despite the abundant data linking obesity and poor prognosis in early breast cancer, there have been relatively few studies evaluating the efficacy and potential benefits of weight loss interventions in breast cancer survivors [54,55]. Several trials have demonstrated that weight loss programs can be successfully implemented in breast cancer survivors and that weight loss can be achieved through a number of different approaches [9,56-61]. However, more research is needed to determine whether purposeful weight loss after breast cancer diagnosis leads to improvements in the risks of breast cancer recurrence and related mortality.

Several large-scale trials have evaluated the feasibility and benefits of weight loss in women with breast cancer. Data are as follows:

The Lifestyle Intervention Study for Adjuvant Treatment of Early Breast Cancer (LISA) randomly assigned 338 postmenopausal women with hormone receptor-positive breast cancer to a two-year telephone-based weight loss intervention or to usual care [9]. LISA demonstrated that women randomized to the intervention lost more weight than controls at one and two years post-randomization as compared with controls (loss of 5.4 percent of baseline weight versus 0.7 percent at one year; and loss of 3.7 percent of baseline weight versus 0.4 percent at two years). Patients in the intervention group also reported significant improvements in physical functioning scores as compared with control participants [9]. Long-term follow-up demonstrated that women who had been randomized to the weight loss group had a nonsignificant reduction in breast cancer recurrence, but the study lacked power to evaluate this endpoint.

The Exercise and Nutrition to Enhance Recovery and Good Health for You (ENERGY) Trial randomized 692 women with a history of breast cancer to a group-based weight loss program or to a less intensive control group that received two meetings with a dietitian and nontargeted print materials [61]. Individuals randomized to the group-based intervention group lost more weight at one and two years post-randomization compared with controls (loss of 6.0 versus 1.5 percent at one year and 3.7 versus 1.3 percent at two years). Intervention participants also had lower blood pressure at one and two years post-randomization compared with controls.

Preliminary findings from the German SUCCESS C trial did not demonstrate benefit from a telephone-based weight loss program on disease outcomes in a subset of overweight and obese women with breast cancer taking part in an adjuvant chemotherapy trial [62]; however, interpretation of these findings is complicated by a high dropout rate (approximately 52 percent of intervention participants dropped out during the two-year intervention period, versus 19 percent of controls) and modest weight loss in the intervention arm. In this study, 2292 patients were randomized to either telephone-based, lifestyle-interventional (LI) counseling or general recommendations for healthy lifestyle. In the intention-to-treat analyses, there were no differences in DFS (HR 0.91, 95% CI 0.70-1.18) or overall survival (HR 0.90, 95% CI 0.63-1.28) between the study arms.

More work is needed to determine if weight loss of the magnitude seen in LISA and ENERGY will result in improvements in breast cancer-specific and overall mortality [61]. A number of ongoing large-scale efforts will provide more information about the impact of lifestyle intervention upon breast cancer outcomes in the coming years:

The Italian DIANA-5 trial tests the impact of a Mediterranean diet in combination with increased physical activity on breast cancer outcomes and serum biomarkers in women with early breast cancer [63].

The Breast cancer WEight Loss (BWEL) trial will test the impact of a telephone-based weight loss intervention on invasive DFS in 3136 overweight and obese women with stage II to III breast cancer in the United States and Canada [64].

Physical activity — Although there is no clear consensus on the "best" type of physical activity for breast cancer survivors, benefits have been seen with a number of types of exercise, and the safety of moderate-intensity exercise has been shown [65]. Data from observational studies show that women who participate in a moderate amount of physical activity after diagnosis have improved outcomes as compared with less active women [2,5,66-69]. However, randomized trials of interventions aimed at increasing physical activity have not been large enough to evaluate whether such interventions improve breast cancer outcomes among survivors.

A meta-analysis of 16 prospective observational studies in breast cancer survivors demonstrated a 48 percent reduction in overall mortality (95% CI 0.42-0.64) and a 28 percent reduction in breast cancer mortality (95% CI 0.60-0.85) in the most versus least active breast cancer survivors. There was a 24 percent (95% CI 11-36 percent) reduction in overall mortality for each 10 metabolic equivalent task-hour/week increase in postdiagnosis physical activity (equivalent to current recommendations of 150 minutes/week of at least moderate intensity activity). Breast cancer survivors who increased their activity after diagnosis relative to prediagnosis levels also had a lower risk of overall mortality (RR 0.61, 95% CI 0.42-0.80) compared with survivors who decreased levels of activity or were inactive at both time points [70].

Numerous randomized trials have tested the feasibility and potential benefits of increasing exercise after diagnosis in breast cancer patients [24,29,71-76]. Although data are limited for the impact of physical activity upon breast cancer outcomes, they suggest that exercise improves multiple clinical outcomes in breast cancer survivors including mental health (anxiety, depression), cancer-related fatigue, physical function and aerobic fitness, strength, health-related quality of life (QOL), body image, body size, and body composition. General exercise recommendations from the American College of Sports Medicine (ACSM) for cancer survivors, including breast cancer, are discussed above. (See 'Exercise and outcomes in cancer survivors' above.)

The impact of upper body strength training on symptoms, such as lymphedema, is discussed separately. (See "Clinical staging and conservative management of peripheral lymphedema", section on 'Diet and exercise'.)

Prostate cancer

Diet — There appears to be an association between dietary fat intake and outcomes for men with prostate cancer. However, the impact of dietary interventions aimed at reducing fat intake on cancer outcomes has not been tested in the setting of large-scale clinical trials. Therefore, dietary change has not been uniformly recommended as part of the treatment strategy for men with prostate cancer.

A number of reports have evaluated the relationship between dietary factors and prognosis in men with prostate cancer [65,77-79]. One review suggested that soy intake or consuming a low-fat, vegan diet may have a favorable impact on prostate-specific antigen (PSA) or PSA doubling time [80]. Other studies suggest that high intake of saturated fat may be associated with worsened outcomes (eg, recurrence or PSA failure), while a plant-based diet could be linked to a lower risk of progression [79,81-83]. In one of these studies, higher BMI and higher intake of saturated fat were both independent predictors of biochemical failure, whereas total caloric intake was not associated with prognosis [83]. Other notable findings were that men who were both obese and consumed a diet high in saturated fat had the shortest biochemical failure-free survival (19 months), while those who were not obese and who consumed a diet low in saturated fat had the longest biochemical failure-free survival (46 months).

Several studies have looked at dietary interventions in prostate cancer survivors [80]. These studies have been unable to draw definitive conclusions and have instead focused on decreasing dietary fat and increasing intake of vegetables, tomatoes/lycopene, and soy products.

The Prostate Cancer Lifestyle Trial (PCLT) enrolled 93 patients with early stage low-grade cancer undergoing watchful waiting and then randomly assigned them to treatment with usual care (controls) or to a multifactorial intervention arm that included counseling and group-based support sessions designed to lower fat to 10 percent of total calories, consume a plant-based diet, increase exercise, and practice stress management. At one year, the multifactorial intervention decreased the PSA by 4 percent, while the PSA rose in 6 percent in those in the control arm. By two years of follow-up, 27 versus 5 percent of patients in the control and experimental arm, respectively, had undergone conventional prostate cancer treatment (radical prostatectomy, radiotherapy, or androgen deprivation) [84,85]. These differences were statistically significant.

