Background:

Serrated polyps (SP) are important colorectal cancer precursors, yet their epidemiology is incompletely understood. We measured risk factors for incident sessile-serrated lesions (SSL) and microvesicular (MVHP) and goblet-cell rich (GCHP) hyperplastic polyp subtypes.

Methods:

We conducted a cohort study of patients undergoing colonoscopic surveillance nested within a chemoprevention trial. Outcomes of interest were ≥1 SPs, including SSLs, MVHPs, and GCHPs specifically. Multivariable generalized estimating equation models were used to estimate adjusted risk ratios (RR) and 95% confidence intervals (CI) for different polyp types.

Results:

Among 2,102 participants, a total of 1,615 SPs (including 212 SSLs) were found among 758 participants during follow-up. Prior history of SPs was strongly associated with subsequent occurrence of SPs. There was no apparent association between age, sex, or education and risk of SPs. Black participants were at lower risk of SSLs and MVHPs, but higher risk of GCHPs compared with white participants [RR, 0.40; 95% CI, 0.16–0.99); RR, 0.63 (95% CI, 0.42–0.96); and RR, 1.83 (95% CI, 1.23–2.72) respectively]. Alcohol and smoking exposure were also associated with SPs, including hyperplastic polyp subtypes in particular.

Conclusions:

In this prospective study, the risk of SP subtypes differed by race, alcohol, and smoking status, and prior history of SPs. Risk factor associations for SPs differ from risk factors for conventional adenomas, supporting the concept of etiologic heterogeneity of colorectal cancer.

Impact:

These findings allow for better risk stratification of patients undergoing colorectal cancer screening and could inform screening test selection.

Serrated polyps are important colorectal cancer precursors, responsible for up to 25% of sporadic colorectal cancer (1–3). The common histologic feature that links serrated polyps is the serration of the colonic crypt epithelium. This characteristic is found in the hyperplastic polyp (HP), the sessile serrated lesion (SSL, also called sessile serrated polyp or sessile serrated adenoma), and the traditional serrated adenoma (TSA; ref. 3). HPs are the most common serrated lesion, accounting for roughly 60% to 70% of all serrated polyps, and the most innocuous. Most HPs are small, located in the distal colorectum, and have little to no malignant potential. SSLs are less common, occurring in up to 14% of average risk patients undergoing screening colonoscopy (4), and likely generate the most colorectal cancers from this pathway. TSAs are also cancer precursors, but are the least commonly found serrated polyp subtype.

Serrated carcinogenesis involves a unique series of genetic and epigenetic alterations that are distinct from those in the traditional adenoma carcinoma sequence (2, 3). There are at least two different pathways by which serrated lesions can develop into cancer, involving activating mutations in RAS or RAF kinase genes, which can result in the CpG island methylation phenotype (CIMP), leading to inactivation of certain tumor suppressor genes (e.g., MLH1, CDKN2A) via promoter hypermethylation. These changes can lead to either microsatellite unstable (MSI) or stable (MSS) colorectal cancer (3). Apart from differences in molecular features and histology, serrated lesions also have a different endoscopic appearance, distribution in the colon, growth characteristics, and epidemiology compared with conventional adenomas.

Understanding who is at risk for colorectal cancer and the different types of precursor lesions is important to be able to optimally prevent this deadly disease. Although the epidemiology of conventional adenomas is fairly well elucidated, serrated lesions are comparatively less well studied with respect to both modifiable and nonmodifiable risk factors, preventive influences, and other important associations. In particular, the field lacks longitudinal studies identifying factors that have a clear temporal or causal relationship with serrated lesion outcomes. In addition, there are few published epidemiologic studies examining HP subtypes. Such data are scientifically valuable because some authors suggest that microvesicular (MVHP) and goblet cell-rich (GCHP) HPs could give rise to premalignant SSLs and TSAs (5, 6).

We used data from a large colonoscopy based chemoprevention trial to study the association between baseline participant characteristics and risk of developing serrated lesions, particularly SSLs, during follow up. Because all polyps were removed at baseline, this study is designed to evaluate risk factors for “incident” serrated lesions.

Study design and participants

This study used data on baseline participant characteristics and serrated colorectal polyps outcomes among participants in the Vitamin D/Calcium Polyp Prevention Study (VCPPS). The VCPPS parent study was a randomized, multicenter, participant, and investigator blinded, placebo-controlled chemoprevention trial that took place at 11 geographically diverse centers across the United States. Participants were enrolled in the trial between July 2004 and July 2008, and colonoscopy outcomes were collected up to June 2016. Detailed methods from this trial have been published previously, including the protocol (7). In brief, individuals ages 45 to 75 were invited to participate if they had undergone a clearing colonoscopy with at least one adenomatous polyp (≥2 mm) detected and removed in the preceding 4 months. All participants were scheduled to undergo surveillance colonoscopy at either 3 or 5 years based on the findings of the index examination. Patients with inflammatory bowel disease, kidney dysfunction, known familial colorectal cancer syndromes, or contraindications to receiving the study treatments were excluded.

Following enrollment, participants underwent a run-in period to confirm minimum pill taking adherence of 80% over at least 2 months. Eligible participants were then randomized to receive calcium carbonate (1,200 mg elemental calcium per day), vitamin D3 (1,000 IU/day), the combination of these agents, or neither agent (placebo). However, eligible women were allowed to choose to receive calcium in their study pills and to be randomized only to vitamin D or placebo. The treatment period continued until the first surveillance colonoscopy at 3 or 5 years. For participants who consented to additional follow-up beyond the treatment phase, data collection continued after the first surveillance colonoscopy (observational period). For this analysis, serrated polyp outcomes were included from any follow-up colonoscopy occurring at least 1-year post-randomization in either the treatment or observational phase. The majority of participants had outcomes determined from a single colonoscopy in both phases.

The VCPPS study was conducted in accordance with recognized ethical guidelines (Declaration of Helsinki) and was approved by the institutional review boards of the central coordinating site (Dartmouth) and all participating clinical sites. All participants provided written informed consent prior to enrollment. The trial was registered at ClinicalTrials.gov (no. NCT00153816).

Measurement of outcomes, exposures, and covariates

At enrollment, trained research staff collected baseline data in a standardized fashion, including demographics, medical history, family history, concomitant medications, health habits, and diet (Block Brief 2000 questionnaire). We collected and systematically abstracted all baseline colonoscopy reports and pathology reports to record data on all polyps found on the baseline (pre-enrollment) exam. The diagnosis for all baseline polyps, adenoma or serrated, was determined per local diagnosis. Height and weight were determined by self-report or measurement, and used to calculate body mass index (BMI). Throughout the trial, patients were contacted by a study coordinator periodically to verify adherence, adverse events, concomitant medications, and receipt of colonoscopy or other colorectal imaging.

All polyps found during post-randomization colonoscopies were reviewed centrally by an expert gastrointestinal pathologist. For this study, the outcomes of interest included all SPs, subtyped as HPs (MVHP and GCHP), SSLs (with or without cytologic dysplasia), and TSAs, classified according to the 2010 WHO criteria (8). Polyp location was categorized as right-sided if proximal to the splenic flexure, and lesions more distal were categorized as left-sided.

