Purpose:

Lynch syndrome (LS) is a hereditary condition with a high lifetime risk of colorectal and endometrial cancers. Exercise is a non-pharmacologic intervention to reduce cancer risk, though its impact on patients with LS has not been prospectively studied. Here, we evaluated the impact of a 12-month aerobic exercise cycling intervention in the biology of the immune system in LS carriers.

Patients and Methods:

To address this, we enrolled 21 patients with LS onto a non-randomized, sequential intervention assignation, clinical trial to assess the effect of a 12-month exercise program that included cycling classes 3 times weekly for 45 minutes versus usual care with a one-time exercise counseling session as control. We analyzed the effects of exercise on cardiorespiratory fitness, circulating, and colorectal-tissue biomarkers using metabolomics, gene expression by bulk mRNA sequencing, and spatial transcriptomics by NanoString GeoMx.

Results:

We observed a significant increase in oxygen consumption (VO2peak) as a primary outcome of the exercise and a decrease in inflammatory markers (prostaglandin E) in colon and blood as the secondary outcomes in the exercise versus usual care group. Gene expression profiling and spatial transcriptomics on available colon biopsies revealed an increase in the colonic mucosa levels of natural killer and CD8+ T cells in the exercise group that were further confirmed by IHC studies.

Conclusions:

Together these data have important implications for cancer interception in LS, and document for the first-time biological effects of exercise in the immune system of a target organ in patients at-risk for cancer.

This article is featured in Selected Articles from This Issue, p. 4315

Translational Relevance

Our study highlights the important role of exercise in modulating inflammation and activity of the resident immune system in the colonic mucosa of patients with Lynch syndrome (LS). Cycling exercise promoted the reduction of inflammatory prostaglandin E2 levels in circulation and the colon as well as reduction of circulating triacyl-glycerides in LS carriers assigned to the exercise intervention and generated an enrichment of natural killer and CD8+ T cell levels when compared with those LS carriers in the control group. These findings suggest that exercise has an important role fostering both the innate and adaptive immune response in a high-risk cancer population. We confirmed that regular aerobic exercise in patients with LS is a safe recommendation for general health benefits and is potentially cancer preventive. A randomized study in LS population is warranted to further explore the preventive efficacy of aerobic exercise training.

Lynch syndrome (LS) is the most common hereditary cancer syndrome, potentially affecting 1.1 million individuals in the United States (1). LS carriers have up to 60% lifetime risk of colorectal cancer (2). Exercise is one potential non-pharmacologic strategy to mitigate excessive colorectal cancer risk in patients with LS. In fact, exercise is associated with reduction in colorectal cancer among the general population (3) with evidence to suggest a robust risk reduction, increased functional capacity and quality of life among individuals who sustain moderate to high levels of activity over time compared with those remaining inactive (4, 5). A similar beneficial association between exercise and colorectal cancer risk has been suggested in patients with LS in retrospective studies showing that physical activity of ≥35 MET-h/week decreased the risk of colorectal cancer (6, 7). However, despite the biological plausibility between exercise and reduction in colorectal cancer risk being robust, the mechanism underlying this association remains unclear with studies showing modulatory effects on the gut microbiome (8, 9), insulin growth factor signaling, and other essential pathways in colorectal cancer development to promote antitumorigenic activity (10, 11).

Overall, exercise alters inflammatory, immune, and insulin growth factor pathways implicated in colorectal cancer development (12–15). In particular, exercise removes excess energy substrate availability, which drives down the formation of reactive oxygen species and subsequent signaling pathways that affect cell-cycle control and survival (16–18). Exercise is also hypothesized to impact prostaglandin (PG)-mediated inflammation, an essential pathway in the development of colorectal cancer (19). We and others have shown that cancer interception strategies such as aspirin and naproxen can successfully inhibit PG synthesis via blockage of cyclooxygenase (COX) enzymes to prevent colon tumorigenesis in LS (20, 21). To date, however, exercise studies on PG synthesis have rendered mixed results at the tissue level. One previous study demonstrated an inverse correlation between self-reported exercise and prostaglandin E2 (PGE2) levels in rectal tissue in patients with a history of polyps (19). In contrast, a randomized clinical trial of healthy adults failed to show an association between exercise training with a change in colonic PGE2 levels (22). Therefore, it remains unclear whether exercise modulates PG levels via COX enzymes and/or other potential pathways implicated in colorectal cancer development through induction of transcriptomic and metabolomic changes. More specifically, the role of exercise to biologically modulate mismatch repair (MMR)-deficient carcinogenesis in the intestinal epithelium to prevent colorectal cancer in LS carriers is unknown.

As such, we sought to (1) directly test the hypothesis that exercise using a sustained aerobic cycling intervention alters gene expression patterns and circulating factors in the colorectal mucosa and circulating PG levels over a 12-month period, and (2) to assess the feasibility of such an intervention in LS carriers as measured by recruitment, retention, and adherence rates. Here, we report the results of ‘the CYCLE-P study’ that enrolled LS participants, who were in an active cancer surveillance program at The University of Texas MD Anderson Cancer Center (MDACC) into a 12-month aerobic exercise intervention or usual care.

Study design and participants

CYCLE-P was a prospective, non-randomized, sequential assignation to intervention, single-site controlled trial that enrolled 21 participants diagnosed with LS in a 12-month aerobic exercise intervention involving cycling classes in a local community (n = 11) followed by enrollment to usual care (n = 10, control group) between April 2018 to January 2019 (Supplementary Fig. S1). Eligible participants were approached prior to their scheduled colonoscopy and all of them provided written informed consent before enrollment. The conduct of the trial complied with the Declaration of Helsinki. Participants were told their allocation when approached for the study and each group signed a different informed consent. The trial protocol and all amendments were approved by the MDACC institutional review board. We followed the Transparent Reporting of Evaluations with Nonrandomized Designs reporting guidelines (23). All authors had access to the study data and reviewed and approved the final manuscript.

Participants were adults between 18 and 50 years of age with LS defined as carriers or obligate carriers by the pedigree of a pathogenic mutation in one of the four DNA MMR genes (i.e., MLH1, MSH2/EPCAM, MSH6, or PMS2) or as participants with a personal history of a non-sporadic MMR-deficient neoplasia defined by IHC or microsatellite instability testing or both. Participants did not have evidence of active or recurrent malignant disease or cancer-directed treatment in the previous 6 months. Only participants willing to undergo yearly screening colonoscopy were eligible. Participants were required to retain a portion of the distal colon or rectosigmoid intact to enable collection of normal mucosa biopsies. Exclusion criteria included previous total proctocolectomy, history of cardiovascular disease, or uncontrolled medical conditions. A lower age cut-off of 18 years was prespecified given standard of care surveillance endoscopy is not performed routinely in patients with LS under 18 years of age. An upper age limit of 50 years was prespecified to recruit healthy patients with LS without co-morbidities who were more likely to achieve the prescribed exercise volume and intensity [≥70% peak heart rate (PHR)] over 52 weeks.

Study interventions

Figure 1 shows the flow and timeline of clinical assessments, interventions, and follow-up testing. Both the intervention group and the usual care group underwent standard of care lower gastrointestinal (GI) endoscopy with biopsies and blood collection at visit 1. Cardiopulmonary exercise testing (CPET) was performed at visit 2 within 30 days of endoscopy. Visit 3 was performed at the 1-year endoscopy for both the intervention and usual care groups, followed by a CPET at visit 4 within 30 days of the 1-year endoscopy. A baseline health questionnaire was used to assess the regular use of aspirin (defined as ≥ 3 times per week) with no participants taking aspirin. Intermittent use of NSAIDs was not recorded throughout the trial.

Figure 1.

A, Trial schema. Patients with LS were consecutively assigned to a 12-month aerobic exercise intervention involving cycling (n = 11) followed by enrollment of patients to usual care (n = 10). Primary endpoints of the trial were feasibility, evaluated by recruitment in terms of eligibility and consent, adherence, and retention rates; changes in the peak oxygen consumption (VO2 peak); and changes in the PG levels in colorectal mucosa and serum. Additional secondary endpoints included gene expression profiling and metabolomics. Blood, urine, and colorectal mucosa biopsies were collected at baseline and 12-months. The numbers in the box represent the number of participants on whom samples/VO2 levels were collected per group at each time point; B, Total weekly minutes of exercise at heart rate≥70% of PHR represented in bars. Blue bars represent the usual care and red bars represent the exercise group; C, Changes in peak oxygen consumption (VO2 peak) before and after intervention in the usual care (blue) and exercise (red) groups; D, Relative change in PGE2 after twelve months follow-up in the usual care control and exercise groups (Wilcoxon signed-rank test; *, P ≤ 0.05); E, Exercise group. UC, Usual Care group.

Figure 1.

