Skip to main content
Erschienen in: Current Colorectal Cancer Reports 5/2017

Open Access 14.09.2017 | Nutrition and Nutritional Interventions in Colorectal Cancer (K Wu, Section Editor)

Lifestyle after Colorectal Cancer Diagnosis in Relation to Survival and Recurrence: A Review of the Literature

verfasst von: Moniek van Zutphen, Ellen Kampman, Edward L. Giovannucci, Fränzel J. B. van Duijnhoven

Erschienen in: Current Colorectal Cancer Reports | Ausgabe 5/2017

Abstract

Purpose of Review

This review summarizes the evidence regarding diet, physical activity, smoking, and body composition after colorectal cancer (CRC) diagnosis in relation to all-cause and CRC-specific mortality and disease recurrence and gives suggestions for future research directions.

Recent Findings

Overall, this review suggests that some, albeit not all, of the well-known modifiable risk factors for cancer incidence might also be associated with CRC survival. CRC prognosis appears to be worse with increased physical inactivity, smoking, or being underweight after CRC diagnosis. Emerging evidence suggests that diets associated with a positive energy balance, e.g., high consumption of sugar-sweetened beverages, may negatively impact survival in CRC survivors. In contrast, there is currently little evidence to support the recommendation to limit red and processed meat or alcohol intake after CRC diagnosis. Whether being overweight and obese after CRC diagnosis improves or worsens CRC prognosis remains controversial and may depend on the measure used to assess body fatness.

Summary

Further research on post-diagnosis lifestyle patterns is needed to understand the multifactorial influence on CRC prognosis. Disease recurrence and the development of comorbidities should be included as key outcomes in future studies and lifestyle should preferably be repeatedly measured.
Hinweise
This article is part of the Topical Collection on Nutrition and Nutritional Interventions in Colorectal Cancer

Introduction

Diet, physical activity, smoking, alcohol, and body weight are associated with risk (incidence) of colorectal cancer (CRC) [1, 2]. In contrast, far fewer studies have examined the influence of these lifestyle factors on survival after CRC diagnosis. Currently, cancer survivors are advised to follow the recommendations formulated for cancer prevention [3]. However, it is currently unclear if making lifestyle changes after diagnosis would impact disease progression and survival.
Emerging evidence shows that lifestyle, including diet, after CRC diagnosis might affect all-cause and CRC-specific mortality risk. Several recent reviews and meta-analyses on observational studies summarized the available evidence on specific aspects of lifestyle, such as diet [4••, 5, 6], physical activity [4••, 5, 710, 11••, 12], smoking [13••, 14], and body composition [5, 10, 15, 16, 17••, 1822], in relation to CRC outcomes. However, none of these reviews included all the aforementioned lifestyle factors in one review. Furthermore, results might differ due to the timing of lifestyle assessment (e.g., pre-diagnosis vs. post-diagnosis) [8, 10, 15] and characteristics of the included study population [15].
To better understand the association between lifestyle and CRC outcomes, we summarized the evidence regarding diet, physical activity, smoking, and body composition after CRC diagnosis across different groups of cancer survivors. Moreover, we also included observational studies, not included in previous reviews [2338, 39••]. We identified three study design categories based on the selection of the included study population: (1) population-based studies including all incident CRC cases, (2) studies in the adjuvant setting limited to survivors treated with adjuvant therapy, and (3) studies in the metastatic setting limited to patients with metastatic disease (Fig. 1). We chose to focus on post-diagnosis lifestyle factors, because this is the period during which CRC survivors could be counseled to alter their behavior. Therefore, we only included studies that examined the association between lifestyle at or after CRC diagnosis and all-cause mortality, CRC-specific mortality, or cancer recurrence. Additionally, we summarized the evidence regarding changes in lifestyle, i.e., from pre- to post-diagnosis or changes made after diagnosis, among CRC survivors and survival outcomes from either observational or intervention studies. We did not include papers that examined lifestyle and CRC survival separately by molecular subtypes. These publications will be reviewed in future issue of this journal. Finally, we conclude with suggestions for future research directions.

Overview of Included Studies

We excluded all studies that did not assess lifestyle at or after CRC diagnosis (e.g., those that assessed only pre-diagnosis factors) or did not adjust for critical confounders (e.g., age, stage). Furthermore, we excluded all studies that dichotomized body mass index (BMI) when examining the association between BMI and mortality or recurrence. Dichotomized BMI is considered a crude classification of BMI by combining diverse categories of body mass and body composition. Thus, dichotomized BMI may not account for potential differential associations between sub-categories of BMI (e.g., by combining overweight and obese in one category) [15].
We included 57 relevant articles (based on 84 different observational studies) that reported on post-diagnosis diet, physical activity, smoking, or body fatness/body composition in CRC survivors in relation to all-cause mortality, CRC-specific mortality, or cancer recurrence. An overview of the number of included articles according to exposure and type of study population is shown in Fig. 2. Additionally, we included 13 relevant articles (one intervention study and 11 different observational studies) that reported on changes in lifestyle among CRC survivors in relation to survival outcomes. In total, 61 articles are discussed in more detail in this review.

Diet after CRC Diagnosis

Five population-based studies and one study in the adjuvant setting provided results on diet and CRC outcomes in 10 publications [2327, 4044] (Table 1). Three US cohorts assessed post-diagnosis diet in population-based cohorts with > 1000 CRC patients: Nurses’ Health Study I (NHS) [23, 44], Health Professional Follow-Up Study (HPFS) [44], and Cancer Prevention Study (CPS) II Nutrition Cohort [27, 40, 41]. All three cohorts consist of participants diagnosed with CRC during follow-up and have updated dietary assessment after diagnosis. Usually, questionnaires that were completed after treatment was finished were utilized in the analyses. In contrast, two non-US cohorts (the German cohort PopGen [24] and BioBank Japan [26]) recruited > 1000 CRC patients after CRC diagnosis. The study in the adjuvant setting, Cancer and Leukemia Group B (CALGB) 89,803 Diet and Lifestyle Companion study [25, 42, 43], was embedded in a randomized trial of adjuvant chemotherapy among ~ 1000 patients with stage III colon cancer. Additionally, three articles, two from the CPS II Nutrition Cohort [27, 40] and one report on a small randomized dietary intervention trial reported on dietary changes among CRC survivors in relation to mortality [27, 40, 78].
Table 1
Cohort studies among individuals with colon or rectal cancer examining lifestyle factors after diagnosis in relation to all-cause mortality, colorectal cancer-specific mortality, or recurrence
First author, year, name of cohort, country
Study population
Time of post-diagnosis exposure assessment
Outcomes assessed
Year of CRC diagnosis and follow-up
Lifestyle factor
All-cause mortality HR (95% CI)
Colorectal cancer-specific mortality HR (95% CI)
Covariates
Dietary patterns—population-based studies
 Fung, 2014,
Nurses’ Health Study I, USA [23]
n = 1201
W only
CRC
Stage I–III
≥ 6 months after CRC diagnosis (mean 21.0 months)
All-cause mortality (n = 435);
CRC-specific mortality (n = 162)
Diagnosis 1986–2008;
Median FU 11.2 years
Western dietary pattern
Q1
Q2
Q3
Q4
Q5
P trend
Prudent dietary pattern
Q1
Q2
Q3
Q4
Q5
P trend
Alternate Healthy Eating Index (AHEI)
Q1
Q2
Q3
Q4
Q5
P trend
Alternate Mediterranean Diet (aMED) score
Q1
Q2
Q3
Q4
Q5
P trend
Dietary Approaches to Stop Hypertension (DASH) score
Q1
Q2
Q3
Q4
Q5
P trend
1.0
1.15 (0.83–1.58)
1.02 (0.72–1.43)
1.37 (0.97–1.94)
1.32 (0.89–1.97)
0.23
1.0
0.84 (0.62–1.13)
0.91 (0.67–1.25)
1.02 (0.73–1.42)
0.93 (0.65–1.34)
0.80
1.0
0.84 (0.63–1.10)
0.71 (0.53–0.94)
0.71 (0.52–0.96)
0.71 (0.52–0.98)
0.01
1.0
1.14 (0.85–1.52)
1.01 (0.75–1.37)
0.92 (0.66–1.27)
0.87 (0.63–1.21)
0.31
1.0
0.92 (0.68–1.24)
0.96 (0.69–1.32)
0.87 (0.65–1.18)
0.98 (0.71–1.35)
0.66
1.0
1.48 (0.87–2.54)
1.00 (0.55–1.83)
1.50 (0.84–2.70)
1.66 (0.85–3.23)
0.09
1.0
0.67 (0.40–1.12)
0.62 (0.37–1.05)
0.91 (0.53–1.55)
0.67 (0.37–1.22)
0.16
1.0
0.69 (0.42–1.12)
0.73 (0.45–1.17)
0.76 (0.47–1.23)
0.72 (0.43–1.21)
0.07
1.0
1.18 (0.73–1.91)
0.96 (0.58–1.56)
0.73 (0.42–1.20)
0.84 (0.50–1.42)
0.19
1.0
0.84 (0.52–1.34)
0.70 (0.41–1.22)
0.72 (0.43–1.20)
0.87 (0.52–1.45)
0.35
Age, PA, BMI, weight change, tumor grade, chemotherapy, smoking, energy intake, tumor site, stage, date of CRC diagnosis
 Ratjen, 2017, PopGen, Germany [24]
n = 1404
M and W
CRC
Stage I–IV
6 years after diagnosis (median)
All-cause mortality (n = 204)
Diagnosis 1993–2005;
Median FU 7 years
Modified Mediterranean Diet Score
Q1
Q2
Q3
Q4
P trend
Per 1-point increment
Healthy Nordic Food Index
Q1
Q2
Q3
Q4
P trend
Per 1-point increment
1.0
0.92 (0.64–1.34)
0.85 (0.59–1.23)
0.48 (0.32–0.74)
0.001
0.88 (0.81–0.96)
1.0
0.87 (0.59–1.27)
0.77 (0.49–1.22)
0.63 (0.39–1.04)
0.06
0.90 (0.82–0.99)
 
Sex, age, BMI, PA, survival time from CRC diagnosis until diet assessment, tumor site, metastases, other cancer, chemotherapy, smoking, total energy intake, and time-varying age, BMI, and metastases
Dietary patterns—studies in the adjuvant setting
 Meyerhardt, 2007, CALGB 89803, USA [42]
n = 1009
M and W
Colon
Stage III
Midway through adjuvant therapy and 6 months after completion of adjuvant therapy
All-cause mortality (n = 251);
Recurrence-free survival (n = 324);
Disease-free survival (n = 352)
Diagnosis 1999–2001
Median FU 5.3 years
Western dietary pattern
Q1
Q2
Q3
Q4
Q5
P trend
Prudent dietary pattern
Q1
Q2
Q3
Q4
Q5
P trend
1.0
0.74 (0.48–1.17)
1.38 (0.90–2.11)
1.66 (1.04–2.65)
2.32 (1.36–3.96)
< 0.001
1.0
1.18 (0.81–1.71)
0.94 (0.62–1.43)
0.72 (0.46–1.13)
1.32 (0.86–2.04)
0.54
1.0
0.92 (0.63–1.36)
1.42 (0.98–2.07)
1.44 (0.94–2.19)
2.85 (1.75–4.63)
< 0.001
1.0
1.07 (0.76–1.51)
1.05 (0.74–1.51)
0.83 (0.57–1.23)
1.13 (0.77–1.67)
0.84
Sex, age, depth of invasion through bowel wall, number of positive lymph nodes, presence of clinical perforation at time of surgery, presence of bowel obstruction at time of surgery, baseline performance status, treatment group, weight change between first and second questionnaire, and time-varying body mass index, PA level, and total calories
Red and processed meats—population-based studies
 McCullough, 2013, CPS II Nutrition Cohort, USA [40]
n = 1186
M and W
CRC
Stage I–III
3 years after diagnosis (mean)
All-cause mortality (n = 472);
CRC-mortality (n = 146);
CVD-mortality (n = 110);
other-mortality (n = 216)
Diagnosis 1992–2009
Mean FU 7.6 years (SD 3.4 years)
Red and processed meat intake
Q1
Q2
Q3
Q4
P trend
1.0
1.17 (0.89–1.55)
1.13 (0.84–1.52)
0.94 (0.68–1.30)
0.36
1.0
1.28 (0.76–2.15)
0.93 (0.53–1.64)
1.10 (0.61–1.91)
0.91
Age, sex, stage, energy intake, weight change between 1992 pre-diagnostic and post-diagnostic questionnaires, and 1992 pre-diagnostic meat intake
 Fung, 2014,
Nurses’ Health Study I, USA [23]
n = 1201
W only
CRC
Stage I–III
≥ 6 months after diagnosis (mean 21.0 months)
All-cause mortality (n = 435);
CRC-specific mortality (n = 162)
Diagnosis 1986–2008;
Median FU 11.2 years
Red/processed meat per serving/day (secondary analyses)
1.07 (0.87–1.30)
1.22 (0.90–1.67)
Age, PA, BMI, weight change, tumor grade, chemotherapy, smoking, energy intake, tumor site, stage, date of CRC diagnosis
Sugar-sweetened beverages—population-based studies
 Fung, 2014, Nurses’ Health Study I, USA [23]
n = 1201
W only
CRC
Stage I–III
≥ 6 months after diagnosis (mean 21.0 months)
All-cause mortality (n = 435)
CRC-specific mortality (n = 162)
Diagnosis 1986–2008;
Median FU 11.2 years
Sugar-sweetened beverages + juices per serving/d (secondary analyses)
1.11 (1.01–1.23)
1.16 (0.99–1.35)
Age, PA, BMI, weight change, tumor grade, chemotherapy, smoking, energy intake, tumor site, stage, date of CRC diagnosis
Sugar-sweetened beverages—studies in the adjuvant setting
 Fuchs, 2014, CALGB 89803, USA [43]
n = 1011
M and W
Colon
Stage III
Midway through adjuvant therapy and 6 months after completion of adjuvant therapy
All-cause mortality (n = 305)
Recurrence-free survival (n = 343);
Disease-free survival (n = 386)
Diagnosis 1999–2001
Median FU 7.3 years
Sugar-sweetened beverages intake
< 2/month
2/month to 2/week
3 to 6/week
1 to < 2/day
≥ 2/day
P trend
1.0
0.74 (0.53–1.04)
1.07 (0.75–1.53)
0.70 (0.43–1.15)
1.41 (0.79–2.50)
0.21
1.0
0.98 (0.72–1.34)
1.34 (0.97–1.87)
1.07 (0.70–1.65)
1.75 (1.04–2.94)
0.04
Age, sex, depth of invasion through bowel wall, number of positive lymph nodes, baseline performance status, treatment group, and the following time-varying covariates total energy intake, BMI, PA level, Western dietary pattern, prudent dietary pattern, and glycemic load
Alcohol—population-based studies
 Fung, 2014, Nurses’ Health Study, USA [23]
n = 1201
W only
Colon and rectum
Stage I–III
≥ 6 months after diagnosis (mean 21.0 months)
All-cause mortality (n = 435);
CRC-specific mortality (n = 162)
Diagnosi: 1986–2008;
Median FU 11.2 years
No alcohol intake
5–15 g/day
> 15 g/day
(secondary analyses)
1.30 (1.05–1.61)
1.0
1.22 (0.85–1.76)
1.32 (0.93–1.87)
1.0
0.97 (0.50–1.87)
Age, PA, BMI, weight change, tumor grade, chemotherapy, smoking, energy intake, tumor site, stage, date of CRC diagnosis
 Lochhead, 2015, Nurses’ Health Study I + Health Professional Follow-Up Study, USA [44]
n = 1550
M and W
CRC
Stage I–III
≥ 1 year but ≤ 4 years after CRC diagnosis (median 29.5 months)
All-cause mortality (n = 641);
CRC-specific mortality (n = 176)
Diagnosis up to 2006;
Median FU 14.9 years
Alcohol intake g/day
0
0.1–14.9
≥ 15
P trend
1.0
0.83 (0.70–0.99)
0.91 (0.72–1.16)
0.41
1.0
0.51 (0.34–0.76)
0.53 (0.28–0.98)
0.33
Pre-diagnostic alcohol consumption, age, year of diagnosis, BMI, family history of CRC, aspirin use, multivitamin use, smoking, PA, folate, vitamin B12, methionine, and vitamin B6 intake, tumor site, tumor differentiation, time from diagnosis to questionnaire return, and stage- and sex-stratified
 Yang, 2017, CPS II Nutrition Cohort, USA [27]
n = 1599
M and W
CRC
Stage I–III
1.9 years after CRC diagnosis (mean)
All-cause mortality (n = 732);
CRC-specific mortality (n = 235);
CVD-mortality (n = 172);
other mortality (n = 325)
Diagnosis: 1992–2011;
Mean FU: 8.2 years (SD 4.7 years)
Alcohol drinking
Never
Former-former
Current-former
Current < 2 drinks/day
Current ≥ 2 drinks/day
1.0
1.09 (0.81–1.48)
1.21 (0.92–1.60)
0.94 (0.77–1.16)
0.92 (0.66–1.26)
1.0
1.28 (0.73–2.23)
1.81 (1.13–2.91)
1.27 (0.87–1.86)
1.44 (0.80–2.60)
Age, six, tumor stage, smoking status, BMI, PA, education, and pre-existing diseases in 1982/1992
 Tamakoshi, 2017, BioBank Japan, Japan [26]
n = 1598
M and W
CRC
Stage I–IV
Within 90 days after CRC diagnosis
All-cause mortality (n = 521)
Diagnosis 2003–2008;
Median FU 7.4 years
Never drinker
Ex drinker
0–15 g/day
15–30 g/day
≥30 g/day
1.0
1.26 (0.98–1.63)
0.73 (0.56–0.97)
0.79 (0.57–1.11)
0.73 (0.56–0.96)
 