Statistical analysis

The primary outcomes for this post hoc analysis of the VCPPS study were one or more serrated polyps (any type) overall, as well as SSLs and HPs specifically. We were unable to assess TSAs as an outcome in modeling due to their rarity. Multivariable generalized estimating equation (GEE) models were used to estimate risk ratios (RR) and 95% confidence intervals (CI) for different serrated polyp types. Separate models were run for each exposure of interest, which included age, sex, race, education, BMI, physical activity level, smoking status and duration, baseline alcohol use, aspirin or NSAID exposure, and baseline polyp findings (categories and referent groups indicated in tables). Models were adjusted for several covariates, including randomization variables (age, sex, race (white, black, other), center [grouped geographically into southeast (Georgia, North Carolina, South Carolina, and Puerto Rico), north (Ohio, New Hampshire, Iowa, and Minnesota), and west (Colorado, Texas, and California)], vitamin D study treatment (placebo, active), and calcium study treatment (placebo, active—this includes women who elected to receive calcium and were not randomized to it), and potential confounding factors (study phase of the outcome (treatment or observational), and either number of serrated polyps at baseline for any serrated polyp and HP outcomes, or baseline SSL or proximal serrated polyps for SSL outcome. P for trend was calculated using a continuous ordinal variable for the categories of the exposure variable.

Two-sided P values of <0.05 were considered statistically significant. Statistical analyses were conducted using SAS software, version 9.4 (SAS Institute).

Data availability

The data generated in this study are available upon request from the corresponding author.

Participants and baseline characteristics

A total of 2,813 participants were enrolled and 2,259 underwent randomization. Among these, 2,102 participants had a colonoscopy at least 1 year after randomization and had sufficient pathology data to determine polyp outcomes during the initial treatment phase; these individuals were included in the analysis presented here. Because of cessation of funding before all subjects reached their second surveillance colonoscopy, only approximately half of the participants (n = 1,127) had at least one colonoscopy with complete data from the observational phase (Supplementary Fig. S1).

The majority of included participants were male (64%) and white race (89%; Table 1). Most participants were ages 55 or older (65%). At baseline, most participants had one or two low-risk tubular adenomas, but 384 (19%) had advanced adenomas. There were 502 participants who had synchronous serrated polyps at baseline including 335 (17%) with a single serrated polyp and 167 (8%) with 2 or more. Most baseline serrated polyps were HPs, but 155 (8%) of participants had either an SSL or proximal serrated polyp at baseline.

Table 1.

Characteristics for all included subjects.

Characteristicsn (%)
Total participants 2,102 (100) 
Age 
 <55 743 (35.4) 
 55–59 542 (25.8) 
 60–64 398 (18.9) 
 ≥65 419 (19.9) 
Sex 
 Male 1,334 (63.5) 
 Female 768 (36.5) 
Race 
 White 1,788 (88.7) 
 Black 160 (7.9) 
 Other 69 (3.4) 
Education 
 <High school graduate 105 (5.0) 
 High school graduate 284 (13.6) 
 Some college 1,205 (57.6) 
 Graduate school 497 (23.8) 
Body mass index 
 <25 kg/m2 487 (23.2) 
 25–29.9 kg/m2 857 (40.8) 
 ≥30 kg/m2 757 (36.0) 
Activity level 
 Low 497 (23.9) 
 Medium 664 (32.0) 
 High 916 (44.1) 
Smoking status 
 Never 1,122 (53.4) 
 Former 783 (37.3) 
 Current 197 (9.4) 
Pack years 
 0 1,122 (53.5) 
 >0 to 13 310 (14.8) 
 >13 to 34 333 (15.9) 
 >34 331 (15.8) 
Maximum amount smoked for 1+ year 
 0 1,122 (53.4) 
 >0 to <1 pack 292 (13.9) 
 1 pack 366 (17.4) 
 > 1 pack 321 (15.3) 
Duration smoked 
 0 1,122 (53.5) 
 >0 to 15 years 299 (14.3) 
 16–34 years 444 (21.2) 
 >34 years 232 (11.1) 
Baseline alcohol use 
 None 639 (32.7) 
 0.1–1 drink/day 793 (40.5) 
 >1 drink/day 524 (26.8) 
Baseline aspirin use n (%) 
 None 932 (44.3) 
 <1 day/week 301 (14.3) 
 1–6 days/week 178 (8.5) 
 7 days/week 691 (32.9) 
Baseline non-aspirin NSAID use 
 None 796 (37.9) 
 <1 day/week 787 (37.4) 
 1–6 days/week 361 (17.2) 
 7 days/week 158 (7.5) 
Baseline advanced adenoma 
 No 1,682 (81.4) 
 Yes 384 (18.6) 
Baseline serrated polyp 
 0 1,524 (75.2) 
 1 335 (16.5) 
 2+ 167 (8.2) 
Baseline SSL or proximal serrated polyp 
 No 1,821 (92.2) 
 Yes 155 (7.8) 
Menopausal status 
 Pre 155 (20.5) 
 Post 603 (79.6) 
Baseline HRT status 
 Never 407 (53.5) 
 Former 242 (31.8) 
 Current 112 (14.7) 
Ever used contraceptives 
 No 133 (17.4) 
 Yes 633 (82.6) 
Age at menarche 
 ≤11 142 (18.8) 
 12 193 (25.5) 
 13 249 (32.9) 
 ≥14 172 (22.8) 
Number of pregnancies 
 0 79 (10.3) 
 1 84 (11.0) 
 2 204 (26.6) 
 3 206 (26.9) 
 ≥4 194 (25.3) 
Number of live births 
 0 114 (14.9) 
 1 106 (13.8) 
 2 273 (35.6) 
 3 169 (22.0) 
 ≥4 105 (13.7) 
Characteristicsn (%)
Total participants 2,102 (100) 
Age 
 <55 743 (35.4) 
 55–59 542 (25.8) 
 60–64 398 (18.9) 
 ≥65 419 (19.9) 
Sex 
 Male 1,334 (63.5) 
 Female 768 (36.5) 
Race 
 White 1,788 (88.7) 
 Black 160 (7.9) 
 Other 69 (3.4) 
Education 
 <High school graduate 105 (5.0) 
 High school graduate 284 (13.6) 
 Some college 1,205 (57.6) 
 Graduate school 497 (23.8) 
Body mass index 
 <25 kg/m2 487 (23.2) 
 25–29.9 kg/m2 857 (40.8) 
 ≥30 kg/m2 757 (36.0) 
Activity level 
 Low 497 (23.9) 
 Medium 664 (32.0) 
 High 916 (44.1) 
Smoking status 
 Never 1,122 (53.4) 
 Former 783 (37.3) 
 Current 197 (9.4) 
Pack years 
 0 1,122 (53.5) 
 >0 to 13 310 (14.8) 
 >13 to 34 333 (15.9) 
 >34 331 (15.8) 
Maximum amount smoked for 1+ year 
 0 1,122 (53.4) 
 >0 to <1 pack 292 (13.9) 
 1 pack 366 (17.4) 
 > 1 pack 321 (15.3) 
Duration smoked 
 0 1,122 (53.5) 
 >0 to 15 years 299 (14.3) 
 16–34 years 444 (21.2) 
 >34 years 232 (11.1) 
Baseline alcohol use 
 None 639 (32.7) 
 0.1–1 drink/day 793 (40.5) 
 >1 drink/day 524 (26.8) 
Baseline aspirin use n (%) 
 None 932 (44.3) 
 <1 day/week 301 (14.3) 
 1–6 days/week 178 (8.5) 
 7 days/week 691 (32.9) 
Baseline non-aspirin NSAID use 
 None 796 (37.9) 
 <1 day/week 787 (37.4) 
 1–6 days/week 361 (17.2) 
 7 days/week 158 (7.5) 
Baseline advanced adenoma 
 No 1,682 (81.4) 
 Yes 384 (18.6) 
Baseline serrated polyp 
 0 1,524 (75.2) 
 1 335 (16.5) 
 2+ 167 (8.2) 
Baseline SSL or proximal serrated polyp 
 No 1,821 (92.2) 
 Yes 155 (7.8) 
Menopausal status 
 Pre 155 (20.5) 
 Post 603 (79.6) 
Baseline HRT status 
 Never 407 (53.5) 
 Former 242 (31.8) 
 Current 112 (14.7) 
Ever used contraceptives 
 No 133 (17.4) 
 Yes 633 (82.6) 
Age at menarche 
 ≤11 142 (18.8) 
 12 193 (25.5) 
 13 249 (32.9) 
 ≥14 172 (22.8) 
Number of pregnancies 
 0 79 (10.3) 
 1 84 (11.0) 
 2 204 (26.6) 
 3 206 (26.9) 
 ≥4 194 (25.3) 
Number of live births 
 0 114 (14.9) 
 1 106 (13.8) 
 2 273 (35.6) 
 3 169 (22.0) 
 ≥4 105 (13.7) 

Note: Ns for a variable may not sum to the total due to missing data. Percentages are based on nonmissing data. Data for menopausal status, HRT and contraceptive use, menarche, pregnancies, and births limited to female participants only.