A, Trial schema. Patients with LS were consecutively assigned to a 12-month aerobic exercise intervention involving cycling (n = 11) followed by enrollment of patients to usual care (n = 10). Primary endpoints of the trial were feasibility, evaluated by recruitment in terms of eligibility and consent, adherence, and retention rates; changes in the peak oxygen consumption (VO2 peak); and changes in the PG levels in colorectal mucosa and serum. Additional secondary endpoints included gene expression profiling and metabolomics. Blood, urine, and colorectal mucosa biopsies were collected at baseline and 12-months. The numbers in the box represent the number of participants on whom samples/VO2 levels were collected per group at each time point; B, Total weekly minutes of exercise at heart rate≥70% of PHR represented in bars. Blue bars represent the usual care and red bars represent the exercise group; C, Changes in peak oxygen consumption (VO2 peak) before and after intervention in the usual care (blue) and exercise (red) groups; D, Relative change in PGE2 after twelve months follow-up in the usual care control and exercise groups (Wilcoxon signed-rank test; *, P ≤ 0.05); E, Exercise group. UC, Usual Care group.

Close modal

Cycling intervention group

Participants were asked to enroll in subsidized cycling classes for 45 minutes 3 times weekly for 12 months through stand-alone businesses, structured gyms (i.e., YMCA), and one participant through a Peloton bike (Supplementary Fig. S1–S2). There was no ramp-up period for physical activity during the initial weeks. However, a 2-week run-in period was allowed to identify and schedule cycling classes, and during this time participants were contacted by an exercise physiologist to discuss barriers or difficulties finding exercise classes. Afterwards, participants were contacted every two weeks to track and encourage compliance throughout the study. Exercise data including exercise minutes, sleep time, and training heart rate was tracked on an electronic wearable device (Fitbit) by retrieving the data from FITABASE over the study period, and intensity was determined by taking a percentage of an individual's PHR determined during the visit 2 CPET. We have not specifically monitored if any group has been involved in other physical activity outside the study intervention. Exercise minutes ≥70% of an individual's PHR continuously for ≥10 minutes were considered valid and counted. In addition, we tracked the average weekly wear of the Fitbit, and assessed total minutes of exercise at 70% to 85% heart peak rate, and at >85% heart peak rate. A bimonthly response to an exercise questionnaire on class attendance and weekly surveillance of valid exercise minutes and electronic wearable device compliance was overseen by an exercise physiologist.

Usual care group and incentives

Participants in the usual care group did not enroll in cycling classes but were counseled on current exercise guideline recommendations by an exercise physiologist at study visit 2. This group was monitored for exercise via electronic wearable devices under the same principles as the intervention group. Incentives were provided to both groups. Participants in the cycling intervention attending cycling classes received $50 at the end of each month for a total of 12 months to cover the cost of the classes. Both groups were given a $100 payment for completing visit 2 and $300 for completing visit 4.

Outcomes

Peak oxygen consumption (VO2peak) was assessed by a symptom-limited CPET on a bicycle with a 12-lead electrocardiogram using the Cosmed Quark metabolic testing system with a standardized protocol for an exercise laboratory, as previously described (24). Feasibility was evaluated by recruitment in terms of eligibility and consent, adherence, and retention rates. The recruitment rate was defined as the proportion of eligible patients who consented out of the total number of patients contacted by the study team. A participant was deemed adherent in a given week if they performed all 3 cycle sessions that week. Adherence was assessed through self-reported email questionnaires. The adherence rate was then calculated as the percent of weeks (of 52) that a participant was adherent. Retention was calculated as the percent of participants in each group who completed CPET at visit 4. This study was considered feasible per protocol if the recruitment rate was at least 20%, along with at least 75% rates for both adherence and retention.

Colonoscopy methods

Standard of care lower GI endoscopy (flexible sigmoidoscopy or colonoscopy depending on the length of the participants’ colon) was performed in all participants. Biopsies were taken from grossly normal rectosigmoid mucosa using jumbo biopsy forceps. Tissue specimens were then processed immediately in formaldehyde for histology, RNAlater for nucleic acid extraction, and flash-frozen in liquid nitrogen for proteomic analysis and storage.

Transcriptomic and metabolomic analysis

Detailed information on mRNA sequencing (mRNA-seq) and bioinformatics analysis, measurement of PGs, metabolomic studies, GeoMx Digital Spatial Profiling (DSP) and IHC are provided in Supplementary Methods.

Statistical analysis

Descriptive statistics were used to summarize demographics, health history, and study assessments. Categorical variables were summarized with counts and percentages, and Fisher exact test was used to test for associations between these characteristics and the study group. Continuous variables were summarized with median and interquartile range (IQR) and compared between groups using the Wilcoxon rank-sum test. A one-sample paired t test determined significance of the mean percent change in relative VO2peak from baseline to 12 months being different from 0 with a 95% confidence interval (CI) for each study group. Statistical significance was determined using a significance level of 0.05. All statistical analyses were performed using SAS 9.4 for Windows (SAS Institute Inc., Cary, NC) and RStudio 1.1.463 (© 2009–2018 RStudio, Inc., Boston, MA). Statistical parameters for the metabolomic and mRNA-seq studies and the mixed-effect model are included in the Supplementary Methods.

Data availability

The data generated in this study can be accessed at GSE183150 for mRNA data and at GSE210011 for DSP data. To uphold patient privacy and consent, individual participant data will not be shared.

Sixty potential participants were approached and invited to enroll in the study during their regular cancer screening visits at MDACC. Twenty-one participants signed informed consent, enrolled in the study, and were allocated to the exercise group (n = 11) or usual care group (n = 10; Fig. 1A). Participant's ages differed between the exercise group (46 years, IQR 40- 48) and a usual care group (35.5 years, IQR 34–38). Otherwise, baseline characteristics and CPET results between groups were similar in terms of sex, body mass index (BMI), and baseline VO2peak (Table 1). No participants had a history of diabetes, hypertension, or were taking NSAIDs such as aspirin or steroids. One patient in the usual care group was receiving treatment with a statin.

Table 1.

Clinical characteristics of trial participants for CYCLE-P study at MDACC from April 2018 to January 2019.