Stratified by sex and institutions and adjusted for age and entry year
Other food groups and nutrients—population-based studies
 Yang, 2014, CPS II Nutrition cohort, USA [41]
n = 1111
M and W
CRC
Stage I–III
2.6 years after CRC diagnosis (mean)
All-cause mortality (n = 429);
CRC-specific mortality (n = 143)
Diagnosis 1992–2009;
Mean FU 7.6 years (SD 3.4)
Total dairy intake
Q1
Q2
Q3
Q4
P trend
Milk intake
Q1
Q2
Q3
Q4
P trend
Total calcium intake
Q1
Q2
Q3
Q4
P trend
Dietary calcium intake
Q1
Q2
Q3
Q4
P trend
Supplemental calcium intake
C1
C2
C3
P trend
Total vitamin D
Q1
Q2
Q3
Q4
P trend
Dietary vitamin D intake
Q1
Q2
Q3
Q4
P trend
1.0
0.91 (0.69–1.21)
0.73 (0.54–0.98)
0.75 (0.56–1.01)
0.05
1.0
0.85 (0.64–1.13)
0.76 (0.52–1.12)
0.72 (0.55–0.94)
0.02
1.0
0.89 (0.67–1.18)
0.72 (0.53–0.98)
0.72 (0.53–0.98)
0.02
1.0
0.84 (0.63–1.11)
0.69 (0.51–0.93)
0.86 (0.65–1.14)
0.21
1.0
0.95 (0.72–1.27)
0.98 (0.73–1.31)
0.55
1.0
0.81 (0.59–1.10)
0.97 (0.67–1.40)
0.88 (0.57–1.35)
0.35
1.0
0.99 (0.75–1.31)
0.95 (0.71–1.27)
0.90 (0.67–1.21)
0.33
1.0
0.73 (0.44–1.23)
0.92 (0.56–1.52)
0.73 (0.44–1.23)
0.32
1.0
0.90 (0.54–1.49)
0.85 (0.44–1.67)
0.93 (0.59–1.49)
0.81
1.0
1.15 (0.71–1.86)
0.81 (0.48–1.38)
0.59 (0.33–1.05)
0.01
1.0
0.85 (0.51–1.41)
0.98 (0.59–1.62)
1.00 (0.61–1.63)
0.83
1.0
1.04 (0.65–1.69)
0.65 (0.38–1.11)
0.13
1.0
0.99 (0.59–1.66)
1.31 (0.66–2.58)
1.74 (0.80–3.77)
0.52
1.0
0.78 (0.46–1.32)
1.11 (0.67–1.85)
1.28 (0.77–2.10)
0.19
Age, sex, stage, energy intake, post-diagnostic energy intake, and total folate intakes
 Fung, 2014, Nurses’ Health Study, USA [23]
n = 1201
W only CRC Stage I–III
≥ 6 months after diagnosis (mean 21.0 months)
Overall mortality (n = 435);
CRC-specific mortality (n = 162)
Diagnosis: 1986–2008;
Median FU: 11.2 years
Per serving/day
Whole fruits
Vegetables
Nuts
Whole grains
(all secondary analyses)
1.08 (0.98–1.20)
(0.94–1.06)
0.98 (0.82–1.17)
0.98 (0.95–1.01)
1.03 (0.87–1.21)
0.94 (0.84–1.04)
0.69 (0.49–0.97)
0.97 (0.93–1.02)
Age, PA, BMI, weight change, tumor grade, chemotherapy, smoking, energy intake, tumor site, stage, date of CRC diagnosis
 Lochhead, 2015, Nurses’ Health Study I + Health Professional Follow-Up Study, USA [44]
n = 1550
M and W
CRC Stage I–III
≥ 1 year but ≤ 4 year after CRC diagnosis (median 29.5 months)
All-cause mortality (n = 641);
CRC-specific mortality (n = 176)
Diagnosis up to 2006;
Median FU 14.9 years
Folate intake
Q1
Q2
Q3
Q4
Q5
P trend
Vitamin B6 intake
Q1
Q2
Q3
Q4
Q5
P trend
Vitamin B12 intake
Q1
Q2
Q3
Q4
Q5
P trend
Methionine
Q1
Q2
Q3
Q4
Q5
P trend
1.0
1.03 (0.81–1.31)
1.17 (0.92–1.49)
0.86 (0.66–1.13)
0.87 (0.65–1.16)
0.13
1.0
0.87 (0.69–1.11)
0.80 (0.62–1.03)
0.94 (0.73–1.22)
0.78 (0.59–1.03)
0.18
1.0
1.19 (0.93–1.52)
0.96 (0.74–1.23)
0.94 (0.72–1.22)
1.11 (0.82–1.50)
0.71
1.0
0.82 (0.63–1.05)
0.92 (0.71–1.19)
1.02 (0.79–1.31)
1.17 (0.92–1.49)
0.053
1.0
1.17 (0.74–1.88)
1.63 (1.04–2.56)
0.76 (0.43–1.35)
1.04 (0.60–1.82)
0.21
1.0
0.95 (0.59–1.51)
1.08 (0.67–1.74)
0.94 (0.57–1.55)
0.93 (0.58–1.49)
0.66
1.0
1.23 (0.77–1.95)
0.70 (0.43–1.14)
0.88 (0.55–1.42)
1.04 (0.62–1.74)
0.99
1.0
0.57 (0.34–0.95)
0.82 (0.51–1.32)
0.79 (0.50–1.27)
0.90 (0.57–1.41)
0.91
Alcohol consumption, age, year of diagnosis, BMI, family history of CRC, aspirin use, multivitamin use, smoking, PA, folate, vitamin B12, methionine, and vitamin B6 intake, tumor site, tumor differentiation, time from diagnosis to questionnaire return, and stage- and sex-stratified
 Tamakoshi, 2017, BioBank Japan, Japan [26]
n = 1598
M and W
CRC
Stage I–IV
Within 90 days after CRC diagnosis
All-cause mortality (n = 521)
Diagnosis 2003–2008
Median FU 7.4 years
Green leafy vegetable consumption
Almost everyday
3–4 days/week
1–2 days/week
Almost never
Meat consumption
Almost everyday
3–4 days/week
1–2 days/week
Almost never
1.0
1.27 (0.99–1.62)
1.61 (1.18–2.20)
1.87 (1.22–2.88)
1.0
1.04 (0.76–1.41)
1.06 (0.78–1.43)
1.21 (0.85–1.71)
 
Stratified by sex and institutions and adjusted for age and entry year
Other food groups and nutrients—studies in the adjuvant setting
 Meyerhardt, 2012, CALGB 89803, USA [45]
n = 1011
M and W
Colon
Stage III
Midway through adjuvant therapy and 6 months after completion of adjuvant therapy
All-cause mortality (n = 305);
Recurrence-free survival (n = 343);
Disease-free survival (n = 386)
Diagnosis 1999–2001;
Median FU 7.3 years
Glycemic load
Q1
Q2
Q3
Q4
Q5
P trend
Glycemic index
Q1
Q2
Q3
Q4
Q5
P trend
Fructose
Q1
Q2
Q3
Q4
Q5
P trend
Carbohydrate intake
Q1
Q2
Q3
Q4
Q5
P trend
1.0
0.83 (0.55–1.23)
1.05 (0.72–1.54)
1.50 (1.04–2.17)
1.74 (1.20–2.51)
< 0.001
1.0
0.94 (0.64–1.37)
1.22 (0.84–1.77)
1.09 (0.74–1.61)
1.23 (0.83–1.82)
0.22
1.0
0.82 (0.57–1.18)
0.74 (0.51–1.08)
0.92 (0.64–1.32)
1.11 (0.79–1.58)
0.40
1.0
1.00 (0.68–1.49)
1.11 (0.76–1.63)
1.60 (1.11–2.32)
1.80 (1.25–2.60)
< 0.001
1.0
1.01 (0.70–1.47)
1.07 (0.74–1.56)
1.70 (1.18–2.40)
1.97 (1.39–2.79)
< 0.001
1.0
0.99 (0.69–1.43)
1.21 (0.85–1.73)
1.21 (0.84–1.73)
1.24 (0.85–1.81)
0.14
1.0
0.82 (0.58–1.17)
0.95 (0.67–1.33)
1.04 (0.74–1.47)
1.43 (1.04–1.98)
0.01
1.0
1.07 (0.73–1.56)
1.20 (0.83–1.73)
1.76 (1.24–2.50)
2.06 (1.45–2.91)
< 0.001
Sex, age, depth of invasion through bowel wall, number of positive lymph nodes, baseline performance status, treatment group, time-varying BMI, time-varying PA, time-varying cereal fiber, and time-varying dietary pattern
 Guercio, 2015, CALGB 89803, USA [25]
n = 953
M and W
Colon
Stage III
Midway through adjuvant therapy and 6 months after completion of adjuvant therapy
All-cause mortality (n = 324);
Recurrence-free survival (n = 329);
Disease-free survival (n = 365)
Diagnosis 1999–2001
Median FU 7.3 years
Total coffee cups/day
0
< 1
1
2–3
≥ 4
P trend
Non-herbal tea cups/day
0
< 1
1
2–3
≥ 4
P trend
1.0
0.97 (0.66–1.44)
0.97 (0.66–1.42)
0.69 (0.47–1.01)
0.66 (0.37–1.18)
0.01
1.0
1.08 (0.81–1.44)
0.87 (0.58–1.30)
0.95 (0.60–1.50)
0.82 (0.40–1.67)
0.36
1.0
0.98 (0.68–1.43)
0.97 (0.66–1.42)
0.80 (0.56–1.14)
0.71 (0.41–1.23)
0.07
1.0
1.09 (0.83–1.43)
0.89 (0.61–1.30)
1.03 (0.67–1.57)
0.86 (0.44–1.68)
0.51
Age, sex, depth of invasion through bowel wall, number of positive lymph nodes, baseline performance status, treatment group, smoking history, multivitamin, and the following time-varying covariates total energy intake, alcohol consumption, BMI, PA level, Wester dietary pattern, prudent dietary pattern, sugar-sweetened beverage intake and dietary glycemic load
Physical activity—population-based studies
 Meyerhardt, 2006, Nurses’ Health Study I, USA [46]
n = 554
W only
CRC
Stage I–III
≥ 1 year but ≤ 4 years after CRC diagnosis (median 22 months)
All-cause mortality (n = 121);
CRC-specific mortality (n = 72)
Diagnosis 1986–2002;
Median FU 9.6 years
Total MET-h activity/week
< 3
3–8.9
9–17.9
≥ 18
P trend
1.0
0.77 (0.48–1.23)
0.50 (0.28–0.90)
0.43 (0.25–0.74)
0.003
1.0
0.92 (0.50–1.69)
0.57 (0.27–1.20)
0.39 (0.18–0.82)
0.008
Age, year of diagnosis, BMI, stage, tumor grade, tumor site, chemotherapy, time from diagnosis to PA measurement, change in BMI before and after diagnosis, and smoking
 Meyerhardt, 2009, Health Professional Follow-Up Study, USA [47]
n = 661
M only
CRC
Stage I–III
≥ 6 months but ≤ 4 years after CRC diagnosis (median 15 months)
All-cause mortality (n = 258);
CRC-specific mortality (n = 88)
Diagnosis 1986–2004;
Median FU 8.6 years
Total MET-h activity/week
≤ 3
3.1–9
9.1–18
18.1–27
≥ 27
P trend
1.0
1.00 (0.68–1.48)
1.12 (0.74–1.70)
0.74 (0.46–1.20)
0.59 (0.41–0.86)
< 0.001
1.0
1.06 (0.55–2.08)
1.30 (0.65–2.59)
0.76 (0.33–1.77)
0.47 (0.24–0.92)
0.002
Age, stage, tumor grade, tumor site, diagnosis year, BMI at diagnosis, time from diagnosis to PA measurement, change in BMI before and after diagnosis, and smoking
 Baade, 2011, Queensland, Australia [48]
n = 1825
M and W
CRC
Stage I–III
5 months after CRC diagnosis
All-cause mortality (n = 462);
CRC-specific mortality (n = 345)
Diagnosis 2003–2004;
Median FU 4.9 years (range 4.0–6.0)
PA min/wk.
0
1–149
≥ 150
P trend
1.0
0.72 (0.57–0.91)
0.75 (0.60–0.94)
0.007
1.0
0.90 (0.69–1.17)
0.88 (0.68–1.15)
0.585
NR
 Kuiper, 2012, Women’s Health Initiative, USA [49]
n = 606
W only
CRC
Stage I–III
1.5 year after CRC diagnosis (median)
All-cause mortality (n = 108);
CRC-specific mortality (n = 51)
Diagnosis ≥ 1993;
Median FU 11.9 years (IQR 10.9–12.9)
Total MET-h activity /week
0
> 0–2.9
3.0–8.9
9.0–17.9
≥ 18
P trend
1.0
0.71 (0.40–1.30)
0.42 (0.23–0.77)
0.57 (0.31–1.07)
0.41 (0.21–0.81)
0.005
1.0
0.49 (0.21–1.14)
0.30 (0.12–0.73)
0.53 (0.22–1.25)
0.29 (0.11–0.77)
0.02
Age, study arm, stage, ethnicity, education, alcohol, smoking, and hormone therapy use, pre-diagnostic BMI, time between baseline measurement and diagnosis
 Campbell, 2013, CPS II Nutrition Cohort, USA [50]
n = 1800
M and W
CRC
Stage I–III
1.4 years after CRC diagnosis (median)
All-cause mortality (n = 588);
CRC-specific mortality (n = 226);
CVD-mortality (n = 127);
Mortality from other causes (n = 235)
Diagnosis 1994–2007;
Mean FU 6.8 years
Total MET-h activity/week
< 3.5
3.5–8.74
≥ 8.75
1.0
0.78 (0.60–1.00)
0.58 (0.47–0.71)
1.0
1.00 (0.64–1.56)
0. 87 (0.61–1.24)
Age, sex, smoking, BMI, red meat intake, stage, leisure time spent sitting, and education
 Arem, 2015, National Institutes of Health-AARP, USA [51]
n = 1759
M and W
CRC
Stage I–III
4.2 years after CRC diagnosis (median)
All-cause mortality (n = 412);
CRC-specific mortality (n = 128);
CVD-specific mortality (n = 82)
Diagnosis 1996–2006;
Median FU 7.1 years
PA h/wk.
0
< 1
1–3.9
4–6.9
≥ 7
P trend
1.0
1.00 (0.72–1.39)
0.88 (0.65–1.19)
0.66 (0.46–0.94)
0.69 (0.49–0.98)
0.006
1.0
0.98 (0.53–1.81)
0.96 (0.57–1.62)
0.69 (0.36–1.29)
0.53 (0.27–1.03)
0.041
Sex, tumor site, tumor grade, stage, surgery, radiation, chemotherapy, time watching TV, smoking, BMI, self-reported health status, pre- and post-diagnosis PA (age is time metric in model)
 Tamakoshi, 2017, BioBank Japan, Japan [26]
n = 1598
M and W
CRC
Stage I–IV
Within 90 days after diagnosis
All-cause mortality (n = 521)
Diagnosis 2003–2008
Median FU 7.4 years
Physical exercise
≥ 3 times/week
1–2 times/week
No habit
1.0
0.60 (0.33–1.08)
1.33 (1.05–1.68)
 