Based on data from the Men's Eating and Living (MEAL) study (CALGB 70807), telephone-based dietary counseling for increased vegetable consumption did not reduce the risk of disease progression in men with prostate cancer. This randomized clinical trial was conducted in 478 men (aged 50 to 80 years) with biopsy-proven prostate adenocarcinoma (International Society of Urological Pathology grade group 1 in those <70 years and ≤2 in those ≥70 years). Tumors were stage cT2a or less, and serum PSA levels were less than 10 ng/mL. Patients were randomly assigned to a counseling behavioral intervention by telephone promoting consumption of seven or more daily vegetable servings or a control group, which received written information about diet and prostate cancer. At two-year follow-up, time to progression was similar between the two intervention groups (HR 0.96, 95% CI 0.75-1.24). [86].

Weight — Obesity is associated with worse outcomes among men at diagnosis with clinically localized prostate cancer. However, whether changes in weight following a diagnosis of prostate cancer can modify prognosis is unknown and is an area of active investigation.

As illustrated by the following studies, compared with leaner men, obese men are at a greater risk for:

More aggressive disease – A study of 1866 men undergoing prostate biopsy showed that the risk of a high-grade prostate cancer (ie, Gleason score ≥7) increased with an increasing BMI [87]. However, there was no relationship between BMI and incidence of lower-grade prostate cancer. In addition, data from the SELECT trial suggest that the relationship between BMI and aggressive prostate cancer at diagnosis varies by ethnicity [88]. Other data suggest a link between BMI at diagnosis and several molecular factors such as the presence of TMPRSS-ERG fusion and chromatin remodeling [89,90].

More advanced disease – A meta-analysis demonstrated a linear relationship between BMI and incidence of advanced prostate cancer (RR 1.09, 95% CI 1.02-1.16 for each 5 kg/m2 increase) and an inverse relationship between BMI and the development of localized prostate cancer (RR 0.94, 95% CI 0.91-0.94 for each 5 kg/m2 increase in BMI) [77].

Less responsive disease – A report of 287 men treated with radical prostatectomy followed by androgen deprivation therapy demonstrated an increased risk for distant metastasis and a nonsignificant trend toward an increased risk of prostate-specific mortality in men with a higher BMI [91].

Higher mortality – A meta-analysis of 22 reports demonstrated a 20 percent increase in the risk of prostate-specific mortality (RR 1.20, 95% CI 0.99-1.46) and a 21 percent increase in the risk of biochemical progression (RR 1.21, 95% CI 1.11-1.31) for each 5 kg/m2 increase in BMI [21].

By contrast, higher BMI may be associated with better overall survival in men with metastatic, castration-resistant prostate cancer [92-94].

Physical activity — There are limited data on the relationship between physical activity patterns and prostate cancer prognosis, although it remains an area of active interest. Two studies illustrate the potential benefits of exercise in this population:

In a study of 1455 men with localized prostate cancer, men who walked for at least three hours/week at a brisk pace had a 57 percent lower rate of disease progression compared with men who walked at an easy pace for less than three hours/week (HR 0.43, 95% CI 0.21-0.91) [95].

In a second report looking at physical activity patterns and outcomes in 2705 men taking part in the Health Professionals Follow-Up Study, men who walked for at least 90 minutes per week at a moderate-to-brisk pace had a 46 percent lower risk of all-cause mortality as compared with men who performed less or slower-paced walking (HR 0.51, 95% CI 0.36-0.72) [96].

One ongoing study, the INTERVAL trial, will test the impact of high-intensity interval training on overall survival in men with castrate-resistant metastatic prostate cancer, providing some of the first evidence regarding the potential risks and benefits of physical activity interventions in individuals with advanced cancer.

Colorectal cancer

Diet — Data suggest that dietary factors may influence outcomes in some patients with colorectal cancer (CRC). However, there are scant data on whether changes in diet after a diagnosis of CRC would be beneficial to CRC survivors. Furthermore, many CRC survivors do not adhere to dietary recommendations. As an example, 84 percent of survivors were not following the American Cancer Society recommendations regarding fruit and vegetable intake [97].

General dietary pattern — Observational studies of patients with colon cancer suggest that dietary pattern may affect prognosis [98-101]. For example:

In one study, the dietary status of 1009 patients with stage III colon cancer enrolled in a clinical trial was assessed at enrollment, four months after surgery (midway through eight months of adjuvant chemotherapy), and 14 months after surgery [98]. Outcomes were analyzed according to the extent to which patients followed both a "prudent" diet (high in fruits, vegetables, fish, poultry, whole grains) and a "Western-style" diet (increased amounts of red and processed meats, sweets, desserts, refined grains).

Compared with patients in the lowest quintile of Western diet consumption, those in the highest quintile had a greater risk of recurrence (HR 2.85, 95% CI 1.75-4.63) and death (HR 2.32, 95% CI 1.36-3.96) on multivariate analyses. By contrast, there were no differences in these outcomes between the highest and lowest quintile of the prudent dietary pattern. In a separate analysis from this study, patients in the highest quintile of dietary glycemic load had a higher risk of recurrence compared with those in the lowest quintile (HR for DFS 1.79, 95% CI 1.29-2.48) [99]. A higher glycemic load was also associated with worse DFS among overweight or obese participants (BMI ≥25 kg/m2; HR 2.26, 95% CI 1.53-3.32), but not in those patients with a lower BMI. In addition, higher total carbohydrate intakes were associated with lower DFS and overall survival.

In a separate study of 529 newly diagnosed CRC patients identified through a familial CRC registry, CRC patients with a high processed meat dietary pattern had worsened DFS (the highest versus the lowest quartile: HR 1.82, 95% CI 1.07-3.09). No associations were observed with the prudent-vegetable or the high-sugar patterns and DFS [100].

Although these analyses controlled for known factors that influence prognosis in CRC, these data should be interpreted cautiously because dietary differences may have antedated the diagnosis of cancer, influencing the subsequent natural history of disease.

Healthy diet in the general population, not specifically in cancer survivors, is discussed separately. (See "Healthy diet in adults".)

Specific dietary factors — Several specific dietary factors have been reported with associations to outcomes in CRC survivors. None have been studied in randomized trials to date. The mechanism associating these factors with CRC biology is not known, these factors have been associated with insulin regulation as well as inflammation, which may influence growth and spread of CRC cells [102]. As these are initial reports and observational, it may be early to incorporate in clinical practice [99,103].

Coffee consumption – Although some evidence suggests that higher levels of coffee consumption may be associated with reduced cancer recurrence and death, causality is not established.

Stage III colon cancer – In one cohort of stage III (node-positive) colon cancer patients who participated in a trial comparing two different adjuvant chemotherapy regimens, patients consuming four or more cups per day of caffeinated coffee during and six months after adjuvant chemotherapy experienced a reduced cancer recurrence or mortality risk compared with abstainers (HR 0.48, 95% CI 0.25-0.91) [104]. The association persisted even after controlling for known or suspected predictors of patient outcomes, including physical activity, dietary glycemic load, and other dietary patterns. The authors postulated that coffee might reduce the risk of colon cancer recurrence and death through improved insulin sensitization and decreased hyperinsulinemia, on the basis of previous studies supporting the role of high-energy balance states in promoting colon cancer recurrence and mortality. However, it is possible that the observed association was related to confounding variables.