Group
ExerciseControlTotal
(N = 11)(N = 10)(N = 21)P value
Age 46 (40–48) 35.5 (34–38) 39 (34–47) 0.063a 
Sex, n (%)    >0.99b 
 Male 3 (27%) 3 (30%) 6 (29%)  
 Female 8 (73%) 7 (70%) 15 (71%)  
Ethnicity, n (%)    0.476b 
 Hispanic 0 (0%) 1 (10%) 1 (5%)  
 Non-Hispanic 11 (100%) 9 (90%) 20 (95%)  
Race, n (%)    >0.99b 
 White 10 (90%) 10 (100%) 20 (95%)  
 Asian 1 (10%) 0 (0%) 1 (5%)  
MMR gene mutated, n (%)     
MLH1 4 (36%) 5 (50%) 9 (42.9%)  
MSH2 5 (45%) 1 (10%) 6 (28.6%)  
MSH6 1 (9.1%) 4 (40%) 5 (23.8%)  
PMS2 1 (9.0%) 0 (0%) 1 (4.7%)  
BMI – B (kg/m227.4 (23.5–31.1) 27.4 (26.1–28.6) 27.4 (23.6–30.2) 0.734a 
BMI – EOS (kg/m225.3 (24.4–26.8) 28.1 (27.2–30.8) 26.9 (24.6–30.1) 0.168a 
Smoking History n (%)    >0.99b 
 Former smoker 2 (18%) 1 (10%) 3 (14%)  
 Never smoker 9 (82%) 9 (90%) 18 (86%)  
Hypertension (Hx + Meds), n (%)    >0.99b 
 No 11 (100%) 10 (0%) 21 (100%)  
 Yes 0 (0%) 0 (0%) 0 (0%)  
Diabetes, n (%)    >0.99b 
 No 11 (100%) 10 (0%) 21 (100%)  
 Yes 0 (0%) 0 (0%) 0 (0%)  
High Cholesterol (Hx + Meds), n (%)    0.476b 
 No 11 (100%) 9 (90%) 20 (95%)  
 Yes 0 (0%) 1 (10%) 1 (5%)  
History of cancer, n (%)     
 No 7 (64%) 7 (70%) 14 (66.7%) 1.0b 
 Yes 4 (36%) 3 (30%) 7 (33.3%)  
Type of Cancer, n (%)     
 Colorectal 2 (50%) 3 (100%) 5 (71%)  
 Thyroid 2 (50%) 2 (29%)  
Self-reported physical activity score 56 (20–83) 28.5 (23–49) 38 (23–62) 0.218a 
Weekly Fitbit wearing time (days) 6.1 (5.7–6.9) 5.7 (5.4–6.1) 6.0 (5.5–6.7) 0.280a 
Total time at 70–85 Peak Hear Rate (min) 6203 (4270–8338) 1076 (506–2727) 3744 (1092–5949) 0.0002a 
Total time at >85 Peak Hear Rate (min) 1668 (1317–1938) 156 (55–532) 851 (173–1659) 0.001a 
Exercise Self-Efficacy Score – B 61.5 (38–82) 64 (56–77) 64 (44.5–79) 0.734a 
Exercise Self-Efficacy Score – EOS 58 (38–77) 48.5 (46.5–52.8) 54 (44–61) 0.409a 
Resting Heart Rate – B (bpm) 71 (62–83) 86 (77–96) 79 (67–93) 0.152a 
Resting Heart Rate – EOS 68 (66–79) 78 (74–86) 77 (66–81) 0.258a 
Average Resting Heart Rate – DS 63 (55–67) 66 (62–68) 66 (60–68) 0.393a 
Resting Systolic BP – B (mm Hg) 125.5 (118–130) 126.5 (120–131) 125.5 (118–131) 0.677a 
Resting Systolic BP – EOS 127 (116–128) 118 (112–129) 123 (116–130) 0.637a 
Resting Diastolic BP – B (mm Hg) 80 (69–85) 75 (72–81) 79.5 (71–82) 0.705a 
Resting Diastolic BP – EOS 81 (69–88) 80 (78–80) 80 (74–86) 0.515a 
Respiratory exchange ratio B -VO2peak 1.2 (1.1–1.3) 1.3 (1.2–1.4) 1.2 (1.2–1.3) 0.036a 
Respiratory exchange ratio EOS 1.2 (1.2–1.3) 1.3 (1.3–1.4) 1.3 (1.2–1.3) 0.120a 
Relative VO2peak – B (mL/kg/min) 24.5 (21.7–33.0) 24.4 (21.1–28.2) 24.5 (21.3–31.1) 0.418a 
Relative VO2peak – EOS 29.1 (27.5–35.0) 24.7 (20.2–26.8) 27.9 (25.2–29.8) 0.007a 
Absolute VO2peak – B (L/min) 1.9 (1.4–2.2) 1.8 (1.7–2.0) 1.8 (1.7–2.1) 0.436a 
Absolute VO2peak – EOS 2.2 (2–2.4) 1.9 (1.2–2) 2 (1.9–2.3) 0.024a 
Metabolic equivalents – B (METs) 7 (5.8–9.4) 7 (6–8.1) 7 (6–8.9) 0.526a 
Metabolic equivalents – EOS 8.3 (7.9–10) 7.1 (5.8–7.7) 8 (7.2–8.5) 0.009a 
Circulating PGE2 – B 0.111 (0.085–0.123) 0.078 (0.068–0.093) 0.091 (0.075–0.111) 0.024a 
Circulating PGE2 – EOS 0.081 (0.075–0.084) 0.078 (0.075–0.095) 0.081 (0.074–0.090) >0.99a 
Colonic Tissue PGE2 – B (ng/mg) 25.6 (18.0–69.1) 9.4 (7.54–17.5) 18.8 (7.63–25.6) 0.0357a 
Colonic Tissue PGE2 – EOS 13.3 (4.38–31.5) 34.2 (18.5–48.8) 21.5 (5.65–39.4) 0.228a 
Circulating 12(s)-HHTrE – B 1.02 (0.54–1.53) 0.96 (0.61–1.08) 0.97 (0.57–1.45) 0.918a 
Circulating 12(s)-HHTrE – EOS 0.52 (0.45–0.67) 0.83 (0.56–1.21) 0.57 (0.47–0.83) 0.133a 
Group
ExerciseControlTotal
(N = 11)(N = 10)(N = 21)P value
Age 46 (40–48) 35.5 (34–38) 39 (34–47) 0.063a 
Sex, n (%)    >0.99b 
 Male 3 (27%) 3 (30%) 6 (29%)  
 Female 8 (73%) 7 (70%) 15 (71%)  
Ethnicity, n (%)    0.476b 
 Hispanic 0 (0%) 1 (10%) 1 (5%)  
 Non-Hispanic 11 (100%) 9 (90%) 20 (95%)  
Race, n (%)    >0.99b 
 White 10 (90%) 10 (100%) 20 (95%)  
 Asian 1 (10%) 0 (0%) 1 (5%)  
MMR gene mutated, n (%)     
MLH1 4 (36%) 5 (50%) 9 (42.9%)  
MSH2 5 (45%) 1 (10%) 6 (28.6%)  
MSH6 1 (9.1%) 4 (40%) 5 (23.8%)  
PMS2 1 (9.0%) 0 (0%) 1 (4.7%)  
BMI – B (kg/m227.4 (23.5–31.1) 27.4 (26.1–28.6) 27.4 (23.6–30.2) 0.734a 
BMI – EOS (kg/m225.3 (24.4–26.8) 28.1 (27.2–30.8) 26.9 (24.6–30.1) 0.168a 
Smoking History n (%)    >0.99b 
 Former smoker 2 (18%) 1 (10%) 3 (14%)  
 Never smoker 9 (82%) 9 (90%) 18 (86%)  
Hypertension (Hx + Meds), n (%)    >0.99b 
 No 11 (100%) 10 (0%) 21 (100%)  
 Yes 0 (0%) 0 (0%) 0 (0%)  
Diabetes, n (%)    >0.99b 
 No 11 (100%) 10 (0%) 21 (100%)  
 Yes 0 (0%) 0 (0%) 0 (0%)  
High Cholesterol (Hx + Meds), n (%)    0.476b 
 No 11 (100%) 9 (90%) 20 (95%)  
 Yes 0 (0%) 1 (10%) 1 (5%)  
History of cancer, n (%)     
 No 7 (64%) 7 (70%) 14 (66.7%) 1.0b 
 Yes 4 (36%) 3 (30%) 7 (33.3%)  
Type of Cancer, n (%)     
 Colorectal 2 (50%) 3 (100%) 5 (71%)  
 Thyroid 2 (50%) 2 (29%)  
Self-reported physical activity score 56 (20–83) 28.5 (23–49) 38 (23–62) 0.218a 
Weekly Fitbit wearing time (days) 6.1 (5.7–6.9) 5.7 (5.4–6.1) 6.0 (5.5–6.7) 0.280a 
Total time at 70–85 Peak Hear Rate (min) 6203 (4270–8338) 1076 (506–2727) 3744 (1092–5949) 0.0002a 
Total time at >85 Peak Hear Rate (min) 1668 (1317–1938) 156 (55–532) 851 (173–1659) 0.001a 
Exercise Self-Efficacy Score – B 61.5 (38–82) 64 (56–77) 64 (44.5–79) 0.734a 
Exercise Self-Efficacy Score – EOS 58 (38–77) 48.5 (46.5–52.8) 54 (44–61) 0.409a 
Resting Heart Rate – B (bpm) 71 (62–83) 86 (77–96) 79 (67–93) 0.152a 
Resting Heart Rate – EOS 68 (66–79) 78 (74–86) 77 (66–81) 0.258a 
Average Resting Heart Rate – DS 63 (55–67) 66 (62–68) 66 (60–68) 0.393a 
Resting Systolic BP – B (mm Hg) 125.5 (118–130) 126.5 (120–131) 125.5 (118–131) 0.677a 
Resting Systolic BP – EOS 127 (116–128) 118 (112–129) 123 (116–130) 0.637a 
Resting Diastolic BP – B (mm Hg) 80 (69–85) 75 (72–81) 79.5 (71–82) 0.705a 
Resting Diastolic BP – EOS 81 (69–88) 80 (78–80) 80 (74–86) 0.515a 
Respiratory exchange ratio B -VO2peak 1.2 (1.1–1.3) 1.3 (1.2–1.4) 1.2 (1.2–1.3) 0.036a 
Respiratory exchange ratio EOS 1.2 (1.2–1.3) 1.3 (1.3–1.4) 1.3 (1.2–1.3) 0.120a 
Relative VO2peak – B (mL/kg/min) 24.5 (21.7–33.0) 24.4 (21.1–28.2) 24.5 (21.3–31.1) 0.418a 
Relative VO2peak – EOS 29.1 (27.5–35.0) 24.7 (20.2–26.8) 27.9 (25.2–29.8) 0.007a 
Absolute VO2peak – B (L/min) 1.9 (1.4–2.2) 1.8 (1.7–2.0) 1.8 (1.7–2.1) 0.436a 
Absolute VO2peak – EOS 2.2 (2–2.4) 1.9 (1.2–2) 2 (1.9–2.3) 0.024a 
Metabolic equivalents – B (METs) 7 (5.8–9.4) 7 (6–8.1) 7 (6–8.9) 0.526a 
Metabolic equivalents – EOS 8.3 (7.9–10) 7.1 (5.8–7.7) 8 (7.2–8.5) 0.009a 
Circulating PGE2 – B 0.111 (0.085–0.123) 0.078 (0.068–0.093) 0.091 (0.075–0.111) 0.024a 
Circulating PGE2 – EOS 0.081 (0.075–0.084) 0.078 (0.075–0.095) 0.081 (0.074–0.090) >0.99a 
Colonic Tissue PGE2 – B (ng/mg) 25.6 (18.0–69.1) 9.4 (7.54–17.5) 18.8 (7.63–25.6) 0.0357a 
Colonic Tissue PGE2 – EOS 13.3 (4.38–31.5) 34.2 (18.5–48.8) 21.5 (5.65–39.4) 0.228a 
Circulating 12(s)-HHTrE – B 1.02 (0.54–1.53) 0.96 (0.61–1.08) 0.97 (0.57–1.45) 0.918a 
Circulating 12(s)-HHTrE – EOS 0.52 (0.45–0.67) 0.83 (0.56–1.21) 0.57 (0.47–0.83) 0.133a 

Note: Median values for each continuous variable in the specified groups were reported with the number in parentheses indicating either the 1st or 3rd quantile. Count numbers for binary or categorical data were also reported with the number in parentheses indicating the percentage of total counts.