Stratified by sex and institutions and adjusted for age and entry year
Physical activity—studies in the adjuvant setting
 Meyerhardt, 2006, CALGB 89803, USA [52]
n = 832
M and W
Colon
Stage III
7.1 months after completion of adjuvant treatment (median)
All-cause mortality (n = 84);
Recurrence-free survival (n = 159);
Disease-free survival (n = 172)
Inclusion 1999–2001;
Median FU 2.7 years
Total MET-h activity/week
< 3
3–8.9
9–17.9
18–26.9
≥ 27
P trend
1.0
0.85 (0.49–1.49)
0.71 (0.36–1.41)
0.71 (0.32–1.59)
0.37 (0.16–0.82)
0.01
1.0
0.86 (0.57–1.30)
0.89 (0.55–1.42)
0.51 (0.26–1.01)
0.60 (0.36–1.01)
0.03
Age, sex, depth of invasion through bowel wall, no. of positive lymph nodes, clinical perforation at time of surgery, baseline CEA, tumor, baseline performance status, treatment arm, weight change between first and second questionnaire, BMI at time of second questionnaire, time between study entry, and completion of second questionnaire
Sedentary behavior—population-based studies
 Campbell, 2013, CPS II Nutrition Cohort, USA [50]
n = 1656
M and W
CRC
Stage I–III
1.9 years after CRC diagnosis (median)
All-cause mortality (n = 477);
CRC-specific mortality (n = 169);
CVD-mortality (n = 110);
Mortality from other causes (n = 198)
Diagnosis 1994–2007;
Mean FU 6.8 years
Leisure time spent sitting
< 3 h/day
3–< 6
≥ 6 h/day
1.0
1.13 (0.91–1.40)
1.27 (0.99–1.64)
1.0
1.23 (0.84–1.78)
1.62 (1.07–2.44)
Age, sex, smoking, BMI, red meat intake, stage, PA, and education
 Arem, 2015, National Institutes of Health-AARP, USA [51]
n = 1759
M and W
CRC
Stage I–III
4.2 years after diagnosis (median)
All-cause mortality (n = 412);
CRC-specific mortality (n = 128);
CVD-specific mortality (n = 82)
Diagnosis 1996–2006;
Median FU 7.1 years
TV viewing
0–2 h/day
3–4 h/day
≥ 5 h/day
P trend
1.0
0.98 (0.75–1.27)
1.25 (0.93–1.67)
0.126
1.0
0.90 (0.56–1.46)
1.45 (0.85–2.47)
0.156
Age as time metric. Sex, tumor site, tumor grade, stage, chemotherapy, PA, smoking, BMI, self-reported health, and pre-diagnosis TV viewing
 Cao, 2015,
Health Professional Follow-Up Study, USA [53]
n = 714
M only
CRC
Stage I–III
≥ 6 months but ≤ 3 years after CRC diagnosis
All-cause mortality (n = 325);
CRC-specific mortality (n = 72);
Mortality from other causes (n = 253)
Diagnosis 1986–2010;
FU until end 2011
Sitting watching TV
0–6 h/week
7–13 h/week
14–20 h/week
≥ 21 h/week
P trend
1.0
0.98 (0.70–1.37)
1.01 (0.72–1.42)
1.16 (0.80–1.68)
0.66
1.0
0.62 (0.27–1.41)
0.68 (0.30–1.54)
1.45 (0.73–2.87)
0.27
Age, year of diagnosis, stage, tumor grade, tumor site, smoking, PA, BMI, AHEI, and pre-diagnosis TV viewing
Smoking—population based studies
 Jadallah, 1999, Dunedin hospital, New Zealand [54]
n = 241
M and W
CRC
Stage I–III
Hospital record
All-cause mortality (n = 81)
Diagnosis 1990–1992;
FU 5 years
Non-smoker
Smoking
1.0
2.26 (1.31–3.90)
 
Blood transfusion, stage
 Ali, 2011, Irish National Cancer Registry, Ireland [55]
n = 22,335
M and W
CRC
Stage I–IV
Cancer registry
All-cause mortality (n = 11,400);
Diagnosis 1994–2005;
Max FU 15 years
Former smoker
Current smoker
Never smoker
Current smoker
1.0
1.15 (1.07–1.23)
1.0
1.20 (1.13–1.28)
 
Age, tumor grade, stage
 Warren, 2013, Roswell Park Cancer Institute, USA [56]
n = 359
M and W
CRC
Stage I–IV
Within 1 month after CRC diagnosis
All-cause mortality (n = NR);
CRC-specific mortality (n = NR):
Diagnosis 1982–1998
FU 12–27.7 years
Men
Former smoker
Current smoker
Never smoker
Current smoker
Women
Former smoker
Current smoker
Never smoker
Current smoker
1.0
1.07 (0.64–1.81)
1.0
1.05 (0.62–1.78)
1.0
0.89 (0.39–2.06)
1.0
1.70 (0.87–3.31)
1.0
1.14 (0.56–2.27)
1.0
0.70 (0.36–1.36)
1.0
1.18 (0.34–4.05)
1.0
1.85 (0.85–4.02)
Disease site, sex, age, stage, race, date of diagnosis, BMI, total pack-years of smoking
 Tao, 2013, Shanghai Cohort Study, China [28]
n = 248
M only
CRC
Stage NR
At diagnosis and yearly thereafter
All-cause mortality (n = 152)
Diagnosis 1986–2010;
Mean FU 5.3 (±4.8) years
Non-smoking
Smoking (time-dependent)
1.0
1.65 (1.14–2.38)
 
Age, education, pack-years of smoking before diagnosis, treatment, and cancer site
 Amri, 2015, Massachusetts General Hospital, USA [57]
n = 1071
M and W
CRC
Stage I–IV
At pre-operative assessment
All-cause mortality (n = NR);
CRC-specific mortality (n = NR);
Metastatic recurrence (n = NR)
Diagnosis 2004–2011;
FU NR
Non-smoking
Current smoking
P trend
1.0
1.44 (1.07–1.94)
0.017
1.0
1.21 (0.80–1.83)
0.36
Age, stage, BMI, comorbidities
 Walter, 2015, DACHS study, Germany [58]
n = 3130
M and W
CRC
Stage I–IV
24 days after CRC diagnosis (median)
All-cause mortality (n = 889);
CRC-specific mortality (n = 644);
Recurrence-free survival (n = 828);
Disease-free survival (n = 1024);
Non-CRC related mortality (n = 232)
Diagnosis 2003–2010;
Median FU 4.9 years (IQR 2.9–5.1)
Non-smoking
< 15 cigarettes/day
≥ 15 cigarettes/day
1.0
1.10 (0.85–1.43)
0.99 (0.73–1.32)
1.0
1.08 (0.83–1.41)
1.14 (0.87–1.51)
Age, sex, BMI, stage, alcohol consumption, red meat consumption, family history of CRC, use of statins, use of NSAIDs, use of beta blockers, diabetes mellitus, history of heart failure, myocardial infarction, angina pectoris or stroke, history of nonCRC cancer; additional adjustment for age × log(time) and cancer × log(time)
 Yang, 2015, CPS II Nutrition Cohort, USA [29]
n = 2256
M and W
CRC
Stage I–III
1.4 years after CRC diagnosis (mean)
All-cause mortality (n = 865);
CRC-specific mortality (n = 324)
Diagnosis 1992–2009;
Mean FU 7.5 years (SD 4.6 years)
Never smoking
Former smoking
Current smoking
1.0
1.21 (1.03–1.42)
2.22 (1.58–3.13)
1.0
0.91 (0.71–1.18)
1.92 (1.15–3.21)
Age, sex, stage, alcohol consumption, BMI, and PA
 Sharp, 2017, National Cancer Registry Ireland, Ireland [14]
n = 18,166
M and W
Colon
Stage I–IV
At diagnosis
CRC-specific mortality (n = 7488)
Diagnosis 1994–2012;
FU 5 years
Never smoker
Ex-smoker
Current smoker
P trend
 
1.0
1.00 (0.94–1.07)
1.14 (1.07–1.22)
< 0.01
Sex, marital status, deprivation category, period of diagnosis, grade, tumor site. With stage and age fitted as stratification factors
 Sharp, 2017, National Cancer Registry Ireland, Ireland [30]
n = 10,794
M and W
Rectum
Stage I–IV
At diagnosis
CRC-specific mortality (n = 4491)
Diagnosis 1994–2012;
FU 5 years
Never smoker
Ex-smoker
Current smoker
P trend
 
1.0
1.02 (0.93–1.11)
1.15 (1.06–1.24)
< 0.01
Sex, marital status, deprivation category, period of diagnosis, grade. With stage and age fitted as stratification factors
 Rasouli, 2017, Kurdistan’s Cancer Registry, Iran [31]
n = 335
M and W
CRC
Stage II–III
Medical record
All-cause mortality (n = 164)
Diagnosis 2009–2014;
Median FU 42.6 ± 2.8 months
Non-smoking
Smoking
1.0
1.34 (0.92–1.95)
 
Age, residence, marital status, occupation, education, socioeconomic status, comorbidity, stage, tumor grade
 Tamakoshi, 2017, BioBank Japan, Japan [26]
n = 1598
M and W
CRC
Stage I–IV
Within 90 days after CRC diagnosis
All-cause mortality (n = 521)
Diagnosis 2003–2008;
Median FU 7.4 years
Never smoker
Ex-smoker
Current smoker
1.0
1.27 (1.02–1.59)
1.38 (1.06–1.81)
 
Stratified by sex and institutions and adjusted for age and entry year
Smoking—studies in the adjuvant setting
 Munro, 2006, Tayside Cancer Centre, UK [59]
n = 284
M and W
CRC
Stage NR
At the first assessment in the oncology department, usually around 4 weeks after surgery
CRC-specific mortality (n = 83)
Diagnosis: 1997–1999;
Median FU: 56 months (range 20–83)
Non-smoker
Current smoker
 
1.0
2.24 (1.25–4.01)
Number of positive nodes, deprivation, co-morbidity, T stage
 McCleary, 2010, CALGB 89803, USA [60]
n = 1045
M and W
Colon
Stage III
4 months after surgery
All-cause mortality (n = 257);
Recurrence-free survival (n = 332);
Disease-free survival (n = 363);
Diagnosis 1999–2001;
Median FU: 5.3 years
Never smoker
Former smoker
Current smoker
1.0
1.17 (0.87–1.57)
1.38 (0.87–2.18)
1.0
1.15 (0.89–1.48)
0.90 (0.58–1.41)
Age, sex, number of positive lymph nodes, extent of invasion through bowel wall, tumor differentiation, BMI, and clinical bowel obstruction at diagnosis
 Phipps, 2013, North Central Cancer Treatment Group N0147, USA [61]
n = 1968
M and W
Colon
Stage III
Within 56 days after surgery
Time-to-recurrence (n = NR);
Disease-free survival (n = NR)
Diagnosis 2004–2009;
Median FU 3.5 years
Never smoker
Former smoker
Current smoker
 
1.0
1.19 (0.97–1.46)
1.47 (1.03–2.11)
Tumor site, number of involved lymph nodes, T stage, mismatch repair status, performance score, PA, BMI, alcohol consumption, age, and sex
BMI—population-based studies
 Asghari-Jafarabadi, 2009, Shahid Beheshti Medical University, Iran [62]
n = 1219
M and W
CRC
Stage I–IV
Hospital record
All-cause mortality (n = NR)
Diagnosis NR
Mean FU 2.1 years
BMI
< 18.5
18.5–24.9
25.0–29.9
≥ 30
2.74 (1.17–6.45)
1.0
0.32 (0.14–0.73)
0.71 (0.25–2.03)
 
Age, alcohol history, inflammatory bowel disease, tumor grade, stage
 Hines, 2009, University of Alabama at Birmingham Hospital, USA [63]
n = 496
M and W
Colon
Stage I–IV
At time of surgery
All-cause mortality (n = 333)
Diagnosis 1981–2002
FU until 2008
BMI
< 18.5
18.5–24.9
≥ 25
1.54 (0.96–2.45)
1.0
0.77 (0.61–0.97)
 
Age, ethnicity, comorbidity, stage, tumor grade, bowel obstruction
 Baade, 2011, Queensland, Australia [48]
n = 1825
M and W
CRC
Stage I–III
5 months after diagnosis
All-cause mortality (n = 462);
CRC-specific mortality (n = 345)
Diagnosis 2003–2004;
Median FU 4.9 years (range 4.0–6.0)
BMI
< 18.5
18.5–24.9
25.0–29.9
≥ 30
2.29 (1.47–3.59)
1.0
0.75 (0.61–0.94)
0.78 (0.59–1.03)
1.74 (1.00–3.04)
1.0
0.75 (0.59–0.97)
0.70 (0.51–0.97)
NR
 Campbell, 2012, CPS II Nutrition Cohort, USA [64]
n = 1957
M and W
Colon
Stage I–III
18 months after diagnosis
All-cause mortality (n = 815);
CRC-specific mortality (n = 380);
CVD-specific mortality (n = 153)
Diagnosis 1994–2007;
Median FU 6.4 years (range 2 days–16.1 years)
BMI
< 18.5
18.5–24.9
25.0–29.9
≥ 30
1.30 (0.82–2.06)
1.0
0.83 (0.70–1.00)
0.93 (0.75–1.17)
0.64 (0.25–1.60)
1.0
0.87 (0.65–1.17)
1.14 (0.81–1.60)
Age, smoking, PA, red meat intake, stage
 Chin, 2012, Taiwan [65]
n = 2135
M and W
Colon
Stage I–III
NR
All-cause mortality (n = NR);
CRC-specific mortality (n NR);
Disease-free survival (n NR)
Diagnosis 1995–2003;
FU at least 5 years or until death
BMI
< 18.5
18.5–24.9
25.0–29.9
≥ 30
1.58 (1.23–2.05)
1.0
0.83 (0.68–1.01)
0.94 (0.74–1.18)
1.33 (0.94–1.87)
1.0
0.96 (0.76–1.22)
1.06 (0.80–1.41)
Stage, age, sex, comorbidities, CEA, hemoglobin, albumin, timing of surgery, postoperative morbidity, tumor site, histolic type, tumor grade
 Kuiper, 2012, Women’s Health Initiative, USA [49]
n = 587
W only
CRC
Stage I–III
0.8 (IQR 0.4–1.7) years after diagnosis (median)
All-cause mortality (n = 108);
CRC-specific mortality (n = 51)
Diagnosis ≥ 1993
Median FU 11.9 years (IQR 10.9–12.9)
BMI
18.5–25.0
25.0–30.0
≥ 30
1.0
0.77 (0.47–1.27)
1.09 (0.65–1.83)
1.0
0.45 (0.22–0.92)
0.95 (0.49–1.85)
Age, study arm, stage, ethnicity, education, alcohol, smoking, and hormone therapy use, pre-diagnostic BMI, time between baseline measurement and diagnosis
 Alipour, 2013, British Columbia Cancer Agency, Canada [66]
n = 913
M and W
Colon
Stage II–III
Recorded at initial consultation
All-cause mortality (n = NR);
CRC-specific mortality (n = NR);
Relapse-free survival (n = NR)
Diagnosis 2001–2005;
Median FU 6.9 (IQR 5.2–8.5) years
BMI
18.5–25.0
25.0–30.0
≥ 30
1.0
0.89 (0.71–1.11)
1.02 (0.78–1.33)
1.0
0.80 (0.61–1.05)
1.05 (0.77–1.42)
Age, gender, stage, number of lymph nodes retrieved, and systemic therapy
 Schlesinger, 2014, PopGen, Germany
[16]
n = 2143
M and W
CRC
Stage I–IV
4 years after diagnosis (mean)
All-cause mortality (n = 349)
Diagnosis 2002–2005;
Mean FU 3.5 years
BMI
< 18.5
18.5–24.9
25.0–29.9
≥ 30
P trend
1.65 (0.79–3.46)
1.0
0.80 (0.62–1.02)
0.84 (0.62–1.14)
0.09
 