Abbreviation: HRT, hormone replacement therapy.

Occurrence of serrated polyps

A total of 1,040 serrated polyps (including 132 SSLs) were found during the treatment phase, and an additional 575 serrated polyps (including 80 SSLs) were found during the observational phase. On the participant level, 565 (27%) participants had at least one serrated polyp in the treatment phase, including 494 (24%) with HPs, 100 (5%) with SSLs, and 8 (0.4%) with TSAs (Table 2). During the observational phase, 327 (30%) participants had serrated polyps during this period, including 269 (25%) with HPs, 62 (6%) with SSLs, and 6 (0.6%) with TSAs. 11 of 62 (18%) observational phase participants with SSLs also had a prior SSL during the treatment phase. Most SSLs (67%) were located in the proximal colon.

Table 2.

Number of participants with each outcome by study phase.

Treatment phaseObservational phase
n with outcome/Na (%)n with outcome/Na (%)
Polyp outcomesN with exam = 2,102N with exam = 1,127
Any serrated polyp 565/2,058 (27.5) 327/1,100 (29.7) 
Sessile serrated lesion 100/2,014 (5.0) 62/1,078 (5.8) 
 >1 SSL 24/2,012 (1.2) 11/1,075 (1.0) 
 SSL ≥ 1 cm 17/2,013 (0.8) 15/1,075 (1.4) 
 SSL with dysplasia 4/2,013 (0.2) 3/1,077 (0.3) 
Traditional serrated adenoma 8/2,013 (0.4) 6/1,076 (0.6) 
Any hyperplastic polyp 494/2,056 (24.0) 269/1,095 (24.6) 
 Microvesicular HP 304/2,038 (14.9) 153/1,087 (14.1) 
 Goblet cell–rich HP 215/2,033 (10.6) 115/1,084 (10.6) 
 HP not subclassified 65/2,021 (3.2) 43/1,079 (4.0) 
Treatment phaseObservational phase
n with outcome/Na (%)n with outcome/Na (%)
Polyp outcomesN with exam = 2,102N with exam = 1,127
Any serrated polyp 565/2,058 (27.5) 327/1,100 (29.7) 
Sessile serrated lesion 100/2,014 (5.0) 62/1,078 (5.8) 
 >1 SSL 24/2,012 (1.2) 11/1,075 (1.0) 
 SSL ≥ 1 cm 17/2,013 (0.8) 15/1,075 (1.4) 
 SSL with dysplasia 4/2,013 (0.2) 3/1,077 (0.3) 
Traditional serrated adenoma 8/2,013 (0.4) 6/1,076 (0.6) 
Any hyperplastic polyp 494/2,056 (24.0) 269/1,095 (24.6) 
 Microvesicular HP 304/2,038 (14.9) 153/1,087 (14.1) 
 Goblet cell–rich HP 215/2,033 (10.6) 115/1,084 (10.6) 
 HP not subclassified 65/2,021 (3.2) 43/1,079 (4.0) 

aN = number with exam and sufficient pathology to determine outcome status.

Risk factor associations

Table 3 presents results for associations of baseline participant characteristics with various serrated polyp outcomes. No apparent association was seen between age or sex and risk of serrated polyps. However, non-white participants, specifically those who identified as black, were at lower risk of SSLs and MVHPs [RR (95% CI) compared with white individuals: 0.40 (0.16–0.99) and 0.63 (0.42–0.96), respectively]. In contrast, black participants appeared to be at increased risk of GCHPs compared with white participants [RR, 1.83; 95% CI, 1.23–2.72; Fig. 1A). In this analysis, we found no association between physical activity, presence of advanced adenoma at baseline, use of NSAIDs or aspirin and serrated polyp outcomes. We also did not identify any meaningful associations between menopausal status, use of hormone replacement, or use of contraceptives and occurrence of serrated polyps overall (Table 4). However, an increasing number of pregnancies and live births were associated with a higher risk of HPs. For example, women with four or more pregnancies had roughly twice the risk of HPs compared with nulliparous women (RR, 2.24; 95% CI, 1.23–4.08; Ptrend = 0.05). A similar pattern was observed for four or more live births versus none. However, there was no clear association between number of pregnancies or births and risk of SSLs.

Table 3.

Association of selected personal characteristics and serrated polyp outcomes.