Abbreviations: EOS, End of Study; B, Baseline; DS, During the Study; BP, blood pressure; bpm, beats per minute.

aWilcoxon rank-sum test P value.

bFisher exact test P value.

Feasibility

The overall recruitment rate to CYCLE-P was 35% (21/60). Adherence to the exercise intervention was self-reported through questionnaires administered via email. The mean adherence rate in the exercise group was 51.3% (range, 11.5%–73.1%). Nine of 10 participants in the exercise group completed CPET at Visit 4, yielding a retention rate of 81.2% (9/11), while 7 participants in the usual care group completed CPET at Visit 4, yielding a retention rate of 70.0% (7/10).

Adverse events

The number of participants with adverse events (AE) of any grade was 5/11 in the exercise group and 2/10 in the usual care group. AEs were graded according to the NCI Common Terminology Criteria for Adverse Events v.4.0. This difference was not statistically significant (P = 0.36). In the Exercise group, there were one grade 2 AE and four grade 1 AEs. In the usual care group, there were three grade 2 AEs. Thus, a total of eight AEs were reported. Grade 2 was the highest grade among all AEs observed. There was no difference in the AE rate by grade between the exercise and usual care groups (P = 0.43; Supplementary Tables S1–S3).

Effects of CYCling exercise on VO2peak and the colonic PGE2 levels

The median total weekly exercise minutes at a heart rate ≥70% over a 52-week period was 164 minutes (IQR, 88–265) in the exercise group and 14 minutes (IQR, 1–57) in the usual care group (Fig. 1B), respectively. There was no statistically significant difference between the exercise and the usual care group in terms of the number of days that participants used the Fitbit and their baseline HR. However, exercise participants had a significantly higher number of minutes with a heart rate of ≥70% of their PHR (Supplementary Fig. S3A–S3C). Compared with baseline, there was a 21.6% increase in mean relative VO2peak (+ 5.7 mL · kg−1 · min−1, P = 0.0056) in the exercise group as opposed to a 2.0% decrease (−0.47 mL · kg−1 · min−1, P = 0.44) in the usual care group, which rendered a significant difference when the exercise group was compared with the usual care group (21.6% increase vs. 2.0% decrease, P = 0.008 in t test; Fig. 1C; and P = 0.0029 in the mixed effect model as described in Supplementary Methods). In parallel, the exercise group had significantly decreased PGE2 levels in the colonic tissue relative to baseline when compared with the usual care group (P < 0.05 in t test; Fig. 1D; P = 0.03 in the mixed effect model). However, no significant differences between the exercise and usual care groups were observed in the levels of intermediate PG metabolites, including tetranor PGE1, 15-keto PGE2, and 13–14-Dihydro-15-Keto-PGE2, in colonic tissue (Supplementary Figures S4A and S4C).

Effects of CYCling exercise on the levels of circulating PGE2

Untargeted metabolomic analyses were performed to assess the effect of the cycling intervention on the serum metabolome. A total of 740 uniquely annotated metabolites were quantified. Differential analyses identified 29 metabolites that were statistically significantly altered following intervention in the exercise group but not in the usual care group (P < 0.05 in t test and in the mixed effect model): 7 were increased following exercise, whereas 22 were decreased (Supplementary Table S4). Among the 22 metabolites decreased after exercise were PGE2 and 12-Hydroxyheptadecatrienoic acid [12(s)-HHTrE], a primary metabolite of arachidonic acid metabolism via thromboxane synthase (Fig. 2AC). Moreover, there was a reduction in several circulating triacylglycerols (TG; n = 12), particularly those containing highly unsaturated long-chain fatty acids on the acyl groups that are key regulators in the production of COX2-mediated PGE2 (Fig. 2D).

Figure 2.

A, Overview of the metabolism of arachidonate to PGE2 and 12(s)-HHTrE; B, Changes in PGE2 before and after intervention in the usual care and exercise groups (*, P < 0.05); C, Waterfall plot illustrating fold-change in serum levels of PGE2 and 12(s)-HHTrE in usual care and exercise groups at the end-of-study relative to baseline. P values represent 2-sided paired t tests (*, P < 0.05); D, Heat map depicts the fold-change (end-of-study compared with baseline) for the 12 individual TGs that were statistically significant (2-sided paired t test P < 0.05) in the exercise group but not the usual care group.

Figure 2.

A, Overview of the metabolism of arachidonate to PGE2 and 12(s)-HHTrE; B, Changes in PGE2 before and after intervention in the usual care and exercise groups (*, P < 0.05); C, Waterfall plot illustrating fold-change in serum levels of PGE2 and 12(s)-HHTrE in usual care and exercise groups at the end-of-study relative to baseline. P values represent 2-sided paired t tests (*, P < 0.05); D, Heat map depicts the fold-change (end-of-study compared with baseline) for the 12 individual TGs that were statistically significant (2-sided paired t test P < 0.05) in the exercise group but not the usual care group.

Close modal

Differential gene expression analysis in colorectal normal mucosa between exercise and usual care groups

Transcriptomic analysis by mRNA-seq showed statistically significant changes in gene expression in colorectal normal mucosa between the exercise and usual care groups. Among differentially expressed genes (DEG) in the exercise group, 13 genes were upregulated, and 33 genes were downregulated (up to 10-fold) when compared with the usual care group (Fig. 3A and B; Supplementary Table S5). The top DEGs were C5orf17 (LINC02899), ABCG8 (ATP Binding Cassette Subfamily G Member 8), B4GALNT2 (Beta-1,4-N-Acetyl-Galactosaminyltransferase 2), and KCNV1 (Potassium Voltage-Gated Channel Modifier Subfamily V Member 1), while the downregulated genes were TMPRSS6 (Transmembrane Serine Protease 6), PRAC1/2 (Small nuclear protein PRAC1/2), and HOXB13 (Homeobox B13), and those genes are indicated as bold (Fig. 3A). Further, pathway enrichment analysis using Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and gene ontology biological process databases revealed that the top activated pathways in the exercise group were related to the immune system (Th17, Th1-Th2 cell differentiation, and Toll-like signaling) and others related to growth, survival, proliferation, and differentiation signals such as Rap1 (Ras-related protein 1), VEGF, and EGFR, as well as key cellular processes of DNA replication and cell-cycle control (Fig. 3C; Supplementary Table S6). The top suppressed pathways were related to the ribosome, cardiac muscle contraction, oxidative phosphorylation, and synaptic vesicle cycle. In addition, gene set enrichment analysis (GSEA) showed engagement of the IL6 pathway in the exercise group (P = 0.055; Supplementary Fig. S5A) using two independent transcriptome data sets generated in vitro for IL6 stimulation (25) and deprivation (26) showing positive enrichment scores (P = 2.9×10−5 and P = 8.9×10−8; respectively; Supplementary Fig. S5B and S5C).

Figure 3.

A, Heat map presenting the log fold change in significant genes compared with baseline after 12 months in the usual care and exercise groups; B, Volcano plot of statistically significant genes and fold-changes between the usual care and exercise groups. Top significantly DEGs are labeled; C, Dot plot presents enriched KEGG pathways. The sizes of the dots represent the count of core enrichment (leading-edge) genes. The colors of the dots represent the BH-adjusted P values. The order of KEGG gene sets is based on the gene ratio (number of significant genes associated with the KEGG gene sets/total number of significant genes). Immune-related pathways are bold; D and E, Scatter plot displaying correlations between selected variables. Blue line represents the linear regression line, and the grey band presents the 0.95 CI of the linear model; D, VO2 peak had a positive correlation with the change in activated NK cells (R = 0.57, P = 0.07); E, Change in PGE2 was negatively correlated (R = −0.58; P = 0.08) with the change in CD8+ T-cell population. Pearson correlation coefficient (R) and P value (P) are presented in the upper right corner of each plot.

Figure 3.