Age, sex, alcohol, smoking, tumor site, family history of CRC, metastases and other cancer
 Kroenke, 2016, Kaiser Permanente Northern California, USA
[32]
n = 3408
M and W
CRC
Stage I–III
At diagnosis and 15 months after diagnosis
All-cause mortality (n = 617);
CRC-specific mortality (n = 411)
Diagnosis 2006–2011;
Median FU 3.5 (range 0.0–7.9) years
BMI at diagnosis
< 18.5
18.5–24.9
25.0–29.9
30–34.9
≥ 35
BMI after diagnosis
< 18.5
18.5–22.9
23–24.9
25.0–27.9
28–29.9
30–34.9
≥ 35
3.01 (1.88–4.83)
1.0
0.81 (0.64–1.03)
1.03 (0.77–1.38)
1.63 (1.13–2.33)
3.38 (2.19–5.20)
1.0
0.72 (0.52–1.02)
0.56 (0.41–0.77)
0.39 (0.26–0.58)
0.51 (0.35–0.73)
0.85 (0.56–1.30)
3.35 (1.92–5.87)
1.0
0.77 (0.57–1.03)
1.06 (0.75–1.50)
1.47 (0.96–2.27)
3.21 (1.88–5.47)
1.0
0.69 (0.46–1.05)
0.50 (0.34–0.75)
0.42 (0.26–0.67)
0.56 (0.36–0.85)
0.84 (0.51–1.37)
Sociodemographics, disease severity, treatment, and pre-diagnosis BMI
 Walter, 2016, DACHS, Germany
[33]
n = 3130
M and W
CRC
Stage I–IV
At diagnosis
All-cause mortality (n = 896);
CRC-specific mortality (n = 649);
Recurrence-free survival (n = 828);
Disease-free survival (n = 1024)
Diagnosis 2003–2010;
Median FU 4.9 years
BMI
< 20
20–24.9
25.0–29.9
≥ 30
1.21 (0.89–1.66)
1.0
0.82 (0.70–0.95)
0.80 (0.66–0.98)
0.95 (0.65–1.41)
1.0
0.84 (0.71–1.01)
0.78 (0.62–0.99)
Age, sex, tumor site, stage, alcohol, smoking, use of statins, use of NSAIDs, use of beta-blockers, hyperlipidemia, diabetes mellitus, history of heart failure, myocardial infarction, angina pectoris or stroke, history of other cancer, age × log(time) and history of other cancer × log(time)
 Tamakoshi, 2017, BioBank Japan, Japan [26]
n = 1598
M and W
CRC
Stage I–IV
Within 90 days after diagnosis
All-cause mortality (n = 521)
Diagnosis 2003–2008
Median FU 7.4 years
BMI
< 18.5
18.5–24.9
25.0–29.9
≥ 30
1.40 (1.12–1.76)
1.0
0.80 (0.62–1.05)
1.54 (0.86–2.76)
 
Stratified by sex and institutions and adjusted for age and entry year
BMI—studies in the adjuvant setting
 Meyerhardt, 2003, Intergroup Trial 0089, USA [67]
n = 3438
M and W
Colon
Stage II–III
Day 1 of chemotherapy
All-cause mortality (n = NR);
Recurrence-free survival (n = NR);
Disease-free survival (n = NR)
Diagnosis 1988–1992;
Median FU 9.4 years (max 12.7)
BMI
< 21
21.0–24.9
25.0–27.49
27.5–29.9
≥ 30
P trend
1.15 (0.98–1.35
1.0
1.10 (0.95–1.26)
1.05 (0.90–1.24)
1.11 (0.96–1.29)
0.20
1.06 (0.88–1.27)
1.0
1.06 (0.88–1.27)
1.12 (0.94–1.33)
1.11 (0.94–1.30)
0.17
Age, sex, race, performance status, bowel obstruction, bowel perforation, stage, peritoneal implants, predominant macroscopic pathologic feature, completion of chemotherapy
 Meyerhardt, 2004, Intergroup Trial 0114, USA [68]
n = 1688
M and W
Rectum
Stage II–III
Day 1 of chemotherapy
All-cause mortality (n = NR);
Recurrence-free survival (n = NR);
Disease-free survival (n = NR)
Diagnosis 1990–1992;
Median FU 9.9 years (max 11.8)
BMI
< 20
20.0–24.9
25–26.9
27–29.9
≥ 30
P trend
1.43 (1.08–1.89)
1.0
0.97 (0.80–1.17)
0.95 (0.78–1.15)
1.09 (0.90–1.33)
0.5
1.16 (0.85–1.58)
1.0
1.01 (0.81–1.24)
0.88 (0.71–1.09)
1.08 (0.87–1.33)
0.8
Age, sex, race, performance status, bowel obstruction, extent of bowel wall invasion, number of positive lymph nodes
 Sinicrope, 2013, pooled analyses ACCENT database (21 studies), USA [69]
n = 25,291
M and W
CRC
Stage II–III
At study enrolment
All-cause mortality (n = 7973);
Time to Recurrence (n = 7973);
Disease-free survival (n = 15,946);
Diagnosis NR;
Median FU 7.8 years
BMI
< 20
20–24.9
25.0–29.9
≥ 30
30–34.9
≥ 35
P trend
1.21 (1.11–1.32)
1.0
0.99 (0.94–1.04)
1.10 (1.04–1.17)
1.10 (1.02–1.18)
1.11 (1.00–1.23)
< 0.0001
1.13 (1.04–1.24)
1.0
0.99 (0.94–1.04)
1.06 (1.00–1.13)
1.05 (0.98–1.20)
1.08 (0.98–1.20)
0.007
Age, stage, treatment, sex
BMI—studies in the metastatic setting
 Patel, 2015, South Australia Clinical registry for metastatic CRC, Australia [34]
n = 1174
M and W
CRC
Stage IV
At first diagnosis of metastatic CRC, prior to treatment with chemotherapy
All-cause mortality (n = NR)
Diagnosis ≥ 2006;
Median FU 24 months
BMI
< 18.5
18.5–24.9
25.0–29.9
30.0–34.9
≥ 35
2.21 (1.53–3.19)
1.0
1.23 (1.03–1.46)
1.20 (0.94–1.51)
0.89 (0.64–1.23)
 
Age, sex, synchronous disease, > 1 met site, number of lines of chemotherapy and number of lines of antibody
 Renfro, 2016, ARCAD database (25 studies) [70]
n = 21,149
M and W
CRC
Stage IV
Baseline BMI
All-cause mortality (n = NR);
Progression-free survival (n = NR)
Diagnosis 1997–2012;
Median FU 18.9 months
Continuous BMI
P < 0.001 with an L-shaped pattern; highest risk for patients with the lowest BMI, it decreased until a BMI of approximately 28 kg/m2, and remained similar for patients with higher BMI
 
Age, sex, performance score, cancer site, number of metastatic sites; previous chemotherapy usage; presence of liver, lung and lymph node metastases
Visceral adipose tissue—population-based studies
 Rickles, 2013, University of Rochester Medical Center, USA [71]
n = 219
M and W
CRC
Stage I–III
CT, preoperative visceral fat volume
All-cause mortality (n = NR);
Recurrence-free survival (n = 34);
Disease-free survival (n = NR)
Diagnosis 2003–2010;
Max FU 96 months
Visceral fat volume
Stage I
< median
> median
Stage II
< median
> median
Stage III
< median
> median
1.0
0.67 (0.18–2.59)
1.0
1.97 (0.78–5.02)
1.0
0.43 (0.17–1.07)
Insufficient number of events
1.0
3.76 (1.12–12.57)
1.0
0.39 (0.16–0.99)
Major complication, intraoperative blood transfusion, laparoscopic approach, smoking history, gender, age, use of adjuvant or neoadjuvant chemotherapy, and tumor size
 Black, 2017, Aberdeen Royal Infirmary, UK [35]
n = 339
M and W
CRC
Stage I–III
CT, preoperative visceral fat index
All-cause mortality (n = 213)
Diagnosis 2006–2014;
Median FU 62 months (range 3–105)
Visceral fat index
High
Medium
Low
1.00 (0.80–1.26)
 
Age, sex, stage, neoadjuvant therapy, adjuvant therapy, lymphovascular invasion, neutrophil count, subcutaneous fat index, skeletal muscle index
 Caan, 2017, Kaiser Permanente Northern California, USA [39••]
n = 3262
M and W
CRC
Stage I–III
CT, within 4 months of diagnosis and before chemotherapy or radiation, visceral fat area
All-cause mortality (n = 788);
CRC-specific mortality (n = 433)
Diagnosis 2006–2011;
Median FU 5.8 years (range 0.0–9.9)
Body composition
Normal
High visceral adiposity and normal muscle
High visceral adiposity and low muscle
1.0
1.22 (0.99–1.49)
1.40 (1.05–1.87)
 
Age, sex, race, stage, chemotherapy, radiation, tumor site, partitioned BMI, subcutaneous adiposity
Visceral adipose tissue—studies in the adjuvant setting
 Clark, 2013, Moffit Cancer Center, USA [72]
n = 96
M and W
Rectum
Stage II–III
CT, diagnostic visceral fat area to subcutaneous fat area ratio and perinephric fat thickness
All-cause mortality (n = NR);
Disease-free survival (n = 26)
Diagnosis 1998–2010;
max FU 7 years
Visceral fat area to subcutaneous fat area ratio
< 0.4
≥ 0.4
Perinephric fat thickness, mm
1.0
2.03 (0.57–7.20)
1.04 (0.99–1.09)
 
Grade and pathologic response
 Lee, 2015, St. Vincent’s University Hospital, Ireland [73]
n = 62
M and W
CRC
Stage I–III
CT, preoperative visceral fat area
All-cause mortality (n = NR);
Disease-free survival (n = NR)
Diagnosis 2006–2009;
Median FU 62.5 months
Visceral fat area
< 130 cm2
> 130 cm2
1.0
7.0 (2.0–24.6)
 
T stage, N stage
Visceral adipose tissue—studies in the metastatic setting
 Guiu, 2010, Georges-François Leclerc Cancer Centre, France [74]
n = 120
M and W
CRC
Stage IV
CT, pre-treatment visceral fat area
All-cause mortality (n = 22);
Disease progression (n = 92)
Diagnosis 2002–2008;
Mean FU 24 months
Visceral fat area, cm2
Bevacizumab group
< 117.88
≥ 117.58
P
Chemotherapy group
< 117.88
≥ 117.58
P
1.0
2.88
0.027
1.0
NR
NS
 
Performance status, CEA, high subcutaneous fat area
Muscle mass—population-based studies
 Miyamoto, 2015, Kumamoto University Hospital, Japan [75]
n = 220
M and W
CRC
Stage I–III
CT, preoperative skeletal muscle index
All-cause mortality (n = 37);
Recurrence-free survival (n = 85)
Diagnosis 2005–2010;
Median FU 41.4 months
Skeletal muscle index
Q1–3
Q4
1.0
2.27 (1.15–4.49)
1.0
2.18 (1.20–3.94)
Sex, performance score, tumor site, histological findings, preoperative serum CEA level
 Malietzis, 2016, St Mark’s Hospital, UK [38]
n = 805
M and W
CRC
Stage I–IV
CT, preoperative skeletal muscle index
All-cause mortality (n = 156);
Disease-free survival (n = 101)
Diagnosis 2006–2011;
Median FU 47 months (IQR 24.9–65.6)
Skeletal muscle index
Normal
Low
1.0
1.70 (1.25–2.31)
 
Age, ASA score, surgical approach, stage, tumor grade, lymphovascular invasion, adjuvant chemotherapy
 Black, 2017, Aberdeen Royal Infirmary, UK [35]
n = 339
M and W
CRC
Stage I–III
CT, preoperative skeletal muscle index
All-cause mortality (n = 213)
Diagnosis 2006–2014;
Median FU 62 months (range 3–105)
Skeletal muscle index
Normal
Low
1.0
0.76 (0.35–1.65)
 
Age, sex, stage, neoadjuvant therapy, adjuvant therapy, lymphovascular invasion, neutrophil count, subcutaneous fat index, visceral fat index
 Caan, 2017, Kaiser Permanente Northern California, USA [39••]
n = 3262
M and W
CRC
Stage I–III
CT, within 4 months of diagnosis and before chemotherapy or radiation, skeletal muscle index and muscle cross-sectional area
All-cause mortality (n = 788);
CRC-specific mortality (n = 433)
Diagnosis 2006–2011;
Median FU 5.8 years (range 0.0–9.9)
Skeletal muscle index
Normal
Low
Muscle, cm2
Low tertile 1
Middle tertile 2
High tertile 3
P trend
1.0
1.27 (1.09–1.48)
1.32 (1.07–1.64)
1.13 (0.93–1.37)
1.0
0.01
1.0
1.46 (1.19–1.79)
1.54 (1.16–2.05)
1.19 (0.92–1.55)
1.0
0.003
Age, sex, race, stage, chemotherapy, radiation, tumor site, partitioned BMI, total adiposity
Muscle mass—studies in the adjuvant setting
 Jung, 2015, Seoul National University Bundag Hospital, South Korea [37]
n = 229
M and W
Colon
Stage III
CT, preoperative psoas muscle cross-sectional area
All-cause mortality (n = 30);
Disease-free survival (n NR)
Diagnosis 2003–2010;
Median FU 61.3 months (IQR 49.7–72.0)
1 SD decrement in the psoas index
1.85 (1.10–3.13)
 
Age, sex, T stage, N stage, chemotherapy dose intensity, comorbidities, and BMI
Muscle mass—studies in the metastatic setting
 van Vledder, 2012, Erasmus Medical Center, the Netherlands [76]
n = 196
M and W
CRC
Stage IV
CT, perioperative skeletal muscle mass
All-cause mortality (n = 84);
Disease-free survival (n NR)
Diagnosis2001–2009;
Median FU 29 (1–97) months
Skeletal muscle mass
Normal
Low
1.0
2.69 (1.67–4.32)
 
No. of metastases, radiofrequency ablation, resection margin
 Thoresen, 2013, St. Olav’s University Hospital/Cross Cancer Institute, Norway/Canada [77]
n = 71
M and W
CRC
Stage IV
CT, skeletal muscle mass cross-sectional area
All-cause mortality (n = 60)
Diagnosis 2004–2006;
Median FU 15.8/20.6 months
Skeletal muscle mass
Normal
Low
1.0
1.74 (0.99–3.03)
 
Nation, age, and gender
 Blauwhoff-Buskermolen, 2016, Vrije Universiteit Medical Center, the Netherlands [36]
n = 67
M and W
CRC
Stage IV
CT, skeletal muscle area
Overall mortality (n NR)
Diagnosis 2011–2014;
Median FU 17.5 months (95% CI 13.3–21.7) for patients receiving first-line chemotherapy and 8.5 months (95% CI 4.4–12.6) for patients receiving second-line chemotherapy or beyond
Muscle mass
Normal
Low
1.0
1.65 (0.85–3.18)
 
Sex, age, lactate dehydrogenase concentration, comorbidity, metastases, chemotherapy line
CRC colorectal cancer, HR hazard ratio, 95% CI 95% confidence interval, SD standard deviation, IQR interquartile range, M men, W women, NR not reported, NS non-significant, Q quintile or quartile, C category, BMI body mass index, PA physical activity, MET-h metabolic equivalent task-hour, CT computed tomography, CEA carcinoembryonic antigen, MSI microsatellite instability, CALGB Cancer and Leukemia Group B, CPS II Cancer Prevention Study II, DACHS German: Darmkrebs: Chancen der Verhutüng durch Screening, English: chances for prevention through screening; ACCENT Adjuvant Colon Cancer Endpoints, ARCAD Aide et Recherche en Cancérologie Digistive
Results are for disease recurrence
In this review, we summarized the available evidence for dietary patterns, red and processed meat, sugar-sweetened beverages, alcohol consumption, other foods and beverages, and CRC survival.