A beneficial impact of postdiagnosis coffee consumption was also reported in an analysis of data from the NHS and the Health Professionals Follow-up Study, which followed 1599 patients diagnosed with stage I to III CRC [105]. Higher total coffee intake was associated with reduced risk for both CRC-specific and all-cause mortality, with the strongest correlation among those with stage III CRC. Among these patients, every cup per day increase in total coffee intake was associated with an 18 percent lower risk of CRC-specific mortality (HR 0.82, 95% CI 0.68-1.0) and a 20 percent lower all-cause mortality (HR 0.80, 95% CI 0.68-0.91). By contrast, no association was found for patients with stage I or II CRC for either CRC-specific or all-cause mortality.

Metastatic colon cancer – Data also suggest an association between coffee (both caffeinated and decaffeinated) and survival in patients with advanced colorectal cancer. As an example, in a prospective observational cohort study of 1171 patients with treatment-naïve metastatic CRC who participated in a randomized trial of different chemotherapy and biological therapy combinations, increased consumption of coffee was associated with decreased risk of cancer progression (HR for 1 cup/day increment 0.95, 95% CI 0.91-1.00) and death (HR for 1 cup/day increment 0.93, 95% CI 0.89-0.98) [106]. Compared with participants who did not drink coffee, the study also demonstrated improved OS for those who consumed two to three (HR 0.82, 95% CI 0.67-1.00) and four or more cups of coffee per day (HR 0.64, 95% CI 0.46-0.87).

Fiber intake – A benefit for higher postdiagnosis fiber intake on CRC outcomes was suggested in a study of 1575 health care professionals with stage I to III CRC who were derived from two cohorts, the NHS and Health Professionals Follow-Up Study [107]. Patients who increased their fiber intake after diagnosis had a lower mortality, and each 5-g/day increase in intake was associated with a 19 percent lower CRC-specific mortality (95% CI 7 to 28 percent), and a 14 percent reduction in all-cause mortality (95% CI 8 to 19 percent). Notably, increased intake of cereal fiber was associated with both CRC-specific and all-cause mortality, while no association was found for fruit fiber.

Despite these data, there have been no randomized trials that have tested the impact of dietary interventions in individuals with early stage CRC.

Nuts – Diets rich in nuts may be associated with reduced rates of cancer recurrence and death in patients with stage III colon cancer. In a prospective, observational study of 826 patients with stage III colon cancer who reported dietary intake on food frequency questionnaires, those who consumed two or more servings of nuts per week experienced improved DFS (HR 0.58, 95% CI 0.37-0.92) and overall survival (HR 0.43, 95% CI 0.25-0.74) relative to those who consumed fewer than two servings of nuts per week [108]. In exploratory multivariate analyses of DFS adjusting for disease, treatment, and patient characteristics, nonsignificant trends towards improved DFS were observed with higher total nut intake, but statistical power to detect differences was limited by small sample size.

Glycemic load, carbohydrates, and sugar-sweetened beverages – Glycemic load, carbohydrates, and sugar-sweetened beverages are dietary measures associated with insulin resistance. Higher dietary glycemic load and total carbohydrate intake were associated with an increased risk of recurrence and mortality in an observational study of 1011 stage III colon cancer patients with stage III colon cancer treated with adjuvant chemotherapy trial [99]. Patients in the highest quintile of dietary glycemic load experienced worsened disease-free survival (DFS; HR 1.79, 95% CI 1.29-2.48) compared with those in the lowest quintile. Similarly, patients in the highest quintile of carbohydrate intake also experienced worsened DFS (HR 1.73, 95% CI 1.22-2.46) compared with those in the lowest quintile.

In another observational study of 1201 women in the NHS with nonmetastatic CRC, a higher combined postdiagnosis intake of sugar-sweetened beverages and fruit juices was associated with worsened overall mortality (HR for each additional serving 1.11, 95% CI 1.01-1.23) [109].

Weight — Increasing BMI, particularly obesity, has been associated with an increased risk of multiple obesity-associated cancers, including CRC [110]. However, BMI may have a paradoxical association with survival in patients with colon cancer, particularly those with BMI in the 25 to 32 kg/m2 range. Furthermore, whether purposeful weight loss provides a benefit in survival outcomes related to CRC is unknown. As such, until there is better understanding of what and how to counsel regarding BMI, as well as potential incorporation of body composition, we do not advise weight loss as a means to prevent recurrence.

In an analysis including 12,000 CRC survivors, those who were overweight had a greater risk of a obesity-related secondary cancers (HR 1.39, 95% CI 1.01-1.92), as did those who were obese (HR 1.47, 95% CI 1.02-2.12), relative to those with a normal BMI. These data suggest that obesity contributed to the elevated cancer risks rather than increased genetic susceptibility. Therefore, they support obesity as a risk factor for both primary and secondary obesity-related malignancies in this population.

In the Cancer Prevention Study-II Nutrition study, patients diagnosed with non-metastatic CRC who had a high prediagnosis BMI ≥30 kg/m2 had poorer overall, cancer-specific, and cardiovascular-specific survival relative to those with a normal BMI [111]. However, there was no association between postdiagnosis BMI and outcomes, which is consistent with the findings of at least one other study [112]. By contrast, another study demonstrated that stage II and III colon cancer patients with BMI >35 kg/m2 after surgery had a statistically significant 38 percent worse DFS compared with normal-weight patients (BMI 18.5 to 24.9 kg/m2) [113].

A population-based cohort of 3408 patients with stage I to III CRC suggested a U-shaped relationship between BMI and outcomes in CRC survivors [114]. Patients who were underweight at diagnosis (BMI <18.5 kg/m2) and those who were class II or III obese (BMI ≥35 kg/m2) had a worse all-cause mortality, compared with patients who were low-normal weight (BMI 18.5 to <23 kg/m2). By contrast, patients who were high-normal weight (BMI 23 to <25 kg/m2), low-overweight (BMI 25 to <28 kg/m2), and high-overweight (BMI 28 to <30 kg/m2) had lower mortality risks. Finally, patients who were class I obese (BMI 30 to <35 kg/m2) showed no difference in risk. These data raise the question of optimal weight in this population.

BMI does not reflect muscle or fat distribution, and there are increasing data supportive of a protective effect of muscle for cancer patients [115-117]. One explanation for the U-shaped curve and possible protective effect of being overweight (BMI 25 to <30 kg/m2) relates to body composition. As an example, in a pooled analysis of 44 observational studies that included 18,891 patients with CRC, over one-third of patients (37 percent) were found to be sarcopenic (ie, low muscle mass) around the time of diagnosis [118]. After CRC surgery, sarcopenia was associated with a higher risk of total postoperative complications (OR 1.84, 95% CI 1.35-2.49), such as postoperative severe complications; postoperative mortality; postoperative infections, and postoperative cardiopulmonary complications. Sarcopenic patients also had lower overall survival (HR 1.83, 95% CI 1.57-2.14) and cancer-specific survival (HR 1.77, 95% CI 1.40-2.23) compared with nonsarcopenic patients.

Physical activity — Higher levels of physical activity may reduce CRC-specific and overall mortality [119,120], in addition to having a beneficial impact on fatigue, QOL, and functional status [121-125].

A systematic review included six studies conducted in CRC survivors [119]. All examined leisure-time activity, and all but one adjusted for stage, CRC risk factors, and BMI. Three studies found that higher levels of physical activity after diagnosis were associated with a reduced risk of CRC-specific mortality, ranging from 43 to 61 percent. In addition, all five studies that examined physical activity after a diagnosis of CRC found that postdiagnosis activity was associated with reduced risks of death from any cause; four were statistically significant.

Despite these findings, some data suggest that CRC survivors do not adhere to physical activity guidelines. As an example, 65 percent of CRC survivors were not following recommendations for physical activity proposed by the American Cancer Society [97]. The ongoing Colon Health and Lifelong Exercise Change (CHALLENGE) study will evaluate the impact of exercise on disease outcomes for CRC survivors [126].