A, Heat map presenting the log fold change in significant genes compared with baseline after 12 months in the usual care and exercise groups; B, Volcano plot of statistically significant genes and fold-changes between the usual care and exercise groups. Top significantly DEGs are labeled; C, Dot plot presents enriched KEGG pathways. The sizes of the dots represent the count of core enrichment (leading-edge) genes. The colors of the dots represent the BH-adjusted P values. The order of KEGG gene sets is based on the gene ratio (number of significant genes associated with the KEGG gene sets/total number of significant genes). Immune-related pathways are bold; D and E, Scatter plot displaying correlations between selected variables. Blue line represents the linear regression line, and the grey band presents the 0.95 CI of the linear model; D, VO2 peak had a positive correlation with the change in activated NK cells (R = 0.57, P = 0.07); E, Change in PGE2 was negatively correlated (R = −0.58; P = 0.08) with the change in CD8+ T-cell population. Pearson correlation coefficient (R) and P value (P) are presented in the upper right corner of each plot.

Close modal

Correlation among aerobic exercise, VO2peak, PGE2 levels and changes in immune cell types

Overall, metabolic and transcriptomic analysis consistently showed a reduction of PGE2 levels and activation of immune-related pathways in the exercise group. Therefore, we performed in silico deconvolution of immune cell types in the colorectal mucosa using CIBERSORT. Overall, the CIBERSORT analysis using stringent criteria showed no significant changes in proportions of different immune cell types present in the colorectal mucosa between the exercise and usual care groups [Benjamini–Hochberg (BH)-Adjusted P < 0.05]; however, it was notable for the enrichment of CD8+ T cells with a non-statistically significant trend (Supplementary Fig. S6; Supplementary Table S7). Because VO2peak represents overall exercise capacity and was higher in the exercise group (Fig. 1C), we also analyzed the correlation between the change in VO2peak after 12 months and the change in other clinical outcomes and immune cell subtypes using scatter plot (Fig. 3D and E) and the Pearson correlation coefficient matrix (BH-Adjusted P < 0.1; Supplementary Fig. S7). These analyses revealed that changes of relative VO2peak had a positive statistically significant correlation with levels of activated natural killer (NK) cells (R = 0.58; BH-Adjusted P = 0.039; Fig. 3D). NK cells play essential roles in innate immunity towards cancer, with the ability to kill circulating cancer cells (27). In parallel, consistent with the known role of PGE2 as a regulator of an adaptive immune response, we observed that changes in PGE2 (reduction in levels) were negatively correlated with changes of CD8+ T cells (R = −0.66; BH-Adjusted P = 0.01; Fig. 3E), thus suggesting that cycling exercise promoted the activation and recruitment of cytotoxic T cells in colorectal mucosa, which is one of the most critical components of adaptive anticancer immunity (28, 29).

CYCling exercise affects cancer-related pathways and changes the NK cell level at colon crypts

To confirm the change of the immune response pathways in trial participants, we performed spatial transcriptomics using the NanoString GeoMx Digital Spatial Profiler (DSP). A total of 16 formalin-fixed, paraffin-embedded (FFPE) samples of normal mucosa from 8 participants were available for this analysis (3 participants in the usual care and 5 in the exercise group; Fig. 4A). We focused the analysis in areas enriched by colonic crypts and assessed differences between the usual care and exercise group observing downregulation of metallothionein genes (MT1A, MT1E, MT1F, MT1G, MT1M, MT1X, and MT2A), and several small and large subunit ribosomal proteins in the exercise group compared with the usual care group (Fig. 4B; Supplementary Fig. S8A). Expression levels of cancer stem cell markers CD24 and TSPAN8, and oncogenes EIF3E, BTF3, ST14, and HMGCS also decreased in the exercise group. GSEA from DSP data revealed in the exercise group suppression of fat and carbohydrate metabolism, androgen and estrogen response, along with several oncogenic pathways including MYC, MTORC1, and TGFβ and activation of KRAS signaling downregulation, IFNγ, and inflammatory responses (Supplementary Fig. S8B). Immune cell deconvolution from DSP expression data confirmed that the exercise group had significantly increased numbers of activated NK cells compared with the usual care group (P = 0.036) and increase of CD8+ T cells (Fig. 4C). Then, we performed analysis of the frequency of CD8+ T and CD57+ NK cells in residual FFPE tissue sections of colonic mucosa of 4 participants in the exercise and 4 in the usual care group using IHC. We confirmed a significant increase of the total number of CD8+ T cells in the exercise group (Supplementary Fig. S8C, left) due to the stromal CD8+ T-cell population (Fig. 4D, left) with an increasing trend in the epithelial CD8+ T cells (Supplementary Fig. S8C, right). A marginal increase in total number of CD57+ NK cells, mainly due to the stromal population, was observed in the exercise group (Fig. 4D, right and Supplementary Fig. S8D). These findings suggest that levels of CTLs are promoted in colonic mucosa by aerobic exercise with concomitant suppression of oncogenic pathways particularly TGFβ signaling which plays a negative regulator of immune cells. In summary, our working model derived from the CYCLE-P study demonstrates that LS participants undergoing exercise through cycling for one year increase cardiorespiratory fitness (VO2peak) and decrease pro-inflammatory and metabolic regulators, most notably, circulating PGE2 and TGs and end-organ colonic PGE2. These molecules converge promoting activation of the immune system in the large intestine by increasing cytotoxic CD8+ T and CD57+ NK cells. Increased VO2peak upon aerobic exercise has been linked to the production of different myokines and cytokines serving as immune modulators (Fig. 4E). Taken together, these pathways triggered by exercise may elicit adaptive antitumor immunity in LS carriers, thus eliciting cancer prevention.

Figure 4.

A, Tissue sections of colorectal mucosa specimens were stained with H&E, analyzed using GeoMx DSP and IHC to validate the change of the immune cell types; B, Heat map presents the log fold change of significant genes comparing 12 months to baseline in the usual care (N = 3) and exercise group (N = 5) in the DSP experiment. Significant genes were separated by colors, metallothionein genes in red, ribosomal genes in green, and cancer related genes in blue; C, Changes in deconvoluted immune cell type proportions derived from DSP data are shown after twelve months of follow-up in usual care and exercise groups using CIBERSORT; D, Stromal CD8+ T cell density per mm2 (left) and CD57+ NK cells (right) in colonic mucosa pre- and post-intervention from usual care (n = 4) and exercise (n = 4) participants; E, Exercise through cycling promotes activation of CD8+ T and CD57+ NK cells via modulation of PGs. Upward green arrow indicates activation and downward red arrow shows inhibition.

Figure 4.

A, Tissue sections of colorectal mucosa specimens were stained with H&E, analyzed using GeoMx DSP and IHC to validate the change of the immune cell types; B, Heat map presents the log fold change of significant genes comparing 12 months to baseline in the usual care (N = 3) and exercise group (N = 5) in the DSP experiment. Significant genes were separated by colors, metallothionein genes in red, ribosomal genes in green, and cancer related genes in blue; C, Changes in deconvoluted immune cell type proportions derived from DSP data are shown after twelve months of follow-up in usual care and exercise groups using CIBERSORT; D, Stromal CD8+ T cell density per mm2 (left) and CD57+ NK cells (right) in colonic mucosa pre- and post-intervention from usual care (n = 4) and exercise (n = 4) participants; E, Exercise through cycling promotes activation of CD8+ T and CD57+ NK cells via modulation of PGs. Upward green arrow indicates activation and downward red arrow shows inhibition.

Close modal

The results of this proof-of-concept and small trial showed that 12 months of sustained aerobic exercise increased absolute VO2peak in LS carriers. Additionally, biomarker studies performed in the normal mucosa and plasma of patients with LS showed that exercise modulates inflammation via the reduction of PGE2 and it has a role in the modulation of the activity of the immune system with increased activation of NK and CD8+ T cells. Overall, our data highlight the role and potential of aerobic exercise as a non-pharmacologic preventive intervention in LS population.

In the study, we achieved a recruitment rate of 35%, which exceeded our prespecified target and was within the range of other exercise clinical trials. The retention rates of 81.2% (9/11) and 70% (7/10) for the exercise and usual groups, respectively, over the 12-month study, were also in line with our expectations of 75%. On the basis of the wearable device information, the intervention group achieved, on average, a PHR of ≥70% during exercise for a significantly greater amount of time (164 min/wk) than the usual care group (14 min/wk) over the 52-week study period. Thus, we conclude that it is feasible to recruit and retain LS participants to a cycling intervention. However, adjustments to measure and promote adherence throughout the study period are warranted for future studies.