Dietary Patterns

Two observational studies, the NHS I [23] and a German cohort of CRC survivors [24], assessed post-diagnosis dietary patterns in a population-based setting [23], while CALGB 89803 [42] reported results in the adjuvant setting (Table 1). Data-driven dietary patterns were assessed within NHS I [23] and CALGB 89803 [42]. Both studies observed patterns that were given the labels a “Western” and a “Prudent” dietary pattern. The Western dietary pattern was characterized by high- and low-fat dairy, refined grains, red and processed meats, desserts, and potatoes, while the Prudent dietary pattern was characterized by high intakes of fruits, vegetables, whole grains, and poultry.
For the Western dietary pattern, both studies reported an increased all-cause mortality risk [23, 42]. However, the association was statistically significant only in the adjuvant setting (CALBG: Q5 vs. Q1: HR 2.32; 95% CI 1.36–3.96; P trend <0.001) [42], and not in the population-based study (NHS I: Q5 vs. Q1: HR 1.32 (0.89–1.97); P trend = 0.23) [23]. Similarly, a statistically significant increased risk of colon cancer recurrence was reported in the adjuvant setting [42], while a non-significant positive association was reported for CRC mortality in the population-based study [23] (Table 1). For the Prudent dietary pattern, both studies reported statistically non-significant associations for all-cause mortality [23, 42], CRC-specific mortality [23], or colon cancer recurrence [42].
Furthermore, several a priori-defined dietary patterns were studied in the two population-based studies [23, 24] (Table 1). Of the a priori-defined dietary patterns, none has been studied in more than one cohort. Some a priori-defined dietary patterns were associated with lower risk of all-cause mortality, but not all [23, 24].
Only one small (n = 111) randomized dietary intervention trial among CRC survivors assessed associations with survival [78]. Throughout the 1.5 months of neoadjuvant radiotherapy patients with rectal cancer randomized to the intervention group received 6 weekly individualized nutrition counseling and education sessions using regular foods, while the control group maintained their usual diet. Overall, the main goal of the intervention was to enable every patient to achieve his or her calculated energy and protein requirements. After long-term follow-up (median follow-up 6.5 (range 4.9–8.1) years), CRC-specific survival was significantly longer in the intervention group after adjustment for age and disease stage (median survival 7.3 vs. 4.9 years).

Red and Processed Meats

Both NHS I [23] and CPS II Nutrition Cohort [40] reported on post-diagnosis red and processed meat intake, although the NHS I paper focused on dietary patterns (Table 1). The CPS II Nutrition Cohort also provided information regarding pre- to post-diagnosis change in red and processed meat consumption [40] (Table 2).
Table 2
Cohort studies among individuals with colon or rectal cancer examining change in lifestyle factors in relation to all-cause mortality, colorectal cancer-specific mortality, or recurrence; changes could be changes from pre- to post-diagnosis or changes made after diagnosis
First author, year, name of cohort, country
Study population
Time of post-diagnosis exposure assessment
Outcomes assessed
Year of CRC diagnosis and follow-up
Lifestyle factor
All-cause mortality HR (95% CI)
Colorectal cancer-specific mortality HR (95% CI)
Covariates
Change in diet—population-based studies
 McCullough, 2013, CPS II Nutrition Cohort, USA [40]
n = 1186
Both genders
Colon and rectum
Stage I–III
Red and processed meat
9 years before CRC diagnosis (mean) and 3 years after diagnosis (mean)
All-cause mortality (n = 472);
CRC-specific mortality (n = 146);
CVD-mortality (n = 110);
other-mortality (n = 216)
Diagnosis 1992–2009;
Mean 7.6 years (SD 3.4 years)
Remained low meat
Remained high meat
Low to high meat
High to low meat
1.0
1.28 (0.98–1.67)
1.25 (0.93–1.67)
1.37 (1.02–1.85)
1.0
1.79 (1.11–2.89)
0.96 (0.55–1.66)
1.43 (0.80–2.57)
Age, sex, stage, 1992 pre-diagnostic energy intake, and post-diagnostic energy intake
 Yang, 2017, CPS II Nutrition Cohort, USA [27]
n = 1599
M and W
CRC
Stage I–III
Alcohol
3.2 years before CRC diagnosis (mean) and 1.9 years after diagnosis (mean)
All-cause mortality (n = 732);
CRC-specific mortality (n = 235);
CVD-mortality (n = 172);
other mortality (n = 325)
Diagnosis 1992–2011;
Mean FU 8.2 years (SD 4.7 years)
Never drinker
Former-former
Current-former
Current < 2 drinks/day
Current ≥ 2 drink/day
1.0
1.09 (0.81–1.48)
1.21 (0.92–1.60)
0.94 (0.77–1.16)
0.92 (0.66–1.28)
1.0
1.28 (0.73–2.23)
1.81 (1.13–2.91)
1.27 (0.87–1.86)
1.44 (0.80–2.60)
Age, sex, tumor stage, smoking, BMI, PA, education, and pre-existing diseases in 1982/1992 (COPD, liver disease, kidney disease)
Change in physical activity—population-based studies
 Meyerhardt, 2006, Nurses’ Health Study I, USA [46]
n = 573
W only
CRC
Stage I–III
6 months before CRC diagnosis (mean) and 22 months after diagnosis (mean)
All-cause mortality (n = 132);
CRC-specific mortality (n = 80)
Diagnosed 1986–2004; Median FU 9.6 years
Decreased PA
No change in PA
Increased PA
MET-hours/week
Stable activity < 9
Stable activity ≥ 9
Increase activity
Pre-diagnosis activity < 9
Pre-diagnosis activity ≥ 9
1.23 (0.79–2.34)
1.0
0.51 (0.30–0.85)
1.0
0.33 (0.11–0.97)
0.26 (0.10–0.66)
0.35 (0.11–1.13)
1.32 (0.74–2.34)
1.0
0.48 (0.24–0.97)
1.0
0.27 (0.09–0.80)
0.36 (0.19–0.67)
0.62 (0.28–1.34)
BMI, stage, differentiation grade, tumor location, age, year of diagnosis, chemotherapy, time from diagnosis to PA measurement, change in BMI, smoking
 Baade, 2011, Queensland, Australia [48]
n = 1825
M and W
CRC
Stage I–III
Pre-diagnosis physical activity was recalled after CRC diagnosis and assessed 5 months after diagnosis
All-cause mortality (n = 462);
CRC-specific mortality (n = 345)
Diagnosis 2003–2004
Mean FU 4.9 years (range 4.0–6.0)
Pre-diagnosis to 5 months post-diagnosis
No change or decreased
Increased ≤ 2 h/week
Increased > 2 h/week
P trend
1.0
1.27 (0.88–1.83)
1.06 (0.65–1.71)
0.449
1.0
1.32 (0.89–1.98)
1.03 (0.59–1.80)
0.389
5 months post-diagnosis PA level, age, stage, smoking status, tumor location, treatment, sex and comorbidities
n = 1554
M and W
CRC
Stage I–III
5 and 12 months after CRC diagnosis
  
5 to 12 months post-diagnosis
No change or decreased
Increased ≤ 2 h/wk.
Increased > 2 h/wk.
P trend
1.0
0.79 (0.59–1.04)
0.69 (0.50–0.94)
0.030
1.0
0.68 (0.48–0.97)
0.64 (0.44–0.93)
0.015
 
Smoking cessation—population-based studies
 Phipps, 2011, Seattle Colon Cancer Family Registry, USA, [79]
n = 2264
M and W
CRC
Stage I–IV
2 years before CRC diagnosis and 8.0 months after diagnosis (mean)
All-cause mortality (n = 831);
CRC-specific mortality (n = 562)
Diagnosis 1998–2007;
FU up to 2010
Remained never smoker
Remained former smoker
Continued smoking
Quit smoking
1.0
1.26 (1.07–1.48)
1.50 (1.14–1.97)
1.52 (1.21–1.90)
1.0
1.14 (0.93–1.38)
1.47 (1.07–2.03)
1.32 (1.00–1.74)
Age, time from diagnosis to interview, history of preventive screening, sex, and education level
 Tao, 2013, Shanghai Cohort Study, China [28]
n = 114
M only
CRC
Stage NR
Yearly assessments of smoking cessation after diagnosis among smokers at diagnosis
All-cause mortality (n = 73)
Diagnosis 1986–2010;
Mean FU 5.3 ± 4.8 years
Quit smoking
Intermittent smoking
Continued smoking Time-dependent smoking
1.0
1.35 (0.68–2.67)
3.46 (1.69–7.10)
2.31 (1.40–3.81)
 
Age, education, pack-years of smoking before diagnosis, treatment, and cancer site
 Walter, 2015, DACHS study, Germany [58]
n = 3130
M and W
CRC
Stage I–IV
Smoking cessation in the year of diagnosis.
All-cause mortality (n = 889)
CRC-specific mortality (n = 828),
Non-CRC related mortality (n = 644),
Recurrence-free survival (n = 1024),
Disease-free survival (n = 232)
Diagnosis 2003–2010;
Median FU 4.9 years.
Nonsmokers
Recent quitters
Continued smokers
1.0
0.97 (0.70–1.33)
1.10 (0.86–1.41)
1.0
0.87 (0.60–1.25)
1.10 (0.83–1.45)
Age, sex, BMI, stage, alcohol consumption, red meat consumption, family history of CRC, use of statins, use of NSAIDs, use of beta-blockers, diabetes mellitus, history of heart failure, myocardial infarction, angina pectoris or stroke, history of non-CRC cancer; additional adjustment for age × log(time) to account for time-dependent effects
 Yang, 2015, CPS II Nutrition Cohort, USA [29]
n = 2256
M and W
CRC
Stage I–III
2.3 years before CRC diagnosis (mean) and 1.4 years after diagnosis (mean)
All-cause mortality (n = 859);
CRC-specific mortality (n = 323)
Diagnosis 1992–2009;
Mean FU 7.5 years (SD 4.6 years)
Remained never smoker
Remained former smoker
Continued smoking
Quit smoking
1.0
1.18 (1.00–1.39)
2.33 (1.62–3.34)
1.94 (1.29–2.91)
1.0
0.86 (0.66–1.11)
2.20 (1.29–3.76)
1.85 (1.02–3.35)
Age, sex, stage, alcohol consumption, BMI, and PA
Weight change—population-based studies
 Baade, 2011, Queensland, Australia [48]
n = 1763
M and W
CRC
Stage I–III
Pre-diagnosis weight was recalled after CRC diagnosis and assessed 5 months after diagnosis
All-cause mortality (n = 462);
CRC-specific mortality (n = 345)
Diagnosis 2003–2004
Mean FU 4.9 years (range 4.0–6.0)
Pre-diagnosis to 5 months post-diagnosis
> 5 kg loss
2–4.9 kg loss
± 2 kg
2–4.9 kg gain
> 5 kg gain
P trend
1.63 (1.29–2.06)
1.10 (0.83–1.46)
1.0
1.12 (0.60–2.09)
1.63 (1.02–2.61)
< 0.001
1.64 (1.24–2.15)
1.02 (0.73–1.42)
1.0
0.90 (0.41–1.96)
1.46 (0.84–2.53)
0.001
5 months post-diagnosis weight, height, PA level, stage, smoking status, tumor site, treatment, sex and comorbidities
 
n = 1503
M and W
CRC
Stage I–III
5 months and 12 months after CRC diagnosis
  
5 to 12 months post-diagnosis
> 5 kg loss
2–4.9 kg loss
± 2 kg
2–4.9 kg gain
> 5 kg gain
P trend
2.92 (1.89–4.49)
1.68 (1.10–2.59)
1.0
0.95 (0.68–1.32)
0.91 (0.69–1.20)
< 0.001
3.21 (1.95–5.31)
1.59 (0.95–2.68)
1.0
1.02 (0.69–1.50)
0.89 (0.64–1.25)
< 0.001
 
 Meyerhardt, 2016, Kaiser Permanente Northern Carolina, USA [80]
n = 2781
M and W
CRC
Stage I–III
Within 3 months after CRC diagnosis (prior to surgery) and approximately 18 months after diagnosis (range 15–21)
All-cause mortality (n = 549);
CRC-specific mortality (n = 311)
Diagnosis 2006–2011;
Median FU 4.2 years (range 0.1–8.1 years)
% Weight change
≥ 10% loss
5–9.9% loss
± 4.9%
5–9.9% gain
≥ 10% gain
P trend loss
P trend gain
3.27 (2.56–4.18)
1.74 (1.34–2.25)
1.0
0.86 (0.65–1.14)
1.20 (0.91–1.58)
< 0.0001
0.27
3.20 (2.33–4.39)
1.58 (1.12–2.23)
1.0
0.84 (0.58–1.22)
0.93 (0.63–1.37)
< 0.0001
0.54
Age, weight at diagnosis, gender, race/ethnicity, stage, grade, chemotherapy, and tumor site
Weight change—studies in the adjuvant setting
 Meyerhardt, 2008, CALGB 89803, USA [81]
n = 1053
M and W
Colon
Stage III
During and 6 months after adjuvant chemotherapy
All-cause mortality (n = 261);
Recurrence-free survival (n = 338);
Disease-free survival (n = 369)
Diagnosis 1999–2001;
Median FU 5.3 years
Weight change (kg)
> 5 loss
2.1–5 loss
± 2
2–4.9 gain
≥ 5 gain
1.13 (0.44–2.93)
0.89 (0.31–2.57)
1.0
0.97 (0.43–2.18)
1.23 (0.65–2.31)
1.35 (0.64–2.81)
1.04 (0.46–2.35)
1.0
1.00 (0.52–1.95)
1.17 (0.70–1.96)
Sex, age, T stage, number of positive lymph nodes, presence of clinical perforation at time of surgery, presence of bowel obstruction at time of surgery, performance status, treatment arm, time between questionnaire one and questionnaire two, time-varying BMI, smoking status at time of questionnaire two, PA level
 Vergidis, 2016, British Columbia Cancer Agency, Canada [82]
n = 539
M and W
Colon
Stage III
At initial oncology consultation visit before the receipt of any systemic therapy and follow-up weights were serially reported at each subsequent clinic visit throughout the entire course of their adjuvant treatment or until 9 months after their first clinic visit, whichever came later. The peak and nadir weights were used to calculate weight change
All-cause mortality (n NR);
Recurrence-free survival (n NR)
Diagnosis 2008–2010;
FU 3–5 years
Weight gain
< 5%
≥ 5%
< 10%
≥ 10%
Weight loss
< 5%
≥ 5%
< 10%
≥ 10%
Weight change
< 5%
≥ 5%
< 10%
≥ 10%
1.0
0.80 (0.39–1.66)
1.0
0.52 (0.24–1.20)
1.0
1.92 (1.00–3.70)
1.0
2.63 (1.04–6.67)
1.0
1.02 (0.54–1.95)
1.0
1.15 (0.59–2.22)
1.0
0.84 (0.46–1.53)
1.0
0.81 (0.40–1.65)
1.0
1.56 (0.88–2.28)
1.0
2.94 (1.39–6.25)
1.0
1.44 (0.79–2.64)
1.0
1.10 (0.64–1.83)
Age, sex, comorbidities, performance status, tumor site, stage, grade, receipt of systemic therapy, type of regimen received
Muscle loss—studies in the metastatic setting
 Blauwhoff-Buskermolen, 2016, Vrije Universiteit Medical Center, the Netherlands [36]
n = 67
M and W
CRC
Stage IV
Skeletal muscle area was measured using CT scans conducted before start of chemotherapy and during chemotherapy
Overall mortality (n = NR)
Diagnosis 2011–2014;
Median FU 17.5 months (95% CI 13.3–21.7) for patients receiving first-line chemotherapy and 8.5 months (95% CI 4.4–12.6) for patients receiving second-line chemotherapy or beyond
Muscle loss
< 9%
≥ 9%
1.0
4.47 (2.21–9.05)
 
Sex, age, lactate dehydrogenase concentration, comorbidity, metastases, chemotherapy line, tumor progression at first evaluation by CT scan
CRC colorectal cancer, HR hazard ratio, 95% CI 95% confidence interval, M men, W women, NR not reported, BMI body mass index, PA physical activity, MET-h metabolic equivalent task-hour, CT computed tomography, CPS II Cancer Prevention Study II, CALGB Cancer and Leukemia Group B, DACHS German: Darmkrebs: Chancen der Verhutüng durch Screening, English: chances for prevention through screening
Results are for disease recurrence
These two studies did not observe an association between red and processed meat intake and both all-cause mortality and CRC-specific mortality [23, 40]. Furthermore, changing meat intake from high (median or higher) before CRC diagnosis to low (below median) after diagnosis was not associated with lower mortality when compared to survivors with a consistently high intake [40].

Sugar-Sweetened Beverages

Both the NHS I [23] and CALGB 89803 [43] reported on post-diagnosis sugar-sweetened beverage intake and CRC outcomes (Table 1).
Both studies [23, 43] reported increased all-cause mortality risk for sugar-sweetened beverage consumption after CRC diagnosis, of which the association in the NHS I was statistically significant [23]. Each additional serving of sugar-sweetened beverages (including fruit juices) after CRC diagnosis was associated with an 11% increased risk for all-cause mortality (HR 1.11; 95% CI 1.01–1.23) [23]. A similar relative risk was reported for CRC-specific mortality, although it was not statistically significant [23]. For colon cancer recurrence, CALGB 89803 reported a statistically significant increased recurrence risk for patients consuming ≥ 2 servings of sugar-sweetened beverages per day (HR 1.75; 95% CI 1.04–2.94) compared to those consuming < 2 servings per month (P trend = 0.04) [43].