Endometrial cancer

Weight — Although obesity is associated with less aggressive histologic types of endometrial cancer [127], it is associated with an increased risk of death [13,127]. As in other cancer types, whether purposeful weight loss after endometrial cancer can improve outcomes is unknown and is a topic of continued investigation.

It is well documented that most women with type I endometrial cancer will not die of their disease; rather, they will die of intercurrent medical illnesses, many of which are also related to obesity [128]. Further, there is evidence that obesity may play a role in endometrial cancer prognosis. Specifically:

Obesity was associated with a higher risk for recurrent endometrial cancer and other malignancies when compared with women of normal weight [13].

Endometrial cancer survivors with obesity and diabetes have a decreased life expectancy when compared with their nonobese, nondiabetic counterparts with the same malignancy [129,130].

Compared with those with a BMI <25 kg/m2, the relative risk of death for obese endometrial cancer patients with BMI of 30 to 34 kg/m2 is 2.53 (95% CI 2.02-3.18) and for BMI >40 kg/m2, 6.25 (95% CI 3.75-10.42) [13].

Unfortunately, endometrial cancer survivors have a high prevalence of continued obesity after treatment has completed [131]. In addition, obese cancer survivors may be less likely to adopt weight loss or healthier lifestyle modifications [132], including the adoption of physical activity [133]. Underlining the complexity of weight change after cancer diagnosis and treatment, one study suggests that an increase or decrease in BMI after therapy may be adversely associated with mortality [134].

Beyond prognosis, studies in endometrial cancer survivors suggest that increasing obesity is associated with lower health-related QOL and is inversely correlated with lower functional domain scores, including lack of fulfillment in work and enjoyment of life [135]. A large population-based study from the Netherlands showed that increasing BMI is related to several QOL outcomes, including decreased vitality, fatigue, and more symptoms associated with lymphedema [136].

Efforts to increase physical activity and address obesity in these patients may be challenging for various reasons, including the presence of other medical conditions that might interfere with an increased active lifestyle, including arthritis, knee pain, and back pain [133]. Nonetheless, if the cancer diagnosis is indeed a "teachable moment," there is an opportunity to change behaviors [137-139].

Physical activity — Whether physical activity improves cancer-specific outcomes in women with endometrial cancer is unknown [140-143]. However, data from observational studies and a randomized trial have demonstrated that an exercise intervention is feasible in this population [144].

In the Survivors of Uterine Cancer Empowered by Exercise and Healthy Diet (SUCCEED) study, women with a BMI ≥25 kg/m2 were randomly assigned to a lifestyle intervention (consisting of a six-month education and counseling session) or to usual care [144]. Adherence to the intervention arm was 84 percent. In addition, women in the intervention arm reported greater endurance measured by physical activity minutes compared with women in the usual care group (mean difference of 89 minutes at 12 months). Whether changes in activity of this magnitude would have impact on survival outcomes requires further follow-up.

CLINICAL LIFESTYLE RECOMMENDATIONS FOR CANCER SURVIVORS — Given the growing body of evidence that exercise, diet, and weight management are important in cancer prevention and control, a number of groups, including the American Cancer Society, the American College of Sports Medicine [29], the Clinical Oncology Society of Australia [145], and the European Society for Medical Oncology [146], have developed nutrition and physical activity guidelines for cancer survivors based on the available evidence linking diet, weight, and physical activity to cancer outcomes [24,27-29,91]. Although recommendations have some individual differences, they broadly recommend the following:

Maintain a healthy weight and attempt weight loss if overweight or obese.

Engage in a physically active lifestyle, ideally consisting of at least 150 minutes of moderate-intensity (or 75 minutes of vigorous) physical activity per week plus two resistance training sessions.

Notably, the ACSM guideline also suggests that lower levels of physical activity (eg, 30 minutes of moderate-intensity aerobic physical activity three times per week or aerobic and resistance training two to three times per week) can also lead to improvements in patient-reported outcomes and fitness in cancer survivors. (See 'Exercise and outcomes in cancer survivors' above.)

Consume a healthy diet, with at least five servings of fruits and vegetables per day and limited ingestion of processed foods and red meats.

Limit alcohol to no more than one drink/day for women and two drinks/day for men. (See "Overview of the risks and benefits of alcohol consumption".)

SUMMARY AND RECOMMENDATIONS

Improving health-related outcomes in cancer survivors – Novel methods to improve outcomes in cancer survivors may offer attractive additions to the currently available treatment options, especially those that are also associated with improvements in quality of life (QOL) and other health benefits. (See 'Introduction' above.)

Energy balance factors – Diet, physical activity, and weight are collectively considered energy balance factors because they describe the relationship between energy consumed (diet), energy expended (physical activity), and energy stored (adiposity). They have each been linked to cancer outcomes. (See 'Energy balance in cancer' above.)

Dietary intake – Dietary intakes have been linked to outcomes in some cancers. However, there are few randomized trials testing the impact of dietary change on cancer outcomes. (See 'Breast cancer' above and 'Prostate cancer' above and 'Colorectal cancer' above and 'Endometrial cancer' above.)

Obesity and weight loss – Obesity (defined as a body mass index [BMI] >30 kg/m2) at diagnosis is also associated with an increased mortality risk in patients with early stage breast cancer and in endometrial cancer, and potentially in those with prostate cancer or colorectal cancer (for those with BMI >35 kg/m2). Whether purposeful weight loss after cancer diagnosis can improve outcomes is unknown and is a topic of continued investigation. (See 'Breast cancer' above and 'Prostate cancer' above and 'Colorectal cancer' above and 'Endometrial cancer' above.)

Physical activity – Observational studies have shown an association between physical activity patterns after cancer diagnosis and prognosis in individuals diagnosed with early-stage cancers of the breast, colon, and prostate. However, there are limited data from randomized trials evaluating the impact of increased physical activity on rates of cancer recurrence or mortality. (See 'Physical activity' above.)

Exercise and outcomes in cancer survivors – Multiple interventional studies have evaluated the safety, feasibility, and benefits of exercise in cancer survivors. The benefits include a reduction in fatigue, improvement in QOL, and improvements in fitness and physical functioning. (See 'Exercise and outcomes in cancer survivors' above.)

Lifestyle recommendations for diet, physical activity, and body weight in cancer survivors – Although limited trial data are available on the effects of interventions aimed at dietary modification, increasing physical activity, and weight loss among cancer survivors, we agree with guidelines for cancer survivors from the American Cancer Society, the American College of Sports Medicine, and other groups that recommend maintaining a healthy weight, adopting a physically active lifestyle, and eating a healthy diet rich in plant sources. (See 'Clinical lifestyle recommendations for cancer survivors' above.)