Two prior studies have assessed the relationship between exercise and PGE2 levels in colorectal tissue. Martinez and colleagues assessed the PGE2 levels in the rectal mucosa of 63 participants with a prior history of polyps and found an inverse association between self-reported exercise (MET-hr-wk) and PGE2 levels (−0.28, P < 0.05) and 28% lower PGE2 levels among those with a reported 22 MET-hr-wk difference in activity (19). Limitations included self-reporting of exercise and the cross-sectional nature of the study design. In a second study, Campbell and colleagues performed a 12-month exercise intervention (combination of supervised and home-based aerobic exercise) in 187 men and women 40 to 75 years of age, reporting a mean 370 minutes a week of exercise in men and 295 minutes a week in women over the study period based on study logs (30). VO2max increased by 2.5 mL · kg−1 · min−1 (10%) in women and 3.3 mL · kg−1 · min−1 (11%) in men in the exercise group and decreased in the usual care group. Of the 187 participants, 89 had follow-up biopsies and subsequent measurement of colonic PGE2 levels, demonstrating no differences in either men (P = 0.74) or women (P = 0.99; ref. 22). Differences in PGE2 levels by VO2peak change were not reported. A combination of factors may explain the discordant findings from these studies and our results. First, the technological advances in PGE2 measurement and the ascertainment of two colonic biopsies for each participant in our CYCLE-P study might likely improve sensitivity for PGE2 detection. Second, it is possible that a more robust change in VO2peak (4.6 mL · kg−1 · min−1) over 12 months in our trial allowed for a greater intervention effect to detect a difference in PGE2. Of note, we tracked minutes of exercise based on participants meeting a threshold heart rate greater than 70% of PHR (not below this threshold), while Campbell and colleagues asked participants to reach 60% to 80% of PHR but reported only overall minutes, not intensity. Therefore, an absolute “exercise dose” difference between the trials cannot be established. Third, we studied a high-risk genetic cohort of LS participants, and Martinez and colleagues recruited participants with a history of polyps, while Campbell and colleagues enrolled participants with no known high-risk features. This poses the question of whether individuals “at-risk” for colorectal cancer are more susceptible to the effects of exercise on colonic tissue. This question will require additional experimental studies applying similar methods across different risk cohorts.

Our findings are biologically plausible, showing an effect of exercise on the COX pathway, consistent with modulation of serum levels of TGs with sustained aerobic exercise. Endogenous TGs are the largest energy source of the body. Fatty acid oxidation is the preferred substrate utilized during sustained bouts of exercise to delay the onset of skeletal muscle glycogen depletion. Our results, supported by prior work (31), showed a decrease in TGs in the resting state among the exercise participants, not observed in the usual care group. Furthermore, we found that long-chain highly unsaturated TGs such as arachidonate along with downstream metabolites, PGE2 and 12 (s) – HHTrE, were lower in the exercise group than in the usual care group. To date, there have been few studies assessing change in the circulating metabolome following chronic exercise utilizing current day metabolomics platforms. Brennen and colleagues found no change in 147 metabolites profiled by liquid chromatography/tandem mass spectrometry among 216 middle-aged abdominally obese men and women following a 24-week exercise intervention (32). In a second study by Koay and colleagues, 52 healthy, normal weight soldiers underwent an 80-day intervention incorporating ∼ 8 MET-hrs per day of exercise, resulting in a 5.34 mL · kg−1 · min−1 in VO2peak following the training period. Metabolomics analysis demonstrated a 1.39-fold decrease in arachidonate (P = 6.6×10−8; ref. 33). Both our study and Koay and colleagues performed a within-group analysis to reduce the interindividual variability of group comparisons, demonstrated a robust effect size change in VO2peak and performed an unbiased assessment of a full range of circulating metabolites. Differences include that our study had a control group (demonstrating no change in arachidonate), and that we delivered an aerobic intervention at a lower exercise dose over a longer time period than Koay and colleagues.

Our transcriptomic data further support the above findings that altered expression of PRAC1/2, ABCG8, B4GALNT2, and HOXB13 affect the modulation of inflammation, immune cell types resident in the mucosa, and cholesterol levels in the LS colorectal environment with sustained aerobic exercise. ABCG8 acts as a regulator of sterols removal (34, 35), B4GALNT2 reduces inflammation in skeletal muscle and regulates the gut microbiome (36, 37), PRAC1/2 genes are expressed in the epithelium of inflamed colon in ulcerative colitis (38), and HOXB13 expression has been reported to correlate with hepatic inflammation and fibrosis (39). In addition, we observed activation of IL6 signaling in the exercise group. IL6 is an important regulator with diverse roles in exercise, PG metabolism, and antitumor immunity through activation of NK, CD8a T cells and other effector T cells (40–43). In fact, PGE2 upregulates the secretion of muscle-derived IL6 into circulation during exercise (44–47). These antitumor functions mainly happen outside the tumor microenvironment. In fact, although the activation of the IL6 pathway was strong, we did not observe overexpression of IL6 mRNA transcripts in the mucosa, thus suggesting that the mechanism of activation may be derived from endocrine secretion by cells outside any tumor microenvironment, such as muscle cells stimulated by exercise rather than colorectal epithelial cells. The main finding among participants in the exercise group was the activation and increased levels of CD8+ T and CD57+ NK cells in the colonic mucosa, though observed nonsignificant increase initially in the immune cell type deconvolution results of total RNA transcriptomic data, but verified independently using a reliable and sensitive approach of NanoString DSP and then by IHC analysis. Furthermore, spatial transcriptomic profiling of the colonic crypts revealed the downregulation of KRAS signaling and activation of immune cell regulatory pathways including inflammatory and IFNγ responses. KRAS signaling is commonly dysfunctional in colon cancer (48). Taken together, we postulate that exercise might be considered as a potential KRAS-targeting cancer treatment and an immune modulator for increased adaptive immune response and a potential KRAS targeting cancer intervention. The added value is that our exercise intervention was very reasonable and so were the cardiorespiratory measurements achieved to the point that it can be considered an exercise prescription for patients with LS seeking to boost immunity and obtain specific pathway-centered anticancer effects.

Limitations to our study include the small sample size without race heterogeneity, and the non-randomized nature of the design. In addition, our validation studies using DSP and IHC may not allow to generalize our results due to interindividual differences among the participants of the exercise group with a relative small sample size. Participants knew their allocation before signing informed consent, which might have impacted their willingness to participate in the trial. In addition, differences in timing of collection of the specimens and storage in the freezer could have driven differences in metabolite levels. Importantly, the study groups were remarkably similar other than age. The exercise group had older participants, which would result in a bias towards a type II error or a false negative result, the opposite of what we have observed. It is important to note that our inclusion criteria were intentionally geared to recruit healthy patients with LS, who could attain high levels of exercise, and to exclude individuals with hypertension, diabetes, cardiovascular disease, or inflammatory disorders. As such, we removed potential confounders to enhance the signal of the exercise intervention. Further, the role of gut microbiome, diet, and supplements taken by participants remain unexplored in this study. This could be critical as several studies have shown that levels of physical activity have differential effects on the intestinal microbiome that may involve in reduction of colorectal cancer risk (8, 49).

In conclusion, our study demonstrates for the first time that aerobic exercise is capable of modulating levels of inflammatory PGs in the colonic tissue and serum in a high-risk genetic cohort for cancer due to mismatch repair deficiency. The significant decrease in mucosal PGE2 levels correlated significantly with an increased activation or recruitment of specific immune cell populations of NK and CD8+ T cells to the colonic mucosa. This may be related to increased immune surveillance in the colon and, therefore, linked to a benefit in colorectal cancer prevention through clearance of dysplastic cell populations that develop in a background of MMR deficiency. Future randomized clinical trials will be needed to confirm the preventive efficacy of aerobic exercise training in LS carriers and to further elucidate the possible immune-related pathways underlying any reductions in colorectal cancer risk.

Written informed consent was obtained from all study participants, and The University of Texas MD Anderson Cancer Center Institutional Review Board (IRB) approved this study (IRB #2017–1035).

M.F. Munsell reports grants from NIH during the conduct of the study. E.T. Hawk reports grants from NCI and Exact Sciences and other support from The University of Texas Boone Pickens Distinguished Chair for Early Prevention of Cancer during the conduct of the study; other support from AACR and grants from Cancer Prevention & Research Institute of Texas outside the submitted work. Y. Li reports other support from Angenus Inc. and Mink Therapeutics Inc. outside the submitted work. J.A. Wargo reports personal fees from Imedex, Dava Oncology, Omniprex, Illumina, Gilead, PeerView, Physician Education Resource, MedImmune, Exelixis, and Bristol-Myers Squibb outside the submitted work; has a patent for PCT/US17/53.717 pending; has served as a consultant/advisory board member for Roche/Genentech, Novartis, AstraZeneca, GlaxoSmithKline, Bristol-Myers Squibb, Micronoma, OSE therapeutics, Merck, and Everimmune; in addition, receives stock options from Micronoma and OSE therapeutics. P. Sharma reports other support from Achelois, Adaptive Biotechnologies, Affini-T, Apricity, Asher Bio, BioAtla LLC, BioNTech, Candel Therapeutics, Catalio, Carisma, Codiak Biosciences Inc., C-Reveal Therapeutics, Dragonfly Therapeutics, Earli Inc, Enable Medicine, Glympse, Henlius/Hengenix, Hummingbird, ImaginAB, Infinity Pharma, Intervenn Biosciences, JSL Health, LAVA Therapeutics, Lytix Biopharma, Marker Therapeutics, Oncolytics, PBM Capital, Phenomic AI, Polaris Pharma, Sporos, Time Bioventures, Trained Therapeutix Discovery, and Two Bear Capital; and other support from Xilis Inc. outside the submitted work. E. Vilar reports grants and personal fees from Janssen Research and Development and personal fees from Recursion Pharma, Guardant Health, and Rising Tide Foundation outside the submitted work. No disclosures were reported by the other authors.