Alcohol

Four population-based studies, NHS I [23, 44], HPFS [44], CPS II Nutrition cohort [27], and a Japanese cohort of CRC survivors [26], reported on post-diagnosis alcohol consumption and CRC outcomes (Table 1).
In NHS I, moderate drinking was used as the reference group and abstaining from alcohol consumption was associated with a statistically significant increased all-cause mortality risk (HR 1.30; 1.05–1.61) compared to women consuming 5–15 g of alcohol per day [23]. Drinking > 15 g/day (approximately 1.5 drinks) was not statistically significantly associated with increased mortality risk. Similarly, abstainers had a higher mortality risk than drinkers in the Japanese cohort [26] and after combining both NHS I and HPFS cohort data [44]. However, the CPS II Nutrition cohort reported that drinking alcohol after diagnosis was not associated with all-cause mortality [27]. For CRC-specific mortality, similar results were reported as for all-cause mortality (Table 1).
The CPS II Nutrition cohort also provided information regarding pre- to post-diagnosis change in alcohol consumption (Table 2). Participants who reported drinking before CRC diagnosis but stopped drinking alcohol after diagnosis had a statistically non-significant increased risk of all-cause and CRC-specific mortality compared to participants who continued to drink alcohol [27].

Other Foods, Beverages, and Nutrients

The intake of some foods, beverages, and nutrients were only reported in one study each (Table 1). Higher nut consumption was associated with lower risk of CRC mortality (HR/serving/day 0.69; 95% CI 0.49–0.97) in the NHS I, while no statistically significant association was reported for all-cause mortality [23]. Furthermore, no associations were observed within the NHS I with either all-cause mortality or CRC-specific mortality for vegetables, fruits, or whole grains [23]. However, in the Japanese study, lower green leafy vegetable intake after CRC diagnosis was associated with an increased all-cause mortality risk [26].
Higher milk intake was statistically significantly associated with lower all-cause mortality risk (Q4 vs. Q1: HR 0.72; 95% CI 0.55–0.94; P trend = 0.02) in the CPS II Nutrition Cohort [41]. A similar risk was reported for overall dairy consumption, although associations did not reach statistical significance [41]. Additionally, higher coffee intake was statistically significantly associated with lower all-cause mortality (≥ 4 vs. 0 cups/day: HR 0.66; 95% CI 0.37–1.18; P trend = 0.01) within CALGB 89803 [25]. No significant associations were reported for non-herbal tea intake [25].
Higher dietary glycemic load and total carbohydrate intake were statistically significant associated with an increased risk of mortality and recurrence in CALGB 89803 [45]. Higher total calcium intake was statistically significantly associated with both lower all-cause mortality and CRC-specific mortality in the CPS II Nutrition Cohort, while no significant associations were reported for vitamin D [41]. Also no significant associations were reported for intake of one-carbon nutrients (folate, vitamins B6 and B12) in NHS I [44].

Diet: Key Points

One small randomized intervention trial which provided individualized nutritional counseling and education about regular foods suggest that making dietary changes may improve cancer-specific survival. No dietary pattern or food has been studied in more than two observational cohorts, with cancer recurrence only studied in one cohort in the adjuvant setting embedded in a randomized chemotherapy trial. While alcohol consumption has been studied more frequently, these studies often used abstainers as comparison group. Abstainers are probably an inappropriate reference group, as this group may, at least in part, include people who stopped drinking because of comorbidities or cancer-related symptoms. Overall, emerging evidence shows that diet after CRC diagnosis might affect survival, but further research is needed to clarify what aspects of diet are important and which dietary changes could affect survival.

Physical Activity after CRC Diagnosis

Seven population-based studies [26, 4651] and one study in the adjuvant setting [52] provided results on physical activity after CRC diagnosis and mortality outcomes (Table 1). Five large US cohorts assessed post-diagnosis physical activity in population-based cohorts with > 500 CRC patients: NHS I [46], HPFS [47], CPS II Nutrition Cohort [50], Women’s Health Initiative [49], and National Institutes of Health-AARP Diet and Health Study [51]. All five cohorts consist of participants diagnosed with CRC during follow-up and have updated physical activity assessment after diagnosis, usually when treatment was completed. In contrast, two non-US cohorts (an Australian cohort [48] and BioBank Japan [26]) recruited > 1500 CRC patients after CRC diagnosis. All studies reported on leisure time physical activity.

Physical Activity

For all-cause mortality, seven studies [26, 4652] were included in previous meta-analyses [710]. These meta-analyses have found highest versus lowest post-diagnostic physical activity to be associated with 40% lower all-cause mortality risk [710]. Five studies that were included in a dose-response meta-analysis showed a 28% lower risk of all-cause mortality (HR 0.72; 95% CI 0.65–0.80) for every 10 metabolic equivalent task-hour per week (MET-hours/week) increase in post-diagnosis physical activity [9], which is equivalent to current recommendations of 150 min/week of at least moderate intensity activity. For CRC-specific mortality, similar risk reductions were reported comparing high versus low physical activity after CRC diagnosis (HR 0.62; 95% CI 0.45–0.86) [11••] and for every 10 MET-hours/week increase in post-diagnosis physical activity (HR 0.75; 95% CI 0.65–0.85) [9].

Changes in Physical Activity

The Australian cohort [48] and NHS I [46] also provided results on changes in physical activity and mortality outcomes in CRC patients (Table 2). An increase of physical activity > 2 h/week between 5 and 12 months post-diagnosis was statistically significantly associated with lower all-cause (HR 0.69; 95% CI 0.50–0.94) and CRC-specific mortality (HR 0.64; 95% CI 0.44–0.93) among Australian CRC survivors [48]. A pre- to post-diagnosis increase in physical activity showed a statistically significant lower all-cause and CRC-specific mortality risk in the NHS I [46], but no association was reported among Australian CRC survivors [48] (Table 2). The first randomized controlled trial designed primarily to assess the impact of physical activity on survival among colon cancer survivors is ongoing [83]. As of April 2017, the trial has enrolled 536 of its planned 972 participants [84] and only 1 year feasibility results have been published so far [85].

Sedentary Behavior

Three of the population-based studies, CPS II Nutrition Cohort [50], National Institutes of Health-AARP Diet and Health [51], and HPFS [53] also reported on post-diagnosis sedentary behavior and all-cause as well as CRC-specific mortality (Table 1). CPS II reported on leisure time spent sitting [50], whereas the other two studies assessed TV viewing [51, 53]. All three studies [50, 51, 53] reported no statistically significant associations between sedentary behavior and all-cause mortality. With regard to CRC-specific mortality, only one study, the CPS II Nutrition Cohort showed a statistically significant positive association between sedentary behavior and CRC-specific mortality (≥ 6 h vs. < 3 h/day sitting time: HR 1.62; 95% CI 1.07–2.44) [50].

Physical Activity: Key Points

Evidence from prospective observational studies has consistently suggested that higher physical activity after CRC diagnosis is associated with a lower risk of CRC-specific and all-cause mortality, but whether physical activity is causally related to CRC mortality remains unclear. A randomized controlled trial is currently ongoing to address whether aerobic physical activity after complement of adjuvant therapy improves survival. Based on a few studies, there is some evidence suggesting that excessive sedentary behavior after CRC diagnosis might be associated with increased CRC-specific mortality, but findings are less consistent than for leisure time physical activity.

Smoking after CRC Diagnosis

Eleven population-based studies [14, 26, 2831, 5458] and three studies in the adjuvant setting [5961] reported on smoking at or after CRC diagnosis and mortality outcomes (Table 1). Four population-based studies used data from a cancer registry [14, 30, 31, 55]; three were from single-institution hospital cohorts [54, 56, 57]; three were non-US cohorts (Shanghai Cohort Study [28], the German cohort DACHS [58], and BioBank Japan [26]); and lastly, the CPS II Nutrition cohort [29]. Two studies in the adjuvant setting were embedded in an adjuvant chemotherapy trial, CALGB 89803 [60] and N0147 [61], while the third study included patients referred to a single institution for consideration of adjuvant treatment [59]. Six studies [28, 31, 54, 5759] compared current smokers with non-smokers, while eight studies [14, 26, 29, 30, 55, 56, 60, 61] compared current smokers with never smokers.

Smoking

For all-cause mortality, eight out of nine population-based studies [26, 28, 29, 31, 5457] reported increased all-cause mortality risk for smoking, of which six [26, 28, 29, 54, 55, 57] were statistically significant. Furthermore, the study in the adjuvant setting also reported a statistically significant increased all-cause mortality risk for smoking [61].
For CRC-specific mortality, five population-based studies [14, 29, 30, 57, 58] reported increased CRC-specific mortality risk for smoking, of which three [14, 29, 30] were statistically significant (Table 1). However, one study that reported results separately for men and women reported a statistically non-significant positive association among women for post-diagnosis smoking, while among men, a statistically non-significant inverse association was reported [56]. Furthermore, one study in the adjuvant setting also reported a statistically significant increased CRC-specific mortality risk for smoking [59].
For colon cancer recurrence, one study embedded in the trial N0147 [61] reported a statistically significant increased cancer recurrence risk for smoking, while CALGB 89803 [60] reported no association with smoking among stage III colon cancer patients treated with adjuvant chemotherapy.

Smoking Cessation

Four population-based studies provided results on smoking cessation and mortality outcomes in CRC patients (Table 2). People who continued smoking after CRC diagnosis had a more than threefold increased risk of all-cause mortality (HR 3.46; 95% CI 1.69–7.10) compared to people who quit smoking after diagnosis [28]. Pre- to post-diagnosis smoking cessation was not statistically significantly associated with all-cause or CRC-specific mortality risk [29, 58, 79], although one of these studies reported lower mortality risk for those who quit smoking compared to those who continued to smoke [29].

Smoking: Key Points

Overall, evidence from observational studies has consistently suggested that smoking after CRC diagnosis increases the risk of CRC-specific and all-cause mortality. It seems plausible that smoking cessation would improve survival outcomes in CRC survivors, although direct evidence is limited.

Body Fatness and Body Composition after CRC Diagnosis

This review first focusses on studies that assessed BMI at or after CRC diagnosis. Next, we discuss weight changes and lastly, we describe the results of studies which quantified visceral adipose tissue or skeletal muscle mass from CT images.

Body Mass Index

Eleven population-based studies [16, 26, 32, 33, 48, 49, 6266], two studies from adjuvant chemotherapy trials [67, 68], and one study among metastatic patients [34] assessed the association of BMI at or after CRC diagnosis and CRC outcomes (Table 1). Furthermore, 21 additional studies in the adjuvant setting were included in a pooled analyses of patients enrolled in trials of adjuvant chemotherapy [69]. Moreover, an additional article with pooled analyses in the metastatic setting included data of 25 treatment trials [70].
For underweight (either BMI < 18.5 or 20 kg/m2), all population-based studies [16, 26, 32, 33, 48, 6265], the pooled analysis of studies in the adjuvant setting [69], and both publications in the metastatic setting [34, 70] reported higher all-cause mortality risk compared to normal weight individuals. The majority of these studies [26, 32, 34, 48, 62, 65, 69, 70] reported statistically significant results (Table 1). In the largest population-based study, ~ 3400 men and women diagnosed with stage I to III CRC from the Kaiser Permanente Northern California population, underweight at diagnosis was associated with a threefold increased all-cause mortality risk (HR 3.01; 95% CI 1.88–4.83) compared to normal weight [32]. However, most other studies report a 1.5- to 2-fold increased risk (Table 1). Generally, similar results were reported for CRC-specific mortality and cancer recurrence (Table 1).
For overweight (defined as BMI 25.0–24.9 kg/m2), all population-based studies [16, 26, 32, 33, 48, 49, 62, 6466] reported lower all-cause mortality risk compared to normal weight individuals, of which three were statistically significant [48, 49, 62]. However, studies in the adjuvant setting of a chemotherapy trial reported that overweight individuals had a similar all-cause mortality risk as normal weight individuals (Table 1). For metastatic patients participating in treatment trials, all-cause mortality risk was lowest at BMI 28 kg/m2 [70], while overweight was associated with an increased all-cause mortality risk among a general population of patients diagnosed with metastatic disease (HR 1.23; 95% CI 1.03–1.46) [34]. Generally, similar results were reported for CRC-specific mortality and cancer recurrence (Table 1).
For obesity (BMI ≥ 30 kg/m2), none of the population-based studies [16, 26, 32, 33, 48, 49, 62, 6466] reported statistically significant associations with all-cause mortality. Nevertheless, the only study (Kaiser Permanente Northern California cohort) that reported on a separate group with class II or III obesity (BMI ≥ 35 kg/m2) reported a statistically significant increased all-cause mortality risk [32]. Within the adjuvant setting pooled analyses showed a modest increased all-cause mortality risk (HR 1.10; 95% CI 1.04–1.17) compared with normal weight [69]. Within the metastatic setting, both publications showed that obese individuals had a somewhat similar, or lower, all-cause mortality risk as normal weight individuals [34, 70]. Generally, similar results were reported for CRC-specific mortality and cancer recurrence (Table 1).

Changes in Weight

Four studies [48, 8082] reported on weight changes (Table 2). Two studies were population-based studies, a cohort from the Kaiser Permanente Northern California population [80] and an Australian cohort [48], and two studies were in the adjuvant setting, CALGB 89803 [81] and a cohort from the British Columbia Cancer Agency [82].
Large post-diagnosis weight loss (> 5 kg or ≥ 10%) was associated with a threefold increased all-cause and CRC mortality risk compared with stable weight in both population-based studies [48, 80]. Modest weight loss (2–4.9 kg or 5–9.9%) was also associated with increased all-cause and CRC mortality risk [48, 80], although only statistically significant in the Kaiser Permanente Northern California cohort [80]. In fact, the association between weight loss and mortality was present regardless of at-diagnosis BMI [80]. Large weight loss during adjuvant chemotherapy was associated with increased all-cause mortality and recurrence risk in a cohort from the British Columbia Cancer Agency [82], but not in CALGB 89803 [81].
Post-diagnosis weight gain was not associated with increased all-cause or CRC-specific mortality risk [48, 80, 81] or colon cancer recurrence [81, 82]. Furthermore, pre- to post-diagnosis weight loss or weight gain of >5 kg were both associated with a statistically significant 60% higher all-cause risk compared to stable weight [48].

Visceral Adipose Tissue

Three population-based studies [35, 39, 71], two studies in the adjuvant setting [72, 73], and one study among metastatic patients [74] reported on post-diagnosis visceral adipose tissue and all-cause mortality (Table 1). Most of these studies were small (n = 62 to 339), except for the population-based cohort from the Kaiser Permanente Northern California population (n ~ 3200) [39••].
For all-cause mortality, all population-based studies [35, 39••, 71] reported statistically non-significant associations with visceral adipose tissue (Table 1). Both studies among patients treated with chemotherapy [72, 73] reported an increased all-cause mortality risk with high visceral adipose tissue, of which one was statistically significant [72]. The study among metastatic CRC patients [74] reported a statistically significant increased all-cause mortality risk for high visceral adipose tissue among patients treated with chemotherapy plus the angiogenesis inhibitor bevacizumab, but not among patients treated with chemotherapy only.

Skeletal Muscle Mass

Four population-based studies [35, 38, 39••, 75], one study in the adjuvant setting [37] and three studies among patients with metastatic disease [36, 76, 77] reported on all-cause mortality (Table 1). Most of these studies were small (n = 67 to 339), except two population-based cohorts, from the Kaiser Permanente Northern California population (n ~ 3200) [39••] and from a single-institution hospital cohort that included stage I–IV patients [38].
Seven out of eight studies [3638, 39••, 7577] reported increased all-cause mortality risk for low skeletal muscle mass, of which five were statistically significant [37, 38, 39••, 75, 76] (Table 1). A meta-analysis, based on three small studies [7577], concluded that a low muscle mass was statistically significantly associated with a more than twofold increased all-cause mortality risk (HR 2.25; 95% CI 1.63–3.09) [20]. The only large population-based cohort with non-metastatic patients, from Kaiser Permanente Northern California, showed an almost 30% increased risk of overall mortality and 50% increased risk of CRC-specific mortality [39••].
One study among metastatic patients reported on loss of muscle mass during chemotherapy [36]. This study showed that ≥ 9% loss of muscle mass during chemotherapy was associated with a more than fourfold increased all-cause mortality risk (HR 4.47; 95% CI 2.21–9.05) [36].