  1. Cancer fact sheets. Global Cancer Observatory (GCO). Available at: https://gco.iarc.fr/today/fact-sheets-cancers (Accessed on April 10, 2018).
  2. Holmes MD, Chen WY, Feskanich D, et al. Physical activity and survival after breast cancer diagnosis. JAMA 2005; 293:2479.
  3. Chlebowski RT, Aiello E, McTiernan A. Weight loss in breast cancer patient management. J Clin Oncol 2002; 20:1128.
  4. Goodwin P. Energy balance and cancer prognosis. In: Cancer Prevention and Management Through Exercise and Weight Control, McTiernan A (Ed), Taylor and Francis, Boca Raton 2006. p.405.
  5. Holick CN, Newcomb PA, Trentham-Dietz A, et al. Physical activity and survival after diagnosis of invasive breast cancer. Cancer Epidemiol Biomarkers Prev 2008; 17:379.
  6. Cespedes Feliciano EM, Kroenke CH, Meyerhardt JA, et al. Metabolic Dysfunction, Obesity, and Survival Among Patients With Early-Stage Colorectal Cancer. J Clin Oncol 2016.
  7. Fair AM, Montgomery K. Energy balance, physical activity, and cancer risk. Methods Mol Biol 2009; 472:57.
  8. Galvão DA, Newton RU. Review of exercise intervention studies in cancer patients. J Clin Oncol 2005; 23:899.
  9. Goodwin PJ, Segal RJ, Vallis M, et al. Randomized trial of a telephone-based weight loss intervention in postmenopausal women with breast cancer receiving letrozole: the LISA trial. J Clin Oncol 2014; 32:2231.
  10. Chlebowski RT, Blackburn GL, Thomson CA, et al. Dietary fat reduction and breast cancer outcome: interim efficacy results from the Women's Intervention Nutrition Study. J Natl Cancer Inst 2006; 98:1767.
  11. Willett WC. Nutritional Epidemiology, 2nd ed, Oxford University Press, New York 1998.
  12. Schwedhelm C, Boeing H, Hoffmann G, et al. Effect of diet on mortality and cancer recurrence among cancer survivors: a systematic review and meta-analysis of cohort studies. Nutr Rev 2016; 74:737.
  13. Calle EE, Rodriguez C, Walker-Thurmond K, Thun MJ. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med 2003; 348:1625.
  14. Larsson SC, Wolk A. Body mass index and risk of non-Hodgkin's and Hodgkin's lymphoma: a meta-analysis of prospective studies. Eur J Cancer 2011; 47:2422.
  15. Kant P, Hull MA. Excess body weight and obesity--the link with gastrointestinal and hepatobiliary cancer. Nat Rev Gastroenterol Hepatol 2011; 8:224.
  16. Anderson GL, Neuhouser ML. Obesity and the risk for premenopausal and postmenopausal breast cancer. Cancer Prev Res (Phila) 2012; 5:515.
  17. Aune D, Greenwood DC, Chan DS, et al. Body mass index, abdominal fatness and pancreatic cancer risk: a systematic review and non-linear dose-response meta-analysis of prospective studies. Ann Oncol 2012; 23:843.
  18. Lauby-Secretan B, Scoccianti C, Loomis D, et al. Body Fatness and Cancer--Viewpoint of the IARC Working Group. N Engl J Med 2016; 375:794.
  19. 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.
  20. Protani M, Coory M, Martin JH. Effect of obesity on survival of women with breast cancer: systematic review and meta-analysis. Breast Cancer Res Treat 2010; 123:627.
  21. Cao Y, Ma J. Body mass index, prostate cancer-specific mortality, and biochemical recurrence: a systematic review and meta-analysis. Cancer Prev Res (Phila) 2011; 4:486.
  22. Bray GA. The Battle of the Bulge: A History of Obesity Research, Dorrance, 2007.
  23. Ligibel JA, Alfano CM, Courneya KS, et al. American Society of Clinical Oncology position statement on obesity and cancer. J Clin Oncol 2014; 32:3568.
  24. Schmitz KH, Courneya KS, Matthews C, et al. American College of Sports Medicine roundtable on exercise guidelines for cancer survivors. Med Sci Sports Exerc 2010; 42:1409.
  25. Mishra SI, Scherer RW, Snyder C, et al. Exercise interventions on health-related quality of life for people with cancer during active treatment. Cochrane Database Syst Rev 2012; :CD008465.
  26. Li T, Wei S, Shi Y, et al. The dose-response effect of physical activity on cancer mortality: findings from 71 prospective cohort studies. Br J Sports Med 2016; 50:339.
  27. Patel AV, Friedenreich CM, Moore SC, et al. American College of Sports Medicine Roundtable Report on Physical Activity, Sedentary Behavior, and Cancer Prevention and Control. Med Sci Sports Exerc 2019; 51:2391.
  28. Schmitz KH, Campbell AM, Stuiver MM, et al. Exercise is medicine in oncology: Engaging clinicians to help patients move through cancer. CA Cancer J Clin 2019; 69:468.
  29. Campbell KL, Winters-Stone KM, Wiskemann J, et al. Exercise Guidelines for Cancer Survivors: Consensus Statement from International Multidisciplinary Roundtable. Med Sci Sports Exerc 2019; 51:2375.
  30. Friedenreich CM, Neilson HK, Farris MS, Courneya KS. Physical Activity and Cancer Outcomes: A Precision Medicine Approach. Clin Cancer Res 2016; 22:4766.
  31. Printz C. An expanded role for exercise in cancer treatment and survivorship: Backed by a trove of studies regarding the benefits of physical activity for patients with cancer and cancer survivors, researchers have updated exercise guidelines for these groups. Cancer 2020; 126:2731.
  32. Biswas A, Oh PI, Faulkner GE, et al. Sedentary time and its association with risk for disease incidence, mortality, and hospitalization in adults: a systematic review and meta-analysis. Ann Intern Med 2015; 162:123.
  33. Ness KK, Plana JC, Joshi VM, et al. Exercise Intolerance, Mortality, and Organ System Impairment in Adult Survivors of Childhood Cancer. J Clin Oncol 2020; 38:29.
  34. Rees-Punia E, Patel AV, Nocera JR, et al. Self-reported physical activity, sitting time, and mental and physical health among older cancer survivors compared with adults without a history of cancer. Cancer 2021; 127:115.
  35. Markes M, Brockow T, Resch KL. Exercise for women receiving adjuvant therapy for breast cancer. Cochrane Database Syst Rev 2006; :CD005001.
  36. Sweegers MG, Altenburg TM, Brug J, et al. Effects and moderators of exercise on muscle strength, muscle function and aerobic fitness in patients with cancer: a meta-analysis of individual patient data. Br J Sports Med 2019; 53:812.
  37. Tonorezos ES, Ford JS, Wang L, et al. Impact of exercise on psychological burden in adult survivors of childhood cancer: A report from the Childhood Cancer Survivor Study. Cancer 2019; 125:3059.
  38. Beebe-Dimmer JL, Ruterbusch JJ, Harper FWK, et al. Physical activity and quality of life in African American cancer survivors: The Detroit Research on Cancer Survivors study. Cancer 2020; 126:1987.
  39. Barlow-Krelina E, Chen Y, Yasui Y, et al. Consistent Physical Activity and Future Neurocognitive Problems in Adult Survivors of Childhood Cancers: A Report From the Childhood Cancer Survivor Study. J Clin Oncol 2020; 38:2041.
  40. Rock CL, Demark-Wahnefried W. Nutrition and survival after the diagnosis of breast cancer: a review of the evidence. J Clin Oncol 2002; 20:3302.