N. Deng: Data curation, formal analysis, writing–original draft. L. Reyes-Uribe: Data curation, formal analysis, writing–original draft. J.F. Fahrmann: Formal analysis, investigation, writing–original draft. W.S. Thoman: Conceptualization, supervision, investigation. M.F. Munsell: Formal analysis, validation. J.B. Dennison: Formal analysis, writing–review and editing. E. Murage: Formal analysis, writing–review and editing. R. Wu: Formal analysis, writing–review and editing. E.T. Hawk: Writing–review and editing. S. Thirumurthi: Investigation, writing–review and editing. P.M. Lynch: Investigation, writing–review and editing. C.M. Dieli-Conwright: Conceptualization. A.J. Lazar: Formal analysis, writing–review and editing. S. Jindal: Formal analysis, validation, writing–review and editing. K. Chu: Formal analysis, validation, writing–review and editing. M. Chelvanambi: Validation, writing–review and editing. K. Basen-Engquist: Writing–review and editing. Y. Li: Formal analysis. J.A. Wargo: Writing–review and editing. F. McAllister: Writing–review and editing. J.P. Allison: Writing–review and editing. P. Sharma: Formal analysis, writing–review and editing. K.M. Sinha: Formal analysis, writing–review and editing. S. Hanash: Writing–review and editing. S.C. Gilchrist: Formal analysis, supervision, investigation, writing–original draft, writing–review and editing. E. Vilar: Conceptualization, supervision, funding acquisition, investigation, methodology, writing–review and editing.

We thank the patients and their families for their participation. We thank the MDACC Clinical Cancer Prevention Research Core for their assistance in the conduction of this study. We thank the staff of the Advanced Technology Genomics Core for the assistance with RNA sequencing and the Immunotherapy Platform for the assistance with the IHC, both at MDACC. The authors are grateful to Karen Colbert Maresso for critically reading the manuscript.

This work was supported by a gift from the Feinberg Family Foundation to E. Vilar; a grant and a faculty fellowship from The University of Texas MD Anderson Cancer Center Duncan Family Institute for Cancer Prevention and Risk Assessment and the T. Boone Pickens Endowment to S.C. Gilchrist and E.T. Hawk; CA016672 (NIH/NCI) to The University of Texas MD Anderson Cancer Center Core Support Grant, P50 CA221707 (NIH/NCI) to The University of Texas MD Anderson Cancer Center GI SPORE Grant, and the generous philanthropic contributions to The University of Texas MD Anderson Cancer Center Moon Shots Program.