Body Fatness and Body Composition: Key Points

Body fatness was studied most often by assessment of body mass index, while only few studies assessed other measures of body composition. Altogether, the results of studies across the three study categories (population-based, adjuvant, and metastatic setting) suggest a J- or L-shaped association between BMI and all-cause mortality or CRC-specific mortality risk. The risk of death was highest among patients who were underweight, while lowest risk was seen in patients with a BMI between 25 and < 30 kg/m2. If obesity confers an additional mortality risk compared to normal weight or overweight patients remains uncertain. Nevertheless, the most recent meta-analysis of post-diagnosis BMI concluded that obesity was statistically significantly associated with a modest 8% increased all-cause mortality risk (HR 1.08; 95% CI 1.03–1.13) compared to normal weight, while no association was found between obesity and CRC-specific mortality [17••]. Weight loss in the first 2 years after diagnosis was consistently associated with increased mortality risk and this association was independent of BMI at CRC diagnosis. Currently, there are no intentional weight loss trials among CRC survivors that assessed mortality risk [86••] and no study that assessed the effect of weight loss after treatment was successfully completed. That being overweight, and in some studies even obese states, seem to be associated with improved survival compared to normal weight is called the “obesity paradox.” The obesity paradox could be explained by several methodological issues, including the crudeness of BMI as a measure of body fatness, especially in a cancer patient population where loss of weight and lean body mass is a strong adverse factor [87].
Other measures used to study the association between body composition and CRC outcomes were visceral adipose tissue and muscle mass quantified from CT images; studies with other measures, such as waist circumference, are currently lacking. There is only limited evidence that visceral adiposity increased mortality risk. Across study categories, studies had mixed results. Only in the adjuvant setting, two small studies consistently showed increased all-cause mortality risk with higher visceral adipose tissue. Even though quantification of adipose tissue from CT scans is regarded as a more precise measure of adiposity than BMI, the usefulness of single-slice analysis might be limited [88]. On the other hand, evidence consistently shows that low muscle mass is associated with reduced survival, although each study used other cut points to define low muscle mass. The notion that the association between overweight and lower mortality is due solely to methodologic biases is refuted by results from the only large population-based study among non-metastatic CRC patients with available data for both BMI and body composition [39••]. Within the overweight BMI range between 25 and < 30 kg/m2, body composition appeared to explain why a BMI higher than normal is associated with the lowest mortality. The majority (78%) of patients in the overweight group had adequate muscle mass, while less than half (43%) of the patients with a normal BMI had adequate muscle mass. Furthermore, the obesity paradox could also be explained by clinical issues [87], such as metabolic health. One study at Kaiser Permanente investigated the combination of obesity and metabolic health and concluded that mortality risk was statistically significantly increased in obese patients with the metabolic syndrome, but not in metabolically healthy obese patients, compared with metabolically healthy non-obese patients [89].

Conclusions and Future Directions

In conclusion, this review suggests that some, albeit not all, modifiable risk factors for cancer incidence might also be associated with mortality risk after CRC diagnosis. CRC prognosis appears to be worse with increased physical inactivity, smoking, or being underweight after CRC diagnosis. Emerging evidence suggests that diets associated with a positive energy balance, e.g., high consumption of sugar-sweetened beverages, may negatively impact survival in CRC survivors. Nonetheless, data relating post-diagnosis diet to CRC prognosis are scarce; with less than three observational studies that have examined associations for each dietary pattern or individual food after CRC diagnosis. In contrast, high red and processed meat or alcohol intake, established risk factors for incident CRC, do not appear to be associated with mortality after CRC diagnosis. Whether overweight and obesity after CRC diagnosis might confer an additional mortality risk compared to normal weight is still controversial and might depend on how body fatness is assessed and whether muscle mass was accounted for.
Since the first review on lifestyle factors in CRC survivors in 2010 [90], many new studies in this evolving area of research were published and summarized in subsequent reviews and meta-analyses. This is the first paper to comprehensively review post-diagnosis diet, physical activity, smoking, and body composition together in one review. Our findings were generally consistent with previous work, regarding diet [4••], physical activity [710, 11••], smoking [13••], and underweight [16, 17••, 19], although we included new publications. Overweight, assessed by BMI, was consistently associated with lowest mortality risk, although discussion remains about the causal claims regarding the effects of BMI on post-diagnosis mortality for CRC survivors. The only large population-based study among non-metastatic CRC patients concluded that body composition, i.e., muscle mass, appeared to explain why a BMI higher than normal is associated with the lowest mortality risk [39••]. Moreover, low muscle mass was consistently associated with increased mortality risk. Besides observational data, there were no reported randomized controlled trials in smoking or alcohol cessation/reduction, while physical activity and/or dietary/excess weight interventions only reported on short-term outcomes [86••]. Only one small randomized trial assessed long-term follow-up among CRC survivors, finding significantly improved cancer-specific survival after dietary counseling [78].
As people do not have isolated behaviors, a multidimensional lifestyle approach would be most informative for exploring mortality risk and cancer recurrence, as well as for translating these findings into meaningful strategies to improve disease prognosis. Some randomized controlled trials with both dietary and physical activity components have included CRC survivors, but they usually did not test the impact of comprehensive lifestyle interventions on risk of cancer recurrence or survival [86••]. Furthermore, only one observational study evaluated the association of post-diagnosis comprehensive lifestyle patterns and CRC outcomes [91]. That study concluded that adherence to the WCRF recommendations on diet, physical activity, and body fatness was not statistically significantly associated with mortality [91]. However, lifestyle was assessed on average 9 years after diagnosis and survivors were therefore at low risk to die from CRC during subsequent follow-up. Further research on post-diagnosis lifestyle patterns is needed to understand the multifactorial nature of risk of mortality and cancer recurrence and, furthermore, to avoid overemphasis of single lifestyle factors.
The existing studies have several limitations. Few observational studies have reported on the association between post-diagnostic lifestyle and CRC outcomes adjusting for pre-diagnostic lifestyle; thus, it is unknown whether the observed associations between post-diagnostic lifestyle and survival are independent of pre-diagnosis lifestyle. Furthermore, only few studies assessed changes in lifestyle over time in relation to CRC outcomes, with weight change and smoking cessation studied most often. Large prospective cohort studies, such as NHS I, HPFS, the COLON study [92], and others [93, 94], provide further opportunities to examine post-diagnosis lifestyle changes in relation to CRC prognosis during different phases of the cancer trajectory.
Studies evaluating lifestyle factors and CRC outcomes mainly focused on mortality, while cancer recurrence and comorbidities are other important outcomes. Disease recurrence was usually reported by studies in the adjuvant setting, but is not commonly reported by population-based studies. Furthermore, definitions of recurrence were inconsistent between studies. Using the standard definitions proposed by Punt et al. [95] may add to the cross-comparability of future studies. In addition, few studies among CRC survivors studied incidence and progression of comorbidities, although some studies included cardiovascular-mortality as an endpoint. Only one study assessed the incidence of comorbidities after CRC diagnosis [96]. This study observed that BMI and sedentary behavior at 5 months post-diagnosis were associated with the development of comorbid cardiovascular disease in the first 3 years after CRC diagnosis.
More research is needed on the mechanisms underlying the impact of lifestyle after CRC diagnosis on prognosis. A lifestyle contributing to a positive energy balance and hyperinsulinemia has been suggested to be implicated in the prognosis of CRC [5, 97]. For instance, determinants of hyperinsulinemia, such as physical inactivity, excessive sedentary behavior, and several aspects of diet, are associated with increased mortality risk. The dietary factors included in this review that might be linked to insulin-related pathways, a Western dietary pattern [23, 42], sugar-sweetened beverages [23, 43], low coffee consumption [25], and higher dietary glycemic load [45] all showed increased mortality risk. Also, a high-insulinogenic diet [98] has been associated with increased mortality risk. However, these studies were almost all conducted in the same cohort embedded in a trial of adjuvant chemotherapy (CALGB 89803) [25, 42, 43, 45].
Overall, evidence is emerging that modifiable lifestyle factors after CRC diagnosis, such as physical activity, smoking, body composition, and diet could impact survival. Although, not all modifiable risk factors for cancer presentation seem relevant for cancer survivors. With increasing CRC survivorship, however, CRC recurrence should be studied as a key outcome within population-based studies of CRC survivors. Additionally, studies are needed to evaluate the development and progression of comorbidities after CRC diagnosis. Studying lifestyle patterns over time, by including multiple lifestyle factors simultaneously at different time points during the cancer trajectory, would lead to a greater understanding of the multifactorial influence on CRC prognosis. Additional data from prospective observational studies and randomized controlled trials are urgently needed and, ultimately, will allow for lifestyle recommendations that are specifically tailored to cancer survivors.

Compliance with Ethical Standards

Conflict of Interest

Moniek van Zutphen has received research support through a grant from the Dutch Cancer Society. Ellen Kampman declares that she has no conflict of interest. Edward L. Giovannucci declares that he has no conflict of interest. Fränzel J.B. van Duijnhoven declares that she has no conflict of interest.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