  41. Cancer Prevention Recommendations. World Cancer Research Fund. Available at: https://www.wcrf.org/dietandcancer/cancer-prevention-recommendations (Accessed on April 10, 2018).
  42. Pierce JP, Natarajan L, Caan BJ, et al. Influence of a diet very high in vegetables, fruit, and fiber and low in fat on prognosis following treatment for breast cancer: the Women's Healthy Eating and Living (WHEL) randomized trial. JAMA 2007; 298:289.
  43. Pan H. Effect of obesity in premenopausal ER+ early breast cancer: EBCTCG data on 80,000 patients in 70 trials. American Society of Clinical Oncology Annual Meeting, Chicago, IL, 2014.
  44. Sestak I, Distler W, Forbes JF, et al. Effect of body mass index on recurrences in tamoxifen and anastrozole treated women: an exploratory analysis from the ATAC trial. J Clin Oncol 2010; 28:3411.
  45. Pfeiler G, Königsberg R, Fesl C, et al. Impact of body mass index on the efficacy of endocrine therapy in premenopausal patients with breast cancer: an analysis of the prospective ABCSG-12 trial. J Clin Oncol 2011; 29:2653.
  46. Sparano JA, Wang M, Zhao F, et al. Obesity at diagnosis is associated with inferior outcomes in hormone receptor-positive operable breast cancer. Cancer 2012; 118:5937.
  47. Chan DS, Vieira AR, Aune D, et al. Body mass index and survival in women with breast cancer-systematic literature review and meta-analysis of 82 follow-up studies. Ann Oncol 2014; 25:1901.
  48. Lohmann AE, Soldera SV, Pimentel I, et al. Association of Obesity With Breast Cancer Outcome in Relation to Cancer Subtypes: A Meta-Analysis. J Natl Cancer Inst 2021; 113:1465.
  49. Griggs JJ, Sorbero ME, Lyman GH. Undertreatment of obese women receiving breast cancer chemotherapy. Arch Intern Med 2005; 165:1267.
  50. Griggs JJ, Bohlke K, Balaban EP, et al. Appropriate Systemic Therapy Dosing for Obese Adult Patients With Cancer: ASCO Guideline Update. J Clin Oncol 2021; 39:2037.
  51. Jung AY, Hüsing A, Behrens S, et al. Postdiagnosis weight change is associated with poorer survival in breast cancer survivors: A prospective population-based patient cohort study. Int J Cancer 2021; 148:18.
  52. Kroenke CH, Chen WY, Rosner B, Holmes MD. Weight, weight gain, and survival after breast cancer diagnosis. J Clin Oncol 2005; 23:1370.
  53. Caan BJ, Emond JA, Natarajan L, et al. Post-diagnosis weight gain and breast cancer recurrence in women with early stage breast cancer. Breast Cancer Res Treat 2006; 99:47.
  54. Shaikh H, Bradhurst P, Ma LX, et al. Body weight management in overweight and obese breast cancer survivors. Cochrane Database Syst Rev 2020; 12:CD012110.
  55. Martel S, Lambertini M, Agbor-Tarh D, et al. Body Mass Index and Weight Change in Patients With HER2-Positive Early Breast Cancer: Exploratory Analysis of the ALTTO BIG 2-06 Trial. J Natl Compr Canc Netw 2021; 19:181.
  56. Shaw C, Mortimer P, Judd PA. Randomized controlled trial comparing a low-fat diet with a weight-reduction diet in breast cancer-related lymphedema. Cancer 2007; 109:1949.
  57. Djuric Z, DiLaura NM, Jenkins I, et al. Combining weight-loss counseling with the weight watchers plan for obese breast cancer survivors. Obes Res 2002; 10:657.
  58. Goodwin P, Esplen MJ, Butler K, et al. Multidisciplinary weight management in locoregional breast cancer: results of a phase II study. Breast Cancer Res Treat 1998; 48:53.
  59. Mefferd K, Nichols JF, Pakiz B, Rock CL. A cognitive behavioral therapy intervention to promote weight loss improves body composition and blood lipid profiles among overweight breast cancer survivors. Breast Cancer Res Treat 2007; 104:145.
  60. Pakiz B, Flatt SW, Bardwell WA, et al. Effects of a weight loss intervention on body mass, fitness, and inflammatory biomarkers in overweight or obese breast cancer survivors. Int J Behav Med 2011; 18:333.
  61. Rock CL, Flatt SW, Byers TE, et al. Results of the Exercise and Nutrition to Enhance Recovery and Good Health for You (ENERGY) Trial: A Behavioral Weight Loss Intervention in Overweight or Obese Breast Cancer Survivors. J Clin Oncol 2015; 33:3169.
  62. Janni W, Rack BK, Friedl TW, et al. Abstract GS5-03: Lifestyle Intervention and Effect on Disease-free Survival in Early Breast Cancer Patients: Interim Analysis from the Randomized SUCCESS C Study. Cancer Res 2019; 79:4S.
  63. Villarini A, Pasanisi P, Traina A, et al. Lifestyle and breast cancer recurrences: the DIANA-5 trial. Tumori 2012; 98:1.
  64. Ligibel JA, Barry WT, Alfano C, et al. Randomized phase III trial evaluating the role of weight loss in adjuvant treatment of overweight and obese women with early breast cancer (Alliance A011401): study design. NPJ Breast Cancer 2017; 3:37.
  65. Hori S, Butler E, McLoughlin J. Prostate cancer and diet: food for thought? BJU Int 2011; 107:1348.
  66. Irwin ML, Smith AW, McTiernan A, et al. Influence of pre- and postdiagnosis physical activity on mortality in breast cancer survivors: the health, eating, activity, and lifestyle study. J Clin Oncol 2008; 26:3958.
  67. Pierce JP, Stefanick ML, Flatt SW, et al. Greater survival after breast cancer in physically active women with high vegetable-fruit intake regardless of obesity. J Clin Oncol 2007; 25:2345.
  68. Chen X, Lu W, Zheng W, et al. Exercise after diagnosis of breast cancer in association with survival. Cancer Prev Res (Phila) 2011; 4:1409.
  69. Chen LH, Irwin MR, Olmstead R, Haque R. Association of Physical Activity With Risk of Mortality Among Breast Cancer Survivors. JAMA Netw Open 2022; 5:e2242660.
  70. Schmid D, Leitzmann MF. Association between physical activity and mortality among breast cancer and colorectal cancer survivors: a systematic review and meta-analysis. Ann Oncol 2014; 25:1293.
  71. Bluethmann SM, Vernon SW, Gabriel KP, et al. Taking the next step: a systematic review and meta-analysis of physical activity and behavior change interventions in recent post-treatment breast cancer survivors. Breast Cancer Res Treat 2015; 149:331.
  72. Bourke L, Homer KE, Thaha MA, et al. Interventions for promoting habitual exercise in people living with and beyond cancer. Cochrane Database Syst Rev 2013; :CD010192.
  73. Zeng Y, Huang M, Cheng AS, et al. Meta-analysis of the effects of exercise intervention on quality of life in breast cancer survivors. Breast Cancer 2014; 21:262.
  74. Brown JC, Sarwer DB, Troxel AB, et al. A randomized trial of exercise and diet on health-related quality of life in survivors of breast cancer with overweight or obesity. Cancer 2021; 127:3856.
  75. McNeil J, Fahim M, Stone CR, et al. Adherence to a lower versus higher intensity physical activity intervention in the Breast Cancer & Physical Activity Level (BC-PAL) Trial. J Cancer Surviv 2022; 16:353.