Any serrated polypSessile serrated lesionAny hyperplastic polypMicrovesicular HPGoblet cell–rich HP
CharacteristicRR (95% CI)RR (95% CI)RR (95% CI)RR (95% CI)RR (95% CI)
Age 
 <55 Reference Reference Reference Reference Reference 
 55–59 1.11 (0.89–1.40) 0.94 (0.59–1.50) 1.10 (0.87–1.40) 1.33 (1.02–1.75) 0.80 (0.57–1.12) 
 60–64 1.18 (0.92–1.51) 1.48 (0.95–2.32) 1.13 (0.87–1.47) 1.25 (0.91–1.73) 0.89 (0.63–1.26) 
 ≥65 1.17 (0.91–1.50) 1.29 (0.81–2.07) 1.05 (0.81–1.37) 1.08 (0.79–1.47) 1.04 (0.73–1.48) 
Ptrend 0.16 0.11 0.57 0.50 0.91 
Sex 
 Male Reference Reference Reference Reference Reference 
 Female 1.02 (0.84–1.24) 0.86 (0.59–1.26) 1.01 (0.83–1.25) 1.03 (0.80–1.31) 0.95 (0.71–1.27) 
Race 
 White Reference Reference Reference Reference Reference 
 Black 0.88 (0.64–1.22) 0.40 (0.16–0.99) 1.08 (0.78–1.49) 0.63 (0.42–0.96) 1.83 (1.23–2.72) 
 Other 1.06 (0.67–1.66) 0.19 (0.03–1.38) 1.30 (0.82–2.05) 0.57 (0.29–1.14) 1.38 (0.70–2.71) 
Education 
 <High school graduate 1.20 (0.68–2.11) 2.32 (0.90–5.98) 1.14 (0.63–2.03) 1.65 (0.85–3.21) 0.64 (0.27–1.57) 
 High school graduate Reference Reference Reference Reference Reference 
 Some college 0.92 (0.70–1.21) 1.32 (0.78–2.25) 0.92 (0.69–1.21) 1.08 (0.77–1.50) 0.69 (0.49–0.98) 
 Graduate school 0.91 (0.67–1.24) 1.49 (0.82–2.70) 0.82 (0.60–1.13) 0.97 (0.67–1.42) 0.62 (0.41–0.94) 
Ptrend 0.35 0.72 0.13 0.28 0.10 
Body mass index 
 <25 kg/m2 Reference Reference Reference Reference Reference 
 25–29.9 kg/m2 0.97 (0.77–1.23) 0.87 (0.56–1.34) 1.00 (0.78–1.29) 0.86 (0.64–1.15) 1.43 (1.00–2.03) 
 ≥30 kg/m2 1.19 (0.94–1.51) 0.75 (0.48–1.19) 1.31 (1.03–1.68) 1.15 (0.87–1.54) 1.54 (1.07–2.21) 
Ptrend 0.08 0.22 0.01 0.15 0.03 
Physical activity level 
 Low Reference Reference Reference Reference Reference 
 Medium 0.93 (0.73–1.18) 0.73 (0.47–1.14) 1.11 (0.86–1.43) 1.11 (0.82–1.49) 0.93 (0.66–1.30) 
 High 0.94 (0.75–1.18) 0.75 (0.49–1.14) 1.05 (0.83–1.34) 1.09 (0.82–1.45) 0.92 (0.67–1.26) 
Ptrend 0.65 0.23 0.79 0.60 0.61 
Smoking status 
 Never Reference Reference Reference Reference Reference 
 Former 1.60 (1.32–1.95) 1.01 (0.69–1.48) 1.71 (1.39–2.10) 1.77 (1.38–2.26) 1.77 (1.33–2.36) 
 Current 1.74 (1.28–2.35) 1.70 (1.00–2.86) 1.86 (1.37–2.52) 1.58 (1.10–2.28) 2.36 (1.59–3.52) 
Ptrend <0.0001 0.15 <0.0001 <0.0001 <0.0001 
Pack years 
 0 Reference Reference Reference Reference Reference 
 >0 to 13 1.51 (1.17–1.95) 1.06 (0.64–1.75) 1.57 (1.20–2.05) 1.75 (1.29–2.37) 1.56 (1.06–2.29) 
 >13 to 34 1.49 (1.16–1.92) 1.54 (0.98–2.42) 1.49 (1.14–1.95) 1.48 (1.08–2.03) 1.68 (1.16–2.42) 
 >34 1.91 (1.48–2.45) 0.88 (0.52–1.50) 2.21 (1.70–2.87) 1.94 (1.42–2.64) 2.63 (1.88–3.68) 
Ptrend <0.0001 0.70 <0.0001 <0.0001 <0.0001 
Maximum amount smoked for 1+ year 
 0 Reference Reference Reference Reference Reference 
 >0 to < 1 pack 1.41 (1.08–1.84) 1.39 (0.84–2.29) 1.37 (1.04–1.81) 1.31 (0.95–1.81) 1.93 (1.33–2.80) 
 1 pack 1.71 (1.35–2.16) 1.11 (0.68–1.80) 1.84 (1.44–2.35) 1.89 (1.42–2.52) 1.75 (1.24–2.47) 
 >1 pack 1.73 (1.33–2.25) 1.00 (0.61–1.63) 1.98 (1.51–2.60) 1.89 (1.37–2.62) 2.07 (1.44–2.96) 
Ptrend <0.0001 0.84 <0.0001 <0.0001 <0.0001 
Duration smoked 
 0 Reference Reference Reference Reference Reference 
 >0 to 15 years 1.51 (1.16–1.97) 1.17 (0.72–1.89) 1.60 (1.22–2.11) 1.76 (1.29–2.40) 1.40 (0.93–2.11) 
 16–34 years 1.56 (1.24–1.96) 1.08 (0.68–1.72) 1.67 (1.31–2.12) 1.68 (1.26–2.23) 1.90 (1.38–2.61) 
 >34 years 1.98 (1.49–2.63) 1.25 (0.73–2.14) 2.10 (1.57–2.82) 1.78 (1.26–2.51) 2.76 (1.89–4.02) 
Ptrend <0.0001 0.44 <0.0001 <0.0001 <0.0001 
Baseline alcohol use 
 None Reference Reference Reference Reference Reference 
 0.1–1 drink/day 1.09 (0.88–1.34) 1.20 (0.79–1.81) 1.06 (0.85–1.33) 1.29 (0.98–1.70) 0.94 (0.68–1.29) 
 >1 drink/day 1.17 (0.92–1.50) 1.05 (0.65–1.70) 1.30 (1.01–1.69) 1.61 (1.18–2.18) 1.10 (0.77–1.57) 
Ptrend 0.24 0.89 0.03 0.005 0.44 
Baseline aspirin use 
 None Reference Reference Reference Reference Reference 
 <1 day/week 0.90 (0.68–1.19) 1.37 (0.82–2.27) 0.84 (0.62–1.13) 0.73 (0.50–1.06) 0.79 (0.52–1.21) 
 1–6 days/week 1.04 (0.75–1.45) 1.00 (0.50–1.99) 1.06 (0.74–1.50) 1.08 (0.72–1.62) 0.82 (0.50–1.33) 
 7 days/week 1.21 (0.99–1.49) 1.41 (0.93–2.13) 1.15 (0.93–1.43) 1.09 (0.85–1.39) 1.28 (0.95–1.72) 
Ptrend 0.06 0.14 0.16 0.40 0.12 
Baseline non-aspirin NSAID use 
 None Reference Reference Reference Reference Reference 
 <1 day/week 1.12 (0.91–1.37) 0.91 (0.61–1.34) 1.14 (0.92–1.42) 1.15 (0.89–1.49) 1.21 (0.90–1.63) 
 1–6 days/week 1.30 (1.00–1.68) 1.14 (0.71–1.84) 1.33 (1.01–1.75) 1.44 (1.05–1.99) 1.17 (0.81–1.71) 
 7 days/week 0.89 (0.62–1.27) 0.48 (0.20–1.13) 1.03 (0.71–1.49) 1.06 (0.68–1.64) 1.01 (0.61–1.68) 
Ptrend 0.46 0.43 0.18 0.13 0.59 
Baseline advanced adenoma 
 No Reference Reference Reference Reference Reference 
 Yes 1.03 (0.82–1.28) 1.49 (1.01–2.20) 0.96 (0.76–1.21) 1.16 (0.89–1.51) 0.69 (0.49–0.98) 
Baseline serrated polypa 
 0 Reference Reference Reference Reference Reference 
 1 1.91 (1.53–2.39) 1.94 (1.26–2.99) 1.81 (1.43–2.29) 1.78 (1.35–2.34) 1.77 (1.27–2.46) 
 2+ 4.47 (3.34–6.00) 3.32 (2.08–5.30) 4.16 (3.10–5.58) 4.10 (3.02–5.56) 3.92 (2.77–5.55) 
Ptrend <0.0001 <0.0001 <0.0001 <0.0001 <0.0001 
Baseline SSL or PSPb 
 No Reference Reference Reference Reference Reference 
 Yes 2.36 (1.75–3.18) 3.11 (1.97–4.93) 1.84 (1.35–2.52) 2.02 (1.43–2.85) 1.74 (1.15–2.64) 