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

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

e.Med Innere Medizin

Kombi-Abonnement

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

Literatur
3.
Zurück zum Zitat World Cancer Research Fund/American Institute for Cancer Research. Food, nutrition, physical activity, and the prevention of cancer: a global perspective. Washington DC: AICR; 2007. World Cancer Research Fund/American Institute for Cancer Research. Food, nutrition, physical activity, and the prevention of cancer: a global perspective. Washington DC: AICR; 2007.
4.
Zurück zum Zitat •• Van Blarigan EL, Meyerhardt JA. Role of physical activity and diet after colorectal cancer diagnosis. J Clin Oncol. 2015. https://doi.org/10.1200/jco.2014.59.7799. This review summarizes the evidence regarding physical activity and diet after CRC diagnosis in relation to quality of life, disease recurrence, and survival. •• Van Blarigan EL, Meyerhardt JA. Role of physical activity and diet after colorectal cancer diagnosis. J Clin Oncol. 2015. https://​doi.​org/​10.​1200/​jco.​2014.​59.​7799. This review summarizes the evidence regarding physical activity and diet after CRC diagnosis in relation to quality of life, disease recurrence, and survival.
8.
11.
Zurück zum Zitat •• Friedenreich CM, Neilson HK, Farris MS, Courneya KS. Physical activity and cancer outcomes: a precision medicine approach. Clinical cancer research: an official journal of the American Association for Cancer Research. 2016;22(19):4766–75. https://doi.org/10.1158/1078-0432.ccr-16-0067. This review summarizes the epidemiologic literature relating postdiagnosis physical activity to cancer outcomes overall and by molecular/genetic subgroups. CrossRef •• Friedenreich CM, Neilson HK, Farris MS, Courneya KS. Physical activity and cancer outcomes: a precision medicine approach. Clinical cancer research: an official journal of the American Association for Cancer Research. 2016;22(19):4766–75. https://​doi.​org/​10.​1158/​1078-0432.​ccr-16-0067. This review summarizes the epidemiologic literature relating postdiagnosis physical activity to cancer outcomes overall and by molecular/genetic subgroups. CrossRef
13.
Zurück zum Zitat •• Walter V, Jansen L, Hoffmeister M, Brenner H. Smoking and survival of colorectal cancer patients: systematic review and meta-analysis. Annals of oncology: official journal of the European Society for Medical Oncology / ESMO. 2014;25(8):1517–25. https://doi.org/10.1093/annonc/mdu040. This systematic review and meta-analysis provides a summary of the epidemiologic literature between smoking and survival of CRC patients, including all-cause mortality, CRC-specific mortality, disease-free survival, and recurrence-free survival. CrossRef •• Walter V, Jansen L, Hoffmeister M, Brenner H. Smoking and survival of colorectal cancer patients: systematic review and meta-analysis. Annals of oncology: official journal of the European Society for Medical Oncology / ESMO. 2014;25(8):1517–25. https://​doi.​org/​10.​1093/​annonc/​mdu040. This systematic review and meta-analysis provides a summary of the epidemiologic literature between smoking and survival of CRC patients, including all-cause mortality, CRC-specific mortality, disease-free survival, and recurrence-free survival. CrossRef
15.
Zurück zum Zitat Parkin E, O'Reilly DA, Sherlock DJ, Manoharan P, Renehan AG. Excess adiposity and survival in patients with colorectal cancer: a systematic review. Obesity reviews: an official journal of the International Association for the Study of Obesity. 2014;15(5):434–51. https://doi.org/10.1111/obr.12140.CrossRef Parkin E, O'Reilly DA, Sherlock DJ, Manoharan P, Renehan AG. Excess adiposity and survival in patients with colorectal cancer: a systematic review. Obesity reviews: an official journal of the International Association for the Study of Obesity. 2014;15(5):434–51. https://​doi.​org/​10.​1111/​obr.​12140.CrossRef
17.
Zurück zum Zitat •• Lee J, Meyerhardt JA, Giovannucci E, Jeon JY. Association between body mass index and prognosis of colorectal cancer: a meta-analysis of prospective cohort studies. PLoS One. 2015;10(3):e0120706. https://doi.org/10.1371/journal.pone.0120706. This meta-analysis of prospective studies summarized the association of pre- and post-diagnostic BMI with CRC-specific mortality and all-cause mortality in patients with CRC. PubMedPubMedCentralCrossRef •• Lee J, Meyerhardt JA, Giovannucci E, Jeon JY. Association between body mass index and prognosis of colorectal cancer: a meta-analysis of prospective cohort studies. PLoS One. 2015;10(3):e0120706. https://​doi.​org/​10.​1371/​journal.​pone.​0120706. This meta-analysis of prospective studies summarized the association of pre- and post-diagnostic BMI with CRC-specific mortality and all-cause mortality in patients with CRC. PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Malietzis G, Aziz O, Bagnall NM, Johns N, Fearon KC, Jenkins JT. The role of body composition evaluation by computerized tomography in determining colorectal cancer treatment outcomes: a systematic review. European journal of surgical oncology : the journal of the European Society of Surgical Oncology and the British Association of Surgical Oncology. 2015;41(2):186–96. https://doi.org/10.1016/j.ejso.2014.10.056.CrossRef Malietzis G, Aziz O, Bagnall NM, Johns N, Fearon KC, Jenkins JT. The role of body composition evaluation by computerized tomography in determining colorectal cancer treatment outcomes: a systematic review. European journal of surgical oncology : the journal of the European Society of Surgical Oncology and the British Association of Surgical Oncology. 2015;41(2):186–96. https://​doi.​org/​10.​1016/​j.​ejso.​2014.​10.​056.CrossRef
24.
28.
Zurück zum Zitat Tao L, Wang R, Gao YT, Yuan JM. Impact of postdiagnosis smoking on long-term survival of cancer patients: the Shanghai Cohort Study. Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2013;22(12):2404–11. https://doi.org/10.1158/1055-9965.epi-13-0805-t.CrossRef Tao L, Wang R, Gao YT, Yuan JM. Impact of postdiagnosis smoking on long-term survival of cancer patients: the Shanghai Cohort Study. Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2013;22(12):2404–11. https://​doi.​org/​10.​1158/​1055-9965.​epi-13-0805-t.CrossRef
29.
Zurück zum Zitat Yang B, Jacobs EJ, Gapstur SM, Stevens V, Campbell PT. Active smoking and mortality among colorectal cancer survivors: the Cancer Prevention Study II nutrition cohort. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2015;33(8):885–93. https://doi.org/10.1200/jco.2014.58.3831.CrossRef Yang B, Jacobs EJ, Gapstur SM, Stevens V, Campbell PT. Active smoking and mortality among colorectal cancer survivors: the Cancer Prevention Study II nutrition cohort. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2015;33(8):885–93. https://​doi.​org/​10.​1200/​jco.​2014.​58.​3831.CrossRef
30.
Zurück zum Zitat Sharp L, McDevitt J, Brown C, Carsin AE, Comber H. Association between smoking at diagnosis and cause-specific survival in patients with rectal cancer: results from a population-based analysis of 10,794 cases. Cancer. 2017. https://doi.org/10.1002/cncr.30583. Sharp L, McDevitt J, Brown C, Carsin AE, Comber H. Association between smoking at diagnosis and cause-specific survival in patients with rectal cancer: results from a population-based analysis of 10,794 cases. Cancer. 2017. https://​doi.​org/​10.​1002/​cncr.​30583.
35.
Zurück zum Zitat Black D, Mackay C, Ramsay G, Hamoodi Z, Nanthakumaran S, Park KGM, et al. Prognostic value of computed tomography: measured parameters of body composition in primary operable gastrointestinal cancers. Ann Surg Oncol. 2017:1–11. https://doi.org/10.1245/s10434-017-5829-z. Black D, Mackay C, Ramsay G, Hamoodi Z, Nanthakumaran S, Park KGM, et al. Prognostic value of computed tomography: measured parameters of body composition in primary operable gastrointestinal cancers. Ann Surg Oncol. 2017:1–11. https://​doi.​org/​10.​1245/​s10434-017-5829-z.
36.
Zurück zum Zitat Blauwhoff-Buskermolen S, Versteeg KS, de van der Schueren MA, den Braver NR, Berkhof J, Langius JA, et al. Loss of muscle mass during chemotherapy is predictive for poor survival of patients with metastatic colorectal cancer. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2016;34(12):1339–44. https://doi.org/10.1200/JCO.2015.63.6043.CrossRef Blauwhoff-Buskermolen S, Versteeg KS, de van der Schueren MA, den Braver NR, Berkhof J, Langius JA, et al. Loss of muscle mass during chemotherapy is predictive for poor survival of patients with metastatic colorectal cancer. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2016;34(12):1339–44. https://​doi.​org/​10.​1200/​JCO.​2015.​63.​6043.CrossRef
37.
Zurück zum Zitat Jung HW, Kim JW, Kim JY, Kim SW, Yang HK, Lee JW, et al. Effect of muscle mass on toxicity and survival in patients with colon cancer undergoing adjuvant chemotherapy. Supportive care in cancer: official journal of the Multinational Association of Supportive Care in Cancer. 2015;23(3):687–94. https://doi.org/10.1007/s00520-014-2418-6.CrossRef Jung HW, Kim JW, Kim JY, Kim SW, Yang HK, Lee JW, et al. Effect of muscle mass on toxicity and survival in patients with colon cancer undergoing adjuvant chemotherapy. Supportive care in cancer: official journal of the Multinational Association of Supportive Care in Cancer. 2015;23(3):687–94. https://​doi.​org/​10.​1007/​s00520-014-2418-6.CrossRef
39.
Zurück zum Zitat •• Caan BJ, Meyerhardt JA, Kroenke CH, Alexeeff S, Xiao J, Weltzien E, et al. Explaining the obesity paradox: the association between body composition and colorectal cancer survival (C-SCANS study). Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2017. https://doi.org/10.1158/1055-9965.EPI-17-0200. This study among non-metastatic CRC patients concluded that body composition appeared to explain why a BMI higher than normal is associated with the lowest mortality risk and therefore they refute the notion that the association between overweight and lower mortality is solely due to methodologic biases. •• Caan BJ, Meyerhardt JA, Kroenke CH, Alexeeff S, Xiao J, Weltzien E, et al. Explaining the obesity paradox: the association between body composition and colorectal cancer survival (C-SCANS study). Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2017. https://​doi.​org/​10.​1158/​1055-9965.​EPI-17-0200. This study among non-metastatic CRC patients concluded that body composition appeared to explain why a BMI higher than normal is associated with the lowest mortality risk and therefore they refute the notion that the association between overweight and lower mortality is solely due to methodologic biases.
40.
41.
Zurück zum Zitat Yang B, McCullough ML, Gapstur SM, Jacobs EJ, Bostick RM, Fedirko V, et al. Calcium, vitamin D, dairy products, and mortality among colorectal cancer survivors: the Cancer Prevention Study-II Nutrition Cohort. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2014;32(22):2335–43. https://doi.org/10.1200/JCO.2014.55.3024.CrossRef Yang B, McCullough ML, Gapstur SM, Jacobs EJ, Bostick RM, Fedirko V, et al. Calcium, vitamin D, dairy products, and mortality among colorectal cancer survivors: the Cancer Prevention Study-II Nutrition Cohort. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2014;32(22):2335–43. https://​doi.​org/​10.​1200/​JCO.​2014.​55.​3024.CrossRef
48.
Zurück zum Zitat Baade PD, Meng X, Youl PH, Aitken JF, Dunn J, Chambers SK. The impact of body mass index and physical activity on mortality among patients with colorectal cancer in Queensland, Australia. Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2011;20(7):1410–20. https://doi.org/10.1158/1055-9965.EPI-11-0079.CrossRef Baade PD, Meng X, Youl PH, Aitken JF, Dunn J, Chambers SK. The impact of body mass index and physical activity on mortality among patients with colorectal cancer in Queensland, Australia. Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2011;20(7):1410–20. https://​doi.​org/​10.​1158/​1055-9965.​EPI-11-0079.CrossRef
51.
Zurück zum Zitat Arem H, Pfeiffer RM, Engels EA, Alfano CM, Hollenbeck A, Park Y, 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. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2015;33(2):180–8. https://doi.org/10.1200/JCO.2014.58.1355.CrossRef Arem H, Pfeiffer RM, Engels EA, Alfano CM, Hollenbeck A, Park Y, 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. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2015;33(2):180–8. https://​doi.​org/​10.​1200/​JCO.​2014.​58.​1355.CrossRef
52.
Zurück zum Zitat Meyerhardt JA, Heseltine D, Niedzwiecki D, Hollis D, Saltz LB, Mayer RJ, et al. Impact of physical activity on cancer recurrence and survival in patients with stage III colon cancer: findings from CALGB 89803. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2006;24(22):3535–41. https://doi.org/10.1200/JCO.2006.06.0863.CrossRef Meyerhardt JA, Heseltine D, Niedzwiecki D, Hollis D, Saltz LB, Mayer RJ, et al. Impact of physical activity on cancer recurrence and survival in patients with stage III colon cancer: findings from CALGB 89803. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2006;24(22):3535–41. https://​doi.​org/​10.​1200/​JCO.​2006.​06.​0863.CrossRef
54.
Zurück zum Zitat Jadallah F, McCall JL, van Rij AM. Recurrence and survival after potentially curative surgery for colorectal cancer. The New Zealand medical journal. 1999;112(1091):248–50.PubMed Jadallah F, McCall JL, van Rij AM. Recurrence and survival after potentially curative surgery for colorectal cancer. The New Zealand medical journal. 1999;112(1091):248–50.PubMed
55.
Zurück zum Zitat Ali RA, Dooley C, Comber H, Newell J, Egan LJ. Clinical features, treatment, and survival of patients with colorectal cancer with or without inflammatory bowel disease. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association. 2011;9(7):584-9.e1–2. https://doi.org/10.1016/j.cgh.2011.04.016. Ali RA, Dooley C, Comber H, Newell J, Egan LJ. Clinical features, treatment, and survival of patients with colorectal cancer with or without inflammatory bowel disease. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association. 2011;9(7):584-9.e1–2. https://​doi.​org/​10.​1016/​j.​cgh.​2011.​04.​016.
58.
59.
Zurück zum Zitat Munro AJ, Bentley AH, Ackland C, Boyle PJ. Smoking compromises cause-specific survival in patients with operable colorectal cancer. Clin Oncol (R Coll Radiol). 2006;18(6):436–40.CrossRef Munro AJ, Bentley AH, Ackland C, Boyle PJ. Smoking compromises cause-specific survival in patients with operable colorectal cancer. Clin Oncol (R Coll Radiol). 2006;18(6):436–40.CrossRef
62.
Zurück zum Zitat Asghari-Jafarabadi M, Hajizadeh E, Kazemnejad A, Fatemi SR. Site-specific evaluation of prognostic factors on survival in Iranian colorectal cancer patients: a competing risks survival analysis. Asian Pacific journal of cancer prevention : APJCP. 2009;10(5):815–21.PubMed Asghari-Jafarabadi M, Hajizadeh E, Kazemnejad A, Fatemi SR. Site-specific evaluation of prognostic factors on survival in Iranian colorectal cancer patients: a competing risks survival analysis. Asian Pacific journal of cancer prevention : APJCP. 2009;10(5):815–21.PubMed
64.
Zurück zum Zitat Campbell PT, Newton CC, Dehal AN, Jacobs EJ, Patel AV, Gapstur SM. Impact of body mass index on survival after colorectal cancer diagnosis: the cancer prevention study-II nutrition cohort. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2012;30(1):42–52. https://doi.org/10.1200/JCO.2011.38.0287.CrossRef Campbell PT, Newton CC, Dehal AN, Jacobs EJ, Patel AV, Gapstur SM. Impact of body mass index on survival after colorectal cancer diagnosis: the cancer prevention study-II nutrition cohort. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2012;30(1):42–52. https://​doi.​org/​10.​1200/​JCO.​2011.​38.​0287.CrossRef
68.
Zurück zum Zitat Meyerhardt JA, Tepper JE, Niedzwiecki D, Hollis DR, McCollum AD, Brady D, et al. Impact of body mass index on outcomes and treatment-related toxicity in patients with stage II and III rectal cancer: findings from intergroup trial 0114. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2004;22(4):648–57. https://doi.org/10.1200/JCO.2004.07.121.CrossRef Meyerhardt JA, Tepper JE, Niedzwiecki D, Hollis DR, McCollum AD, Brady D, et al. Impact of body mass index on outcomes and treatment-related toxicity in patients with stage II and III rectal cancer: findings from intergroup trial 0114. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2004;22(4):648–57. https://​doi.​org/​10.​1200/​JCO.​2004.​07.​121.CrossRef
71.
Zurück zum Zitat Rickles AS, Iannuzzi JC, Mironov O, Deeb AP, Sharma A, Fleming FJ, et al. Visceral obesity and colorectal cancer: are we missing the boat with BMI? Journal of gastrointestinal surgery: official journal of the Society for Surgery of the Alimentary Tract. 2013;17(1):133–143; discussion p 43. https://doi.org/10.1007/s11605-012-2045-9.CrossRef Rickles AS, Iannuzzi JC, Mironov O, Deeb AP, Sharma A, Fleming FJ, et al. Visceral obesity and colorectal cancer: are we missing the boat with BMI? Journal of gastrointestinal surgery: official journal of the Society for Surgery of the Alimentary Tract. 2013;17(1):133–143; discussion p 43. https://​doi.​org/​10.​1007/​s11605-012-2045-9.CrossRef
80.
Zurück zum Zitat Meyerhardt JA, Kroenke CH, Prado CM, Kwan ML, Castillo A, Weltzien E, et al. Association of Weight Change after colorectal cancer diagnosis and outcomes in the Kaiser Permanente northern California population. Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2016;26(1):30–7. https://doi.org/10.1158/1055-9965.EPI-16-0145.CrossRef Meyerhardt JA, Kroenke CH, Prado CM, Kwan ML, Castillo A, Weltzien E, et al. Association of Weight Change after colorectal cancer diagnosis and outcomes in the Kaiser Permanente northern California population. Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2016;26(1):30–7. https://​doi.​org/​10.​1158/​1055-9965.​EPI-16-0145.CrossRef
81.
Zurück zum Zitat Meyerhardt JA, Niedzwiecki D, Hollis D, Saltz LB, Mayer RJ, Nelson H, 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. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2008;26(25):4109–15. https://doi.org/10.1200/JCO.2007.15.6687.CrossRef Meyerhardt JA, Niedzwiecki D, Hollis D, Saltz LB, Mayer RJ, Nelson H, 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. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2008;26(25):4109–15. https://​doi.​org/​10.​1200/​JCO.​2007.​15.​6687.CrossRef
83.
Zurück zum Zitat Courneya KS, Booth CM, Gill S, O'Brien P, Vardy J, Friedenreich CM, 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(6):279–85.PubMedCrossRef Courneya KS, Booth CM, Gill S, O'Brien P, Vardy J, Friedenreich CM, 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(6):279–85.PubMedCrossRef
85.
Zurück zum Zitat Courneya KS, Vardy JL, O'Callaghan CJ, Friedenreich CM, Campbell KL, Prapavessis H, et al. Effects of a structured exercise program on physical activity and fitness in colon cancer survivors: one year feasibility results from the CHALLENGE trial. Cancer Epidemiol Biomark Prev. 2016;25(6):969–77. https://doi.org/10.1158/1055-9965.epi-15-1267.CrossRef Courneya KS, Vardy JL, O'Callaghan CJ, Friedenreich CM, Campbell KL, Prapavessis H, et al. Effects of a structured exercise program on physical activity and fitness in colon cancer survivors: one year feasibility results from the CHALLENGE trial. Cancer Epidemiol Biomark Prev. 2016;25(6):969–77. https://​doi.​org/​10.​1158/​1055-9965.​epi-15-1267.CrossRef
86.
Zurück zum Zitat •• Moug SJ, Bryce A, Mutrie N, Anderson AS. Lifestyle interventions are feasible in patients with colorectal cancer with potential short-term health benefits: a systematic review. Int J Color Dis. 2017;32(6):765–75. https://doi.org/10.1007/s00384-017-2797-5. This systematic review assessed the evidence for the feasibility of performing lifestyle interventions in CRC patients and evaluated any short- and long-term health benefits. CrossRef •• Moug SJ, Bryce A, Mutrie N, Anderson AS. Lifestyle interventions are feasible in patients with colorectal cancer with potential short-term health benefits: a systematic review. Int J Color Dis. 2017;32(6):765–75. https://​doi.​org/​10.​1007/​s00384-017-2797-5. This systematic review assessed the evidence for the feasibility of performing lifestyle interventions in CRC patients and evaluated any short- and long-term health benefits. CrossRef
91.
Zurück zum Zitat Inoue-Choi M, Robien K, Lazovich D. Adherence to the WCRF/AICR guidelines for cancer prevention is associated with lower mortality among older female cancer survivors. Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2013;22(5):792–802. https://doi.org/10.1158/1055-9965.EPI-13-0054.CrossRef Inoue-Choi M, Robien K, Lazovich D. Adherence to the WCRF/AICR guidelines for cancer prevention is associated with lower mortality among older female cancer survivors. Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2013;22(5):792–802. https://​doi.​org/​10.​1158/​1055-9965.​EPI-13-0054.CrossRef
93.
Zurück zum Zitat Soares-Miranda L, Abreu S, Silva M, Peixoto A, Ramalho R, da Silva PC, et al. Cancer survivor study (CASUS) on colorectal patients: longitudinal study on physical activity, fitness, nutrition, and its influences on quality of life, disease recurrence, and survival. Rationale and design. Int J Color Dis. 2016. https://doi.org/10.1007/s00384-016-2671-x. Soares-Miranda L, Abreu S, Silva M, Peixoto A, Ramalho R, da Silva PC, et al. Cancer survivor study (CASUS) on colorectal patients: longitudinal study on physical activity, fitness, nutrition, and its influences on quality of life, disease recurrence, and survival. Rationale and design. Int J Color Dis. 2016. https://​doi.​org/​10.​1007/​s00384-016-2671-x.
94.
Zurück zum Zitat van Roekel EH, Bours MJL, de Brouwer CPM, Ten Napel H, Sanduleanu S, Beets GL, et al. The applicability of the international classification of functioning, disability, and health to study lifestyle and quality of life of colorectal cancer survivors. Cancer Epidemiol Biomark Prev. 2014;23(7):1394–405. https://doi.org/10.1158/1055-9965.epi-13-1144.CrossRef van Roekel EH, Bours MJL, de Brouwer CPM, Ten Napel H, Sanduleanu S, Beets GL, et al. The applicability of the international classification of functioning, disability, and health to study lifestyle and quality of life of colorectal cancer survivors. Cancer Epidemiol Biomark Prev. 2014;23(7):1394–405. https://​doi.​org/​10.​1158/​1055-9965.​epi-13-1144.CrossRef
95.
Metadaten
Titel
Lifestyle after Colorectal Cancer Diagnosis in Relation to Survival and Recurrence: A Review of the Literature
verfasst von
Moniek van Zutphen
Ellen Kampman
Edward L. Giovannucci
Fränzel J. B. van Duijnhoven
Publikationsdatum
14.09.2017
Verlag
Springer US
Erschienen in
Current Colorectal Cancer Reports / Ausgabe 5/2017
Print ISSN: 1556-3790
Elektronische ISSN: 1556-3804
DOI
https://doi.org/10.1007/s11888-017-0386-1

Update Onkologie

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