  76. Fraser SF, Gardner JR, Dalla Via J, Daly RM. The Effect of Exercise Training on Lean Body Mass in Breast Cancer Patients: A Systematic Review and Meta-analysis. Med Sci Sports Exerc 2022; 54:211.
  77. Discacciati A, Orsini N, Wolk A. Body mass index and incidence of localized and advanced prostate cancer--a dose-response meta-analysis of prospective studies. Ann Oncol 2012; 23:1665.
  78. Davies NJ, Batehup L, Thomas R. The role of diet and physical activity in breast, colorectal, and prostate cancer survivorship: a review of the literature. Br J Cancer 2011; 105 Suppl 1:S52.
  79. Berkow SE, Barnard ND, Saxe GA, Ankerberg-Nobis T. Diet and survival after prostate cancer diagnosis. Nutr Rev 2007; 65:391.
  80. Van Patten CL, de Boer JG, Tomlinson Guns ES. Diet and dietary supplement intervention trials for the prevention of prostate cancer recurrence: a review of the randomized controlled trial evidence. J Urol 2008; 180:2314.
  81. Demark-Wahnefried W, Polascik TJ, George SL, et al. Flaxseed supplementation (not dietary fat restriction) reduces prostate cancer proliferation rates in men presurgery. Cancer Epidemiol Biomarkers Prev 2008; 17:3577.
  82. Meyer F, Bairati I, Shadmani R, et al. Dietary fat and prostate cancer survival. Cancer Causes Control 1999; 10:245.
  83. Strom SS, Yamamura Y, Flores-Sandoval FN, et al. Prostate cancer in Mexican-Americans: identification of risk factors. Prostate 2008; 68:563.
  84. Ornish D, Weidner G, Fair WR, et al. Intensive lifestyle changes may affect the progression of prostate cancer. J Urol 2005; 174:1065.
  85. Frattaroli J, Weidner G, Dnistrian AM, et al. Clinical events in prostate cancer lifestyle trial: results from two years of follow-up. Urology 2008; 72:1319.
  86. Parsons JK, Zahrieh D, Mohler JL, et al. Effect of a Behavioral Intervention to Increase Vegetable Consumption on Cancer Progression Among Men With Early-Stage Prostate Cancer: The MEAL Randomized Clinical Trial. JAMA 2020; 323:140.
  87. Fowke JH, Motley SS, Concepcion RS, et al. Obesity, body composition, and prostate cancer. BMC Cancer 2012; 12:23.
  88. Barrington WE, Schenk JM, Etzioni R, et al. Difference in Association of Obesity With Prostate Cancer Risk Between US African American and Non-Hispanic White Men in the Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA Oncol 2015; 1:342.
  89. Pettersson A, Lis RT, Meisner A, et al. Modification of the association between obesity and lethal prostate cancer by TMPRSS2:ERG. J Natl Cancer Inst 2013; 105:1881.
  90. Ebot EM, Gerke T, Labbé DP, et al. Gene expression profiling of prostate tissue identifies chromatin regulation as a potential link between obesity and lethal prostate cancer. Cancer 2017; 123:4130.
  91. Rock CL, Doyle C, Demark-Wahnefried W, et al. Nutrition and physical activity guidelines for cancer survivors. CA Cancer J Clin 2012; 62:243.
  92. Vidal AC, Howard LE, de Hoedt A, et al. Obese patients with castration-resistant prostate cancer may be at a lower risk of all-cause mortality: results from the Shared Equal Access Regional Cancer Hospital (SEARCH) database. BJU Int 2018; 122:76.
  93. Cushen SJ, Power DG, Murphy KP, et al. Impact of body composition parameters on clinical outcomes in patients with metastatic castrate-resistant prostate cancer treated with docetaxel. Clin Nutr ESPEN 2016; 13:e39.
  94. Mohammed AA, El-Tanni H, Ghanem HM, et al. Impact of body mass index on clinico-pathological parameters and outcome in patients with metastatic prostate cancer. J Egypt Natl Canc Inst 2015; 27:155.
  95. Richman EL, Kenfield SA, Stampfer MJ, et al. Physical activity after diagnosis and risk of prostate cancer progression: data from the cancer of the prostate strategic urologic research endeavor. Cancer Res 2011; 71:3889.
  96. Kenfield SA, Stampfer MJ, Giovannucci E, Chan JM. Physical activity and survival after prostate cancer diagnosis in the health professionals follow-up study. J Clin Oncol 2011; 29:726.
  97. Blanchard CM, Courneya KS, Stein K, American Cancer Society's SCS-II. Cancer survivors' adherence to lifestyle behavior recommendations and associations with health-related quality of life: results from the American Cancer Society's SCS-II. J Clin Oncol 2008; 26:2198.
  98. Meyerhardt JA, Niedzwiecki D, Hollis D, et al. Association of dietary patterns with cancer recurrence and survival in patients with stage III colon cancer. JAMA 2007; 298:754.
  99. Meyerhardt JA, Sato K, Niedzwiecki D, et al. Dietary glycemic load and cancer recurrence and survival in patients with stage III colon cancer: findings from CALGB 89803. J Natl Cancer Inst 2012; 104:1702.
  100. Zhu Y, Wu H, Wang PP, et al. Dietary patterns and colorectal cancer recurrence and survival: a cohort study. BMJ Open 2013; 3.
  101. Cheng E, Ou FS, Ma C, et al. Diet- and Lifestyle-Based Prediction Models to Estimate Cancer Recurrence and Death in Patients With Stage III Colon Cancer (CALGB 89803/Alliance). J Clin Oncol 2022; 40:740.
  102. Cheng E, Shi Q, Shields AF, et al. Association of Inflammatory Biomarkers With Survival Among Patients With Stage III Colon Cancer. JAMA Oncol 2023; 9:404.
  103. Fuchs MA, Sato K, Niedzwiecki D, et al. Sugar-sweetened beverage intake and cancer recurrence and survival in CALGB 89803 (Alliance). PLoS One 2014; 9:e99816.
  104. Guercio BJ, Sato K, Niedzwiecki D, et al. Coffee Intake, Recurrence, and Mortality in Stage III Colon Cancer: Results From CALGB 89803 (Alliance). J Clin Oncol 2015; 33:3598.
  105. Hu Y, Ding M, Yuan C, et al. Association Between Coffee Intake After Diagnosis of Colorectal Cancer and Reduced Mortality. Gastroenterology 2018; 154:916.
  106. Mackintosh C, Yuan C, Ou FS, et al. Association of Coffee Intake With Survival in Patients With Advanced or Metastatic Colorectal Cancer. JAMA Oncol 2020; 6:1713.
  107. Song M, Wu K, Meyerhardt JA, et al. Fiber Intake and Survival After Colorectal Cancer Diagnosis. JAMA Oncol 2018; 4:71.
  108. Fadelu T, Zhang S, Niedzwiecki D, et al. Nut Consumption and Survival in Patients With Stage III Colon Cancer: Results From CALGB 89803 (Alliance). J Clin Oncol 2018; 36:1112.
  109. Fung TT, Kashambwa R, Sato K, et al. Post diagnosis diet quality and colorectal cancer survival in women. PLoS One 2014; 9:e115377.
  110. Gibson TM, Park Y, Robien K, et al. Body mass index and risk of second obesity-associated cancers after colorectal cancer: a pooled analysis of prospective cohort studies. J Clin Oncol 2014; 32:4004.
  111. Campbell PT, Newton CC, Dehal AN, et al. Impact of body mass index on survival after colorectal cancer diagnosis: the Cancer Prevention Study-II Nutrition Cohort. J Clin Oncol 2012; 30:42.
  112. Meyerhardt JA, Niedzwiecki D, Hollis D, et al. Impact of body mass index and weight change after treatment on cancer recurrence and survival in patients with stage III colon cancer: findings from Cancer and Leukemia Group B 89803. J Clin Oncol 2008; 26:4109.