Any serrated polypSessile serrated lesionAny hyperplastic polypMicrovesicular HPGoblet cell–rich HP
CharacteristicRR (95% CI)RR (95% CI)RR (95% CI)RR (95% CI)RR (95% CI)
Age 
 <55 Reference Reference Reference Reference Reference 
 55–59 1.11 (0.89–1.40) 0.94 (0.59–1.50) 1.10 (0.87–1.40) 1.33 (1.02–1.75) 0.80 (0.57–1.12) 
 60–64 1.18 (0.92–1.51) 1.48 (0.95–2.32) 1.13 (0.87–1.47) 1.25 (0.91–1.73) 0.89 (0.63–1.26) 
 ≥65 1.17 (0.91–1.50) 1.29 (0.81–2.07) 1.05 (0.81–1.37) 1.08 (0.79–1.47) 1.04 (0.73–1.48) 
Ptrend 0.16 0.11 0.57 0.50 0.91 
Sex 
 Male Reference Reference Reference Reference Reference 
 Female 1.02 (0.84–1.24) 0.86 (0.59–1.26) 1.01 (0.83–1.25) 1.03 (0.80–1.31) 0.95 (0.71–1.27) 
Race 
 White Reference Reference Reference Reference Reference 
 Black 0.88 (0.64–1.22) 0.40 (0.16–0.99) 1.08 (0.78–1.49) 0.63 (0.42–0.96) 1.83 (1.23–2.72) 
 Other 1.06 (0.67–1.66) 0.19 (0.03–1.38) 1.30 (0.82–2.05) 0.57 (0.29–1.14) 1.38 (0.70–2.71) 
Education 
 <High school graduate 1.20 (0.68–2.11) 2.32 (0.90–5.98) 1.14 (0.63–2.03) 1.65 (0.85–3.21) 0.64 (0.27–1.57) 
 High school graduate Reference Reference Reference Reference Reference 
 Some college 0.92 (0.70–1.21) 1.32 (0.78–2.25) 0.92 (0.69–1.21) 1.08 (0.77–1.50) 0.69 (0.49–0.98) 
 Graduate school 0.91 (0.67–1.24) 1.49 (0.82–2.70) 0.82 (0.60–1.13) 0.97 (0.67–1.42) 0.62 (0.41–0.94) 
Ptrend 0.35 0.72 0.13 0.28 0.10 
Body mass index 
 <25 kg/m2 Reference Reference Reference Reference Reference 
 25–29.9 kg/m2 0.97 (0.77–1.23) 0.87 (0.56–1.34) 1.00 (0.78–1.29) 0.86 (0.64–1.15) 1.43 (1.00–2.03) 
 ≥30 kg/m2 1.19 (0.94–1.51) 0.75 (0.48–1.19) 1.31 (1.03–1.68) 1.15 (0.87–1.54) 1.54 (1.07–2.21) 
Ptrend 0.08 0.22 0.01 0.15 0.03 
Physical activity level 
 Low Reference Reference Reference Reference Reference 
 Medium 0.93 (0.73–1.18) 0.73 (0.47–1.14) 1.11 (0.86–1.43) 1.11 (0.82–1.49) 0.93 (0.66–1.30) 
 High 0.94 (0.75–1.18) 0.75 (0.49–1.14) 1.05 (0.83–1.34) 1.09 (0.82–1.45) 0.92 (0.67–1.26) 
Ptrend 0.65 0.23 0.79 0.60 0.61 
Smoking status 
 Never Reference Reference Reference Reference Reference 
 Former 1.60 (1.32–1.95) 1.01 (0.69–1.48) 1.71 (1.39–2.10) 1.77 (1.38–2.26) 1.77 (1.33–2.36) 
 Current 1.74 (1.28–2.35) 1.70 (1.00–2.86) 1.86 (1.37–2.52) 1.58 (1.10–2.28) 2.36 (1.59–3.52) 
Ptrend <0.0001 0.15 <0.0001 <0.0001 <0.0001 
Pack years 
 0 Reference Reference Reference Reference Reference 
 >0 to 13 1.51 (1.17–1.95) 1.06 (0.64–1.75) 1.57 (1.20–2.05) 1.75 (1.29–2.37) 1.56 (1.06–2.29) 
 >13 to 34 1.49 (1.16–1.92) 1.54 (0.98–2.42) 1.49 (1.14–1.95) 1.48 (1.08–2.03) 1.68 (1.16–2.42) 
 >34 1.91 (1.48–2.45) 0.88 (0.52–1.50) 2.21 (1.70–2.87) 1.94 (1.42–2.64) 2.63 (1.88–3.68) 
Ptrend <0.0001 0.70 <0.0001 <0.0001 <0.0001 
Maximum amount smoked for 1+ year 
 0 Reference Reference Reference Reference Reference 
 >0 to < 1 pack 1.41 (1.08–1.84) 1.39 (0.84–2.29) 1.37 (1.04–1.81) 1.31 (0.95–1.81) 1.93 (1.33–2.80) 
 1 pack 1.71 (1.35–2.16) 1.11 (0.68–1.80) 1.84 (1.44–2.35) 1.89 (1.42–2.52) 1.75 (1.24–2.47) 
 >1 pack 1.73 (1.33–2.25) 1.00 (0.61–1.63) 1.98 (1.51–2.60) 1.89 (1.37–2.62) 2.07 (1.44–2.96) 
Ptrend <0.0001 0.84 <0.0001 <0.0001 <0.0001 
Duration smoked 
 0 Reference Reference Reference Reference Reference 
 >0 to 15 years 1.51 (1.16–1.97) 1.17 (0.72–1.89) 1.60 (1.22–2.11) 1.76 (1.29–2.40) 1.40 (0.93–2.11) 
 16–34 years 1.56 (1.24–1.96) 1.08 (0.68–1.72) 1.67 (1.31–2.12) 1.68 (1.26–2.23) 1.90 (1.38–2.61) 
 >34 years 1.98 (1.49–2.63) 1.25 (0.73–2.14) 2.10 (1.57–2.82) 1.78 (1.26–2.51) 2.76 (1.89–4.02) 
Ptrend <0.0001 0.44 <0.0001 <0.0001 <0.0001 
Baseline alcohol use 
 None Reference Reference Reference Reference Reference 
 0.1–1 drink/day 1.09 (0.88–1.34) 1.20 (0.79–1.81) 1.06 (0.85–1.33) 1.29 (0.98–1.70) 0.94 (0.68–1.29) 
 >1 drink/day 1.17 (0.92–1.50) 1.05 (0.65–1.70) 1.30 (1.01–1.69) 1.61 (1.18–2.18) 1.10 (0.77–1.57) 
Ptrend 0.24 0.89 0.03 0.005 0.44 
Baseline aspirin use 
 None Reference Reference Reference Reference Reference 
 <1 day/week 0.90 (0.68–1.19) 1.37 (0.82–2.27) 0.84 (0.62–1.13) 0.73 (0.50–1.06) 0.79 (0.52–1.21) 
 1–6 days/week 1.04 (0.75–1.45) 1.00 (0.50–1.99) 1.06 (0.74–1.50) 1.08 (0.72–1.62) 0.82 (0.50–1.33) 
 7 days/week 1.21 (0.99–1.49) 1.41 (0.93–2.13) 1.15 (0.93–1.43) 1.09 (0.85–1.39) 1.28 (0.95–1.72) 
Ptrend 0.06 0.14 0.16 0.40 0.12 
Baseline non-aspirin NSAID use 
 None Reference Reference Reference Reference Reference 
 <1 day/week 1.12 (0.91–1.37) 0.91 (0.61–1.34) 1.14 (0.92–1.42) 1.15 (0.89–1.49) 1.21 (0.90–1.63) 
 1–6 days/week 1.30 (1.00–1.68) 1.14 (0.71–1.84) 1.33 (1.01–1.75) 1.44 (1.05–1.99) 1.17 (0.81–1.71) 
 7 days/week 0.89 (0.62–1.27) 0.48 (0.20–1.13) 1.03 (0.71–1.49) 1.06 (0.68–1.64) 1.01 (0.61–1.68) 
Ptrend 0.46 0.43 0.18 0.13 0.59 
Baseline advanced adenoma 
 No Reference Reference Reference Reference Reference 
 Yes 1.03 (0.82–1.28) 1.49 (1.01–2.20) 0.96 (0.76–1.21) 1.16 (0.89–1.51) 0.69 (0.49–0.98) 
Baseline serrated polypa 
 0 Reference Reference Reference Reference Reference 
 1 1.91 (1.53–2.39) 1.94 (1.26–2.99) 1.81 (1.43–2.29) 1.78 (1.35–2.34) 1.77 (1.27–2.46) 
 2+ 4.47 (3.34–6.00) 3.32 (2.08–5.30) 4.16 (3.10–5.58) 4.10 (3.02–5.56) 3.92 (2.77–5.55) 
Ptrend <0.0001 <0.0001 <0.0001 <0.0001 <0.0001 
Baseline SSL or PSPb 
 No Reference Reference Reference Reference Reference 
 Yes 2.36 (1.75–3.18) 3.11 (1.97–4.93) 1.84 (1.35–2.52) 2.02 (1.43–2.85) 1.74 (1.15–2.64) 