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

1.
Lynch
HT
,
Snyder
CL
,
Shaw
TG
,
Heinen
CD
,
Hitchins
MP
.
Milestones of lynch syndrome: 1895–2015
.
Nat Rev Cancer
2015
;
15
:
181
94
.
2.
Dominguez-Valentin
M
,
Sampson
JR
,
Seppälä
TT
,
ten Broeke
SW
,
Plazzer
J-P
,
Nakken
S
, et al
.
Cancer risks by gene, age, and gender in 6350 carriers of pathogenic mismatch repair variants: findings from the prospective lynch syndrome database
.
Genet Med
2020
;
22
:
15
25
.
3.
Wolin
KY
,
Yan
Y
,
Colditz
GA
,
Lee
I-M
.
Physical activity and colon cancer prevention: a meta-analysis
.
Br J Cancer
2009
;
100
:
611
.
4.
Lee
I-M
,
Paffenbarger
RS
Jr
,
Hsieh
C
.
Physical activity and risk of developing colorectal cancer among college alumni
.
J Natl Cancer Inst
1991
;
83
:
1324
9
.
5.
Kraemer
MB
,
Priolli
DG
,
Reis
IGM
,
Pelosi
AC
,
Garbuio
ALP
,
Messias
LHD
.
Home-based, supervised, and mixed exercise intervention on functional capacity and quality of life of colorectal cancer patients: a meta-analysis
.
Sci Rep
2022
;
12
:
2471
.
6.
Coletta
AM
,
Peterson
SK
,
Gatus
LA
,
Krause
KJ
,
Schembre
SM
,
Gilchrist
SC
, et al
.
Energy balance related lifestyle factors and risk of endometrial and colorectal cancer among individuals with lynch syndrome: a systematic review
.
Fam Cancer
2019
;
18
:
399
420
.
7.
Dashti
SG
,
Win
AK
,
Hardikar
SS
,
Glombicki
SE
,
Mallenahalli
S
,
Thirumurthi
S
, et al
.
Physical activity and the risk of colorectal cancer in Lynch syndrome
.
Int J Cancer
2018
;
143
:
2250
60
.
8.
Boytar
AN
,
Nitert
MD
,
Morrision
M
,
Skinner
TL
,
Jenkins
DG
.
Exercise-induced changes to the human gut microbiota and implications for colorectal cancer: a narrative review
.
J Physiol
2022
;
600
:
5189
201
.
9.
Song
M
,
Chan
AT
,
Sun
J
.
Influence of the gut microbiome, diet, and environment on risk of colorectal cancer
.
Gastroenterology
2020
;
158
:
322
40
.
10.
Friedenreich
CM
,
Neilson
HK
,
Farris
MS
,
Courneya
KS
.
Physical activity and cancer outcomes: a precision medicine approach
.
Clin Cancer Res
2016
;
22
:
4766
75
.
11.
Hanyuda
A
,
Kim
SA
,
Martinez-Fernandez
A
,
Qian
ZR
,
Yamauchi
M
,
Nishihara
R
, et al
.
Survival benefit of exercise differs by tumor IRS1 expression status in colorectal cancer
.
Ann Surg Oncol
2016
;
23
:
908
17
.
12.
Kerr
J
,
Anderson
C
,
Lippman
SM
.
Physical activity, sedentary behavior, diet, and cancer: an update and emerging new evidence
.
Lancet Oncol
2017
;
18
:
e457
71
.
13.
Koh
H
,
Hamada
T
,
Song
M
,
Liu
L
,
Cao
Y
,
Nowak
JA
, et al
.
Physical activity and colorectal cancer prognosis according to tumor-infiltrating T cells
.
JNCI Cancer Spectr
2018
;
2
:
pky058
.
14.
Darband
SG
,
Sadighparvar
S
,
Ghaderi Pakdel
F
,
Naderi
S
,
Majidinia
M
.
Involvement of IGF/IGFBP/Erk axis in the exercise-mediated preventive effects on colorectal cancer in rats
.
Int J Clin Exp Pathol
2021
;
14
:
608
17
.
15.
Onerup
A
,
Thörn
S-E
,
Angenete
E
,
Bock
D
,
Grybäck Gillheimer
E
,
Haglind
E
, et al
.
Effects of a home-based exercise program on the insulin-like growth factor axis in patients operated for colorectal cancer in Sweden: results from the randomized controlled trial PHYSSURG-C
.
Growth Horm IGF Res
2020
;
51
:
27
33
.
16.
Wadley
AJ
,
Aldred
S
,
Coles
SJ
.
An unexplored role for peroxiredoxin in exercise-induced redox signaling?
Redox Biol
2016
;
8
:
51
8
.
17.
Dethlefsen
C
,
Hansen
LS
,
Lillelund
C
,
Andersen
C
,
Gehl
J
,
Christensen
JF
, et al
.
Exercise-induced catecholamines activate the hippo tumor suppressor pathway to reduce risks of breast cancer development
.
Cancer Res
2017
;
77
:
4894
904
.
18.
Rogers
CJ
,
Colbert
LH
,
Greiner
JW
,
Perkins
SN
,
Hursting
SD
.
Physical activity and cancer prevention: pathways and targets for intervention
.
Sports Med
2008
;
38
:
271
96
.
19.
Martinez
ME
,
Heddens
D
,
Earnest
DL
,
Bogert
CL
,
Roe
D
,
Einspahr
J
, et al
.
Physical activity, body mass index, and prostaglandin E2 levels in rectal mucosa
.
J Natl Cancer Inst
1999
;
91
:
950
3
.
20.
Reyes-Uribe
L
,
Wu
W
,
Gelincik
O
,
Bommi
PV
,
Francisco-Cruz
A
,
Solis
LM
, et al
.
Naproxen chemoprevention promotes immune activation in Lynch syndrome colorectal mucosa
.
Gut
2021
;
70
:
555
66
.
21.
Burn
J
,
Gerdes
A-M
,
Macrae
F
,
Mecklin
J-P
,
Moeslein
G
,
Olschwang
S
, et al
.
Long-term effect of aspirin on cancer risk in carriers of hereditary colorectal cancer: an analysis from the CAPP2 randomized controlled trial
.
Lancet
2011
;
378
:
2081
7
.
22.
Abrahamson
PE
,
King
IB
,
Ulrich
CM
,
Rudolph
RE
,
Irwin
ML
,
Yasui
Y
, et al
.
No effect of exercise on colon mucosal prostaglandin concentrations: a 12-month randomized controlled trial
.
Cancer Epidemiol Biomarkers Prev
2007
;
16
:
2351
6
.
23.
Haynes
AB
,
Haukoos
JS
,
Dimick
JB
.
TREND reporting guidelines for nonrandomized/quasi-experimental study designs
.
JAMA Surgery
2021
;
156
:
879
80
.
24.
Myers
J
,
Arena
R
,
Franklin
B
,
Pina
I
,
Kraus
WE
,
McInnis
K
, et al
.
Recommendations for clinical exercise laboratories: a scientific statement from the American heart association
.
Circulation
2009
;
119
:
3144
61
.
25.
Dasu
MR
,
Hawkins
HK
,
Barrow
RE
,
Xue
H
,
Herndon
DN
.
Gene expression profiles from hypertrophic scar fibroblasts before and after IL6 stimulation
.
J Pathol
2004
;
202
:
476
85
.
26.
Croonquist
PA
,
Linden
MA
,
Zhao
F
,
Van Ness
BG
.
Gene profiling of a myeloma cell line reveals similarities and unique signatures among IL6 response, N-ras-activating mutations, and coculture with bone marrow stromal cells
.
Blood
2003
;
102
:
2581
92
.
27.
Jobin
G
,
Rodriguez-Suarez
R
,
Betito
K
.
Association between natural killer cell activity and colorectal cancer in high-risk subjects undergoing colonoscopy
.
Gastroenterology
2017
;
153
:
980
7
.
28.
Galon
J
,
Costes
A
,
Sanchez-Cabo
F
,
Kirilovsky
A
,
Mlecnik
B
,
Lagorce-Pagès
C
, et al
.
Type, density, and location of immune cells within human colorectal tumors predict clinical outcome
.
Science
2006
;
313
:
1960
4
.
29.
Zhang
L
,
Conejo-Garcia
JR
,
Katsaros
D
,
Gimotty
PA
,
Massobrio
M
,
Regnani
G
, et al
.
Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer
.
N Engl J Med
2003
;
348
:
203
13
.
30.
Campbell
KL
,
McTiernan
A
,
Li
SS
,
Sorensen
BE
,
Yasui
Y
,
Lampe
JW
, et al
.
Effect of a 12-month exercise intervention on the apoptotic regulating proteins Bax and Bcl-2 in colon crypts: a randomized controlled trial
.
Cancer Epidemiol Biomarkers Prev
2007
;
16
:
1767
74
.
31.
Kraus
WE
,
Houmard
JA
,
Duscha
BD
,
Knetzger
KJ
,
Wharton
MB
,
McCartney
JS
, et al
.
Effects of the amount and intensity of exercise on plasma lipoproteins
.
N Engl J Med
2002
;
347
:
1483
92
.
32.
Brennan
AM
,
BensoN
M
,
Morningstar
J
,
Herzig
M
,
Robbins
J
,
Gerszten
RE
, et al
.
Plasma metabolite profiles in response to chronic exercise
.
Med Sci Sports Exerc
2018
;
50
:
1480
6
.
33.
Koay
YC
,
Stanton
K
,
Kienzle
V
,
Li
M
,
Yang
J
,
Celermajer
DS
, et al
.
Effect of chronic exercise in healthy young male adults: a metabolomic analysis
.
Cardiovasc Res
2021
;
117
:
613
22
.
34.
Baldan
A
,
Bojanic
DD
,
Edwards
PA
.
The ABCs of sterol transport
.
J Lipid Res
2009
;
50
:
S80
5
.
35.
Fitzgerald
ML
,
Mujawar
Z
,
Tamehiro
N
.
ABC transporters, atherosclerosis, and inflammation
.
Atherosclerosis
2010
;
211
:
361
70
.
36.
Xu
R
,
Singhal
N
,
Serinagaoglu
Y
,
Chandrasekharan
K
,
Joshi
M
,
Bauer
JA
, et al
.
Deletion of Galgt2 (B4Galnt2) reduces muscle growth in response to acute injury and increases muscle inflammation and pathology in dystrophin-deficient mice
.
Am J Pathol
2015
;
185
:
2668
84
.
37.
Rausch
P
,
Steck
N
,
Suwandi
A
,
Seidel
JA
,
Künzel
S
,
Bhullar
K
, et al
.
Expression of the blood group–related gene B4galnt2 alters susceptibility to salmonella infection
.
PLoS Pathog
2015
;
11
:
e1005008
.
38.
Barnicle
A
,
Seoighe
C
,
Greally
JM
,
Golden
A
,
Egan
LJ
.
Inflammation-associated DNA methylation patterns in epithelium of ulcerative colitis
.
Epigenetics
2017
;
12
:
591
606
.
39.
Zuo
L
,
Tan
T
,
Wei
C
,
Wang
H
,
Tan
L
,
Hao
Y
, et al
.
HOXB13 expression is correlated with hepatic inflammatory activity of patients with hepatic fibrosis
.
J Mol Histol
2020
;
51
:
183
9
.
40.
Miller
S
,
Senior
PV
,
Prakash
M
,
Apostolopoulos
V
,
Sakkal
S
,
Nurgali
K
.
Leukocyte populations and IL6 in the tumor microenvironment of an orthotopic colorectal cancer model
.
Acta Biochim Biophys Sin
2016
;
48
:
334
41
.
41.
Gustafsson
K
,
Ingelsten
M
,
Bergqvist
L
,
Nyström
J
,
Andersson
B
,
Karlsson-Parra
A
.
Recruitment and activation of natural killer cells in vitro by a human dendritic cell vaccine
.
Cancer Res
2008
;
68
:
5965
71
.
42.
Huang
S-P
,
Wu
M-S
,
Shun
C-T
,
Wang
H-P
,
Lin
M-T
,
Kuo
M-L
, et al
.
Interleukin-6 increases vascular endothelial growth factor and angiogenesis in gastric carcinoma
.
J Biomed Sci
2004
;
11
:
517
27
.
43.
Van Pelt
DW
,
Guth
LM
,
Horowitz
JF
.
Aerobic exercise elevates markers of angiogenesis and macrophage IL6 gene expression in the subcutaneous adipose tissue of overweight-to-obese adults
.
J Appl Physiol
2017
;
123
:
1150
9
.
44.
Ostrowski
K
,
Rohde
T
,
Zacho
M
,
Asp
S
,
Pedersen
BK
.
Evidence that interleukin-6 is produced in human skeletal muscle during prolonged running
.
J Physiol
1998
;
508
:
949
53
.
45.
Chowdhury
S
,
Schulz
L
,
Palmisano
B
,
Singh
P
,
Berger
JM
,
Yadav
VK
, et al
.
Muscle-derived interleukin 6 increases exercise capacity by signaling in osteoblasts
.
J Clin Invest
2020
;
130
:
2888
902
.
46.
Standley
RA
,
Liu
SZ
,
Jemiolo
B
,
Trappe
SW
,
Trappe
TA
.
Prostaglandin E2 induces transcription of skeletal muscle mass regulators interleukin-6 and muscle RING finger-1 in humans
.
Prostaglandins Leukot Essent Fatty Acids
2013
;
88
:
361
4
.
47.
Steensberg
A
,
Hall
G
,
Osada
T
,
Sacchetti
M
,
Saltin
B
,
Pedersen
BK
.
Production of interleukin-6 in contracting human skeletal muscles can account for the exercise-induced increase in plasma interleukin-6
.
J Physiol
2000
;
529
Pt 1
:
237
42
.
48.
Dinu
D
,
Dobre
M
,
Panaitescu
E
,
Bîrlă
R
,
Iosif
C
,
Hoara
P
, et al
.
Prognostic significance of KRAS gene mutations in colorectal cancer–preliminary study
.
J Med Life
2014
;
7
:
581
7
.
49.
Himbert
C
,
Stephens
WZ
,
Gigic
B
,
Hardikar
S
,
Holowatyj
AN
,
Lin
T
, et al
.
Differences in the gut microbiome by physical activity and BMI among colorectal cancer patients
.
Am J Cancer Res
2022
;
12
:
4789
801
.
This open access article is distributed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) license.

Supplementary data