  113. Dignam JJ, Polite BN, Yothers G, et al. Body mass index and outcomes in patients who receive adjuvant chemotherapy for colon cancer. J Natl Cancer Inst 2006; 98:1647.
  114. Kroenke CH, Neugebauer R, Meyerhardt J, et al. Analysis of Body Mass Index and Mortality in Patients With Colorectal Cancer Using Causal Diagrams. JAMA Oncol 2016; 2:1137.
  115. Caan BJ, Meyerhardt JA, Kroenke CH, et al. Explaining the Obesity Paradox: The Association between Body Composition and Colorectal Cancer Survival (C-SCANS Study). Cancer Epidemiol Biomarkers Prev 2017; 26:1008.
  116. Hopkins JJ, Reif RL, Bigam DL, et al. The Impact of Muscle and Adipose Tissue on Long-term Survival in Patients With Stage I to III Colorectal Cancer. Dis Colon Rectum 2019; 62:549.
  117. Chung E, Lee HS, Cho ES, et al. Changes in Body Composition During Adjuvant FOLFOX Chemotherapy and Overall Survival in Non-Metastatic Colon Cancer. Cancers (Basel) 2019; 12.
  118. Trejo-Avila M, Bozada-Gutiérrez K, Valenzuela-Salazar C, et al. Sarcopenia predicts worse postoperative outcomes and decreased survival rates in patients with colorectal cancer: a systematic review and meta-analysis. Int J Colorectal Dis 2021; 36:1077.
  119. Ballard-Barbash R, Friedenreich CM, Courneya KS, et al. Physical activity, biomarkers, and disease outcomes in cancer survivors: a systematic review. J Natl Cancer Inst 2012; 104:815.
  120. Brown JC, Ma C, Shi Q, et al. Physical Activity in Stage III Colon Cancer: CALGB/SWOG 80702 (Alliance). J Clin Oncol 2023; 41:243.
  121. Lynch BM, Cerin E, Owen N, et al. Prospective relationships of physical activity with quality of life among colorectal cancer survivors. J Clin Oncol 2008; 26:4480.
  122. Peddle CJ, Au HJ, Courneya KS. Associations between exercise, quality of life, and fatigue in colorectal cancer survivors. Dis Colon Rectum 2008; 51:1242.
  123. Morey MC, Snyder DC, Sloane R, et al. Effects of home-based diet and exercise on functional outcomes among older, overweight long-term cancer survivors: RENEW: a randomized controlled trial. JAMA 2009; 301:1883.
  124. Buffart LM, Thong MS, Schep G, et al. Self-reported physical activity: its correlates and relationship with health-related quality of life in a large cohort of colorectal cancer survivors. PLoS One 2012; 7:e36164.
  125. Arem H, Pfeiffer RM, Engels EA, et al. Pre- and postdiagnosis physical activity, television viewing, and mortality among patients with colorectal cancer in the National Institutes of Health-AARP Diet and Health Study. J Clin Oncol 2015; 33:180.
  126. Courneya KS, Booth CM, Gill S, et al. The Colon Health and Life-Long Exercise Change trial: a randomized trial of the National Cancer Institute of Canada Clinical Trials Group. Curr Oncol 2008; 15:279.
  127. Everett E, Tamimi H, Greer B, et al. The effect of body mass index on clinical/pathologic features, surgical morbidity, and outcome in patients with endometrial cancer. Gynecol Oncol 2003; 90:150.
  128. von Gruenigen VE, Tian C, Frasure H, et al. Treatment effects, disease recurrence, and survival in obese women with early endometrial carcinoma : a Gynecologic Oncology Group study. Cancer 2006; 107:2786.
  129. Kaaks R, Lukanova A, Kurzer MS. Obesity, endogenous hormones, and endometrial cancer risk: a synthetic review. Cancer Epidemiol Biomarkers Prev 2002; 11:1531.
  130. Abu-Abid S, Szold A, Klausner J. Obesity and cancer. J Med 2002; 33:73.
  131. Courneya KS, Karvinen KH, Campbell KL, et al. Associations among exercise, body weight, and quality of life in a population-based sample of endometrial cancer survivors. Gynecol Oncol 2005; 97:422.
  132. von Gruenigen VE, Waggoner SE, Frasure HE, et al. Lifestyle challenges in endometrial cancer survivorship. Obstet Gynecol 2011; 117:93.
  133. Basen-Engquist K, Scruggs S, Jhingran A, et al. Physical activity and obesity in endometrial cancer survivors: associations with pain, fatigue, and physical functioning. Am J Obstet Gynecol 2009; 200:288.e1.
  134. Matsuo K, Moeini A, Cahoon SS, et al. Weight Change Pattern and Survival Outcome of Women with Endometrial Cancer. Ann Surg Oncol 2016; 23:2988.
  135. Fader AN, Frasure HE, Gil KM, et al. Quality of life in endometrial cancer survivors: what does obesity have to do with it? Obstet Gynecol Int 2011; 2011:308609.
  136. Oldenburg CS, Boll D, Nicolaije KA, et al. The relationship of body mass index with quality of life among endometrial cancer survivors: a study from the population-based PROFILES registry. Gynecol Oncol 2013; 129:216.
  137. Demark-Wahnefried W, Peterson B, McBride C, et al. Current health behaviors and readiness to pursue life-style changes among men and women diagnosed with early stage prostate and breast carcinomas. Cancer 2000; 88:674.
  138. Demark-Wahnefried W, Aziz NM, Rowland JH, Pinto BM. Riding the crest of the teachable moment: promoting long-term health after the diagnosis of cancer. J Clin Oncol 2005; 23:5814.
  139. Jernigan AM, Maurer KA, Cooper K, et al. Referring survivors of endometrial cancer and complex atypical hyperplasia to bariatric specialists: a prospective cohort study. Am J Obstet Gynecol 2015; 213:350.e1.
  140. Arem H, Chlebowski R, Stefanick ML, et al. Body mass index, physical activity, and survival after endometrial cancer diagnosis: results from the Women's Health Initiative. Gynecol Oncol 2013; 128:181.
  141. Arem H, Park Y, Pelser C, et al. Prediagnosis body mass index, physical activity, and mortality in endometrial cancer patients. J Natl Cancer Inst 2013; 105:342.
  142. Arem H, Pfeiffer RM, Moore SC, et al. Body mass index, physical activity, and television time in relation to mortality risk among endometrial cancer survivors in the NIH-AARP Diet and Health Study cohort. Cancer Causes Control 2016; 27:1403.
  143. Tarasenko YN, Linder DF, Miller EA. Muscle-strengthening and aerobic activities and mortality among 3+ year cancer survivors in the U.S. Cancer Causes Control 2018; 29:475.
  144. von Gruenigen V, Frasure H, Kavanagh MB, et al. Survivors of uterine cancer empowered by exercise and healthy diet (SUCCEED): a randomized controlled trial. Gynecol Oncol 2012; 125:699.
  145. Cormie P, Atkinson M, Bucci L, et al. Clinical Oncology Society of Australia position statement on exercise in cancer care. Med J Aust 2018; 209:184.
  146. Physical activity guidelines. In: Rehabilitation Issues During Cancer Treatment and Follow-Up, European Society for Medical Oncology (ESMO). Available at: https://oncologypro.esmo.org/education-library/esmo-handbooks/rehabilitation-issues-during-cancer-treatment-and-follow-up/Physical-Activity-Guidelines (Accessed on January 14, 2021).
Topic 14222 Version 43.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