Note: Each serrated polyp phenotype was investigated in a separate GEE model, adjusted for age, sex, center [grouped geographically into southeast (Georgia, North Carolina, South Carolina, and Puerto Rico), north (Ohio, New Hampshire, Iowa, and Minnesota), and west (Colorado, Texas, and California)], race (white, black, other), study phase (treatment, observational), vitamin D study treatment (placebo, active), calcium study treatment (placebo, active—this includes women who elected to receive calcium and were not randomized to it), and either number of serrated polyps at baseline (0, 1, 2+) for any serrated polyp and hyperplastic polyp outcomes or baseline SSL/right-sided serrated polyp (no, yes) for SSL outcome.

Abbreviations: PSP, proximal serrated polyp; SSL, sessile serrated lesion.

aNot adjusted for number of serrated polyps at baseline.

bNot adjusted for SSL or proximal serrated polyps at baseline.

Figure 1.

Risk of different serrated polyp subtypes by race (A), presence of sessile serrated lesions or proximal serrated polyps on baseline exam (B), and smoking status (C). GCHP, goblet cell hyperplastic polyp; MVHP, microvesicular hyperplastic polyp; REF, referent category; SSL, sessile serrated lesion.

Figure 1.

Risk of different serrated polyp subtypes by race (A), presence of sessile serrated lesions or proximal serrated polyps on baseline exam (B), and smoking status (C). GCHP, goblet cell hyperplastic polyp; MVHP, microvesicular hyperplastic polyp; REF, referent category; SSL, sessile serrated lesion.

Close modal
Table 4.

Association of selected reproductive variables and serrated polyp outcomes.

Any serrated polypSessile serrated lesionAny hyperplastic polypMicrovesicular HPGoblet cell–rich HP
CharacteristicRR (95% CI)RR (95% CI)RR (95% CI)RR (95% CI)RR (95% CI)
Menopausal status 
 Pre Reference Reference Reference Reference Reference 
 Post 1.01 (0.68–1.48) 1.32 (0.61–2.85) 0.89 (0.60–1.33) 1.07 (0.66–1.74) 0.67 (0.38–1.16) 
Baseline HRT status 
 Never Reference Reference Reference Reference Reference 
 Former 1.05 (0.76–1.47) 2.04 (1.11–3.75) 0.92 (0.65–1.31) 1.06 (0.70–1.59) 0.64 (0.38–1.07) 
 Current 0.69 (0.45–1.05) 0.44 (0.13–1.53) 0.66 (0.42–1.03) 0.63 (0.36–1.11) 0.66 (0.34–1.29) 
Ptrend 0.13 0.89 0.07 0.12 0.15 
Ever used contraceptives 
 No Reference Reference Reference Reference Reference 
 Yes 0.97 (0.66–1.42) 2.12 (0.84–5.36) 0.79 (0.53–1.18) 1.04 (0.64–1.71) 0.72 (0.41–1.27) 
Age at menarche 
 11 Reference Reference Reference Reference Reference 
 12 1.10 (0.72–1.69) 1.92 (0.77–4.76) 1.09 (0.69–1.72) 1.27 (0.74–2.20) 0.91 (0.47–1.74) 
 13 1.12 (0.75–1.67) 1.51 (0.61–3.77) 1.04 (0.68–1.61) 1.35 (0.80–2.29) 1.13 (0.63–2.02) 
 ≥14 0.74 (0.45–1.19) 0.78 (0.25–2.40) 0.77 (0.46–1.29) 0.89 (0.47–1.70) 0.88 (0.44–1.76) 
Ptrend 0.29 0.61 0.33 0.89 0.95 
Number of pregnancies 
 0 Reference Reference Reference Reference Reference 
 1 1.30 (0.68–2.46) 0.65 (0.18–2.33) 1.81 (0.92–3.58) 1.08 (0.46–2.54) 3.44 (1.20–9.86) 
 2 1.48 (0.85–2.58) 0.98 (0.35–2.73) 1.92 (1.06–3.48) 1.60 (0.82–3.13) 2.52 (0.94–6.78) 
 3 1.33 (0.76–2.31) 0.85 (0.30–2.42) 1.63 (0.90–2.97) 1.23 (0.61–2.49) 2.44 (0.93–6.42) 
 ≥4 1.61 (0.92–2.81) 0.60 (0.20–1.78) 2.24 (1.23–4.08) 1.79 (0.91–3.53) 3.34 (1.27–8.75) 
Ptrend 0.16 0.36 0.05 0.14 0.07 
Number of live births 
 0 Reference Reference Reference Reference Reference 
 1 1.50 (0.87–2.58) 1.15 (0.39–3.39) 1.67 (0.94–2.94) 0.78 (0.37–1.65) 2.20 (0.99–4.88) 
 2 1.32 (0.84–2.09) 1.10 (0.44–2.76) 1.50 (0.93–2.43) 1.14 (0.65–1.98) 1.53 (0.75–3.11) 
 3 1.39 (0.84–2.28) 1.05 (0.38–2.93) 1.41 (0.83–2.38) 1.09 (0.60–2.00) 1.37 (0.63–2.97) 
 ≥4 1.86 (1.05–3.27) 0.76 (0.23–2.52) 2.24 (1.24–4.04) 1.78 (0.92–3.46) 2.16 (0.95–4.89) 
Ptrend 0.08 0.58 0.05 0.11 0.28 
Any serrated polypSessile serrated lesionAny hyperplastic polypMicrovesicular HPGoblet cell–rich HP
CharacteristicRR (95% CI)RR (95% CI)RR (95% CI)RR (95% CI)RR (95% CI)
Menopausal status 
 Pre Reference Reference Reference Reference Reference 
 Post 1.01 (0.68–1.48) 1.32 (0.61–2.85) 0.89 (0.60–1.33) 1.07 (0.66–1.74) 0.67 (0.38–1.16) 
Baseline HRT status 
 Never Reference Reference Reference Reference Reference 
 Former 1.05 (0.76–1.47) 2.04 (1.11–3.75) 0.92 (0.65–1.31) 1.06 (0.70–1.59) 0.64 (0.38–1.07) 
 Current 0.69 (0.45–1.05) 0.44 (0.13–1.53) 0.66 (0.42–1.03) 0.63 (0.36–1.11) 0.66 (0.34–1.29) 
Ptrend 0.13 0.89 0.07 0.12 0.15 
Ever used contraceptives 
 No Reference Reference Reference Reference Reference 
 Yes 0.97 (0.66–1.42) 2.12 (0.84–5.36) 0.79 (0.53–1.18) 1.04 (0.64–1.71) 0.72 (0.41–1.27) 
Age at menarche 
 11 Reference Reference Reference Reference Reference 
 12 1.10 (0.72–1.69) 1.92 (0.77–4.76) 1.09 (0.69–1.72) 1.27 (0.74–2.20) 0.91 (0.47–1.74) 
 13 1.12 (0.75–1.67) 1.51 (0.61–3.77) 1.04 (0.68–1.61) 1.35 (0.80–2.29) 1.13 (0.63–2.02) 
 ≥14 0.74 (0.45–1.19) 0.78 (0.25–2.40) 0.77 (0.46–1.29) 0.89 (0.47–1.70) 0.88 (0.44–1.76) 
Ptrend 0.29 0.61 0.33 0.89 0.95 
Number of pregnancies 
 0 Reference Reference Reference Reference Reference 
 1 1.30 (0.68–2.46) 0.65 (0.18–2.33) 1.81 (0.92–3.58) 1.08 (0.46–2.54) 3.44 (1.20–9.86) 
 2 1.48 (0.85–2.58) 0.98 (0.35–2.73) 1.92 (1.06–3.48) 1.60 (0.82–3.13) 2.52 (0.94–6.78) 
 3 1.33 (0.76–2.31) 0.85 (0.30–2.42) 1.63 (0.90–2.97) 1.23 (0.61–2.49) 2.44 (0.93–6.42) 
 ≥4 1.61 (0.92–2.81) 0.60 (0.20–1.78) 2.24 (1.23–4.08) 1.79 (0.91–3.53) 3.34 (1.27–8.75) 
Ptrend 0.16 0.36 0.05 0.14 0.07 
Number of live births 
 0 Reference Reference Reference Reference Reference 
 1 1.50 (0.87–2.58) 1.15 (0.39–3.39) 1.67 (0.94–2.94) 0.78 (0.37–1.65) 2.20 (0.99–4.88) 
 2 1.32 (0.84–2.09) 1.10 (0.44–2.76) 1.50 (0.93–2.43) 1.14 (0.65–1.98) 1.53 (0.75–3.11) 
 3 1.39 (0.84–2.28) 1.05 (0.38–2.93) 1.41 (0.83–2.38) 1.09 (0.60–2.00) 1.37 (0.63–2.97) 
 ≥4 1.86 (1.05–3.27) 0.76 (0.23–2.52) 2.24 (1.24–4.04) 1.78 (0.92–3.46) 2.16 (0.95–4.89) 
Ptrend 0.08 0.58 0.05 0.11 0.28 

Note: Analysis limited to female participants. Each characteristic is run in a separate GEE model, adjusted for age, center [grouped geographically into southeast (Georgia, North Carolina, South Carolina, and Puerto Rico), north (Ohio, New Hampshire, Iowa, and Minnesota), and west (Colorado, Texas, and California), race (white, black, other), study phase (treatment, observational), vitamin D study treatment (placebo, active), calcium study treatment (placebo, active—this includes women who elected to receive calcium and were not randomized to it)], and either number of serrated polyps at baseline (0, 1, 2+) for any serrated polyp and hyperplastic polyp outcomes or baseline SSL/right-sided serrated polyp (no, yes) for SSL outcome. Men were included in the models and treated as a separate group; they were dropped from the model to obtain P for trend.

Abbreviation: HRT, hormone replacement therapy.

A history of serrated polyps at baseline was the strongest risk factor for serrated polyps during follow-up [RR (95% CI) for 1 or 2+ serrated polyps at baseline vs. none: 1.91 (1.53–2.39) and 4.47 (3.34–6.00), respectively, Ptrend < 0.0001; Fig. 1B]. Smoking status was also significantly associated with serrated polyps. Current smokers at baseline exhibited the highest risk of serrated polyps during follow-up for all serrated polyp subtypes [current smoker vs. never smoker RR (95% CI) for MVHPs: 1.58 (1.10–2.28); GCHPs: 2.36 (1.59–3.52); and SSLs: 1.70 (1.00–2.86); Fig. 1C]. We also observed a dose-related trend with respect to duration smoked and maximum amount smoked and HP occurrence, although a clear relationship between these variables and SSL risk was not demonstrated. Alcohol use was associated with an increased risk of HPs and MVHPs specifically [RR (95% CI) for >1 drink/day vs. none: 1.30 (1.01–1.69) and 1.61 (1.18–2.18), respectively], but was not associated with risk of GCHPs. Obesity was associated with an elevated risk of HPs, and GCHPs specifically [RR (95% CI) for BMI ≥30 vs. <25 kg/m2: 1.31 (1.03–1.68) and 1.54 (1.07–2.21), respectively], although no clear association between obesity and either SSLs or MVHPs was identified.

In this study using data from a large, multicenter, prospective study, we found that patients who reported smoking at baseline had an increased risk of incident serrated polyps of all types, and specifically HPs. Nonwhite participants had a lower risk of developing SSLs and MVHPs, but had a higher risk of developing GCHPs. Not surprisingly, those with a prior history of serrated polyps at their baseline colonoscopy exhibited an increased risk of developing serrated polyps and specifically SSLs during the follow-up period. We did not find evidence that baseline use of NSAIDs or aspirin reduced the risk of serrated polyps in contrast to prior work (9–11).

A limited number of studies have examined risk factors for serrated polyps, both in aggregate and for serrated polyp subtypes of more clinical importance, namely SSLs and TSAs. We did not find a relationship between age or sex and risk of serrated polyps, which is consistent with most prior literature demonstrating minimal or no increased risk of SSLs beyond age 50, and roughly equivalent risk of SSLs among men and women (4, 12, 13). This is not necessarily in conflict with findings that women are at higher risk of BRAF-mutated cancers than men (14, 15), because female sex is inversely associated with risk of conventional adenomas and KRAS-mutated cancers, which are more common.

A meta-analysis of modifiable risk factors for serrated polyps published in 2017 (comprising six studies with data on SSLs) found that smoking and alcohol intake were associated with SSLs in particular (16). In addition, an analysis of pooled data from two large cohort studies (n = 141,143) also found that smoking and alcohol (as well as BMI) were associated with serrated polyps (17). A prior study by our group found that smoking was associated with increased risk of serrated polyps, particularly in the left colon (18). Similar to these other reports, we found that smoking and alcohol were associated with serrated class polyps. In particular, we found that smoking (including smoking duration and maximum amount smoked) was most strongly related to HP occurrence, but this effect was attenuated for SSLs. The implications of this finding are not entirely clear, as HPs are generally not thought to be premalignant lesions.

Smoking has been associated with serrated pathway cancers as well as polyps. Studies of invasive colorectal cancer have consistently identified a stronger relationship between cigarette smoking and risk of MSI, CIMP, and/or BRAF-mutated cancers compared with conventional (WNT/KRAS driven) cancers (19, 20). Although the reasons for this are not entirely clear, cigarette smoke has direct immunomodulatory effects in the colon, which promote Th1 inflammation and impede T-cell memory capacity (21–23). Smoking may also increase immune checkpoint expression, thereby perhaps decreasing immune surveillance in early colorectal carcinogenesis (24).

Regarding race, our finding that black individuals appear to be at lower risk of SSLs (and conversely that white individuals are at higher risk of SSLs) is also consistent with prior work (10, 25–28). Reasons for this finding are uncertain. As race is largely a social construct, it is possible that this relationship is confounded by other factors such as colonoscopy quality or socioeconomic status. However, a prior study demonstrated that even within a low-income and uninsured colonoscopy screening population, black individuals were still at lower risk of serrated polyps, suggesting that socioeconomic status alone does not explain these differences (29). One prior study found that higher levels of education were associated with serrated polyps (and SSLs specifically; ref. 30), but we did not see a clear association between education levels and serrated polyp outcomes in our analysis. The contrasting associations between race and MVHPs and GCHPs is somewhat puzzling. Interestingly, this finding has also been reported by other researchers as well. In a single-center US cross-sectional study of over 3,500 patients undergoing colonoscopy, Qazi and colleagues found that compared with white participants, black participants were at a decreased risk for MVHPs but an increased risk of GCHPs [OR (95% CI) 0.55 (0.40–0.75) and 1.82 (1.37–2.45), respectively], a pattern consistent with our findings for risk of incident lesions (5). These polyps have different molecular features: MVHPs are frequently BRAF mutated, whereas GCHPs are not, and KRAS mutation is more common in GCHPs than MVHPs (31). This raises the possibility that differences in genetic ancestry (measured by race as a proxy) could explain why black individuals appeared to be at higher risk of GCHPs, despite a lower risk of MVHPs and SSLs. Indeed, KRAS mutation rates appear to be 10% to 15% higher in colorectal cancers among black patients compared with white patients (32–36). Furthermore, differences in immune responses have been observed in black individuals that may influence the risk, growth, and pathway trajectory of serrated neoplasia (37–41). Parenthetically, multiple studies have shown that black individuals have a higher prevalence of proximal (nonserrated) adenomas and advanced adenomas compared with white individuals, which suggests possible biologic differences related to conventional pathway lesions as well (29, 42–44). These findings could be important to explore along with mutational profiles of different polyp types in future studies.

Similar to others, we found that prior history of serrated polyps predicts future occurrence (45, 46). In contrast, presence of advanced adenomas at baseline did not predict occurrence of serrated polyps during follow-up in this study. These findings support the concept that the traditional adenoma-carcinoma sequence and serrated pathway(s) are distinct, with differing risk factors, although patients can certainly harbor both types of polyps (12).

We previously published an analysis of the treatment effects of calcium and vitamin D on serrated polyp outcomes (47). That study found that calcium and the combination of calcium and vitamin D led to increased risk of SSLs, particularly in the later years of the study. Because of this, we carefully controlled for treatment assignment in this study, so as to isolate nontreatment participant characteristics associated with the development of serrated polyps and particularly SSLs. Furthermore, because treatment was randomized, this is unlikely to be a significant confounder of our results.

Strengths of this study include the use of data from a relatively large number of participants from a multicenter trial. We examined incident serrated polyps within a prospective trial, which is an advantage with respect to studying causal associations compared with cross-sectional studies. Although pathology interpretation of serrated polyps can be a pitfall of epidemiology studies of this kind, we performed central path review by an expert GI pathologist who helped author the WHO criteria for serrated polyps. Furthermore, the fact that participants were included from multiple geographically dispersed sites across the US contributes to the external validity of our results. However, we had relatively few outcomes, particularly with respect to SSLs and TSAs, which is common among studies of these lesions given their relatively low incidence compared with conventional adenomas. We were unable to perform detailed analyses on TSA risk factors given their rarity. We aimed to perform a broad analysis of possible serrated polyp risk factors, but given the multiple comparisons performed, it is possible that some positive associations occurred by chance alone. Despite use of multivariable modeling, residual confounding is also possible.

Multiple studies have demonstrated variable detection of serrated polyps and SSLs specifically (4, 48), and this could have also affected our study. If SSLs were underdetected at baseline, lesions detected during follow-up would not be truly “incident” but rather pre-existing lesions. This should not materially affect our results regarding risk factor associations. If SSLs were underdetected during follow-up (a form of measurement bias), it could bias results though imperfect sensitivity of outcome measures is generally thought to have minimal effects on risk ratios when specificity is high (49). We used local pathologist diagnoses for baseline results, which could have been inaccurate given known variability in pathology diagnosis of serrated lesions among non-expert pathologists (50). We attempted to minimize the potential impact of imperfect baseline diagnoses of serrated polyps by combining SSLs with proximal hyperplastic polyps in our analyses. Finally, all participants in the VCPPS trial had to have at least one adenomatous polyp on baseline exam, so this study did not include patients with isolated serrated polyps. Prior research suggests that roughly half of patients with SSLs have synchronous conventional adenomas (51). Although we have no reason to believe that risk factor associations for serrated class polyps would be different in this group, it is possible that this feature of the study design impacts the external validity of our findings.

In conclusion, using data from a large, multicenter prospective trial, we found evidence that smoking, presence of serrated polyps at baseline, and white race were associated with incident SSLs. We also saw that black participants were at higher risk of GCHPs, and white participants were at higher risk of MVHPs, for reasons that are unclear. Additional research is needed to explore the underlying mechanisms for these findings. Understanding these risk factors could allow for better risk stratification of patients undergoing colorectal cancer screening, and could potentially guide screening test selection, as serrated class polyps are less-well detected by noninvasive screening tests (52). Furthermore, risk factor associations for SPs differ from known risk factors for conventional adenomas, which supports the concept of etiologic heterogeneity of colorectal cancer.

S.D. Crockett reports grants from National Institutes of Health during the conduct of the study as well as other support from Exact Sciences, Freenome, and Guardant outside the submitted work. E.L. Barry reports grants from NIH/NCI during the conduct of the study. L.A. Mott reports grants from NIH/NCI during the conduct of the study. D.C. Snover reports personal fees from Dartmouth University during the conduct of the study. J.A. Baron reports grants from NCI during the conduct of the study; in addition, J.A. Baron has a patent for chemopreventive use of aspirin issued and a patent for chemopreventive use of calcium issued. No disclosures were reported by the other authors.

S.D. Crockett: Conceptualization, supervision, investigation, methodology, writing–original draft, writing–review and editing. E.L. Barry: Conceptualization, data curation, formal analysis, methodology, writing–original draft, project administration, writing–review and editing. L.A. Mott: Data curation, formal analysis, investigation, methodology, writing–original draft, writing–review and editing. D.C. Snover: Investigation, writing–review and editing. K. Wallace: Writing–review and editing. J.A. Baron: Conceptualization, resources, supervision, funding acquisition, investigation, methodology, writing–review and editing.

This study was funded by a grant from the NIH, NCI (CA098286, PI: J.A. Baron). S.D. Crockett's effort was supported in part by a grant from the NIH (KL2TR001109).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Leggett
B
,
Whitehall
V
.
Role of the serrated pathway in colorectal cancer pathogenesis
.
Gastroenterology
2010
;
138
:
2088
100
.
2.
Snover
DC
.
Update on the serrated pathway to colorectal carcinoma
.
Hum Pathol
2011
;
42
:
1
10
.
3.
Crockett
SD
,
Nagtegaal
I
.
Terminology, molecular features, epidemiology, and management of serrated colorectal neoplasia
.
Gastroenterology
2019
;
157
:
949
66
.
4.
Ijspeert
JEG
,
de Wit
K
,
van der Vlugt
M
,
Bastiaansen
BA
,
Fockens
P
,
Dekker
E
.
Prevalence, distribution and risk of sessile serrated adenomas/polyps at a center with a high adenoma detection rate and experienced pathologists
.
Endoscopy
2016
;
48
:
740
6
.
5.
Qazi
TM
,
O'Brien
MJ
,
Farraye
FA
,
Gould
RW
,
Chen
CA
,
Schroy
PC
3rd
.
Epidemiology of goblet cell and microvesicular hyperplastic polyps
.
Am J Gastroenterol
2014
;
109
:
1922
32
.
6.
Rex
DK
,
Ahnen
DJ
,
Baron
JA
,
Batts
KP
,
Burke
CA
,
Burt
RW
, et al
.
Serrated lesions of the colorectum: review and recommendations from an expert panel
.
Am J Gastroenterol
2012
;
107
:
1315
29
;
quiz 4, 30
.
7.
Baron
JA
,
Barry
EL
,
Mott
LA
,
Rees
JR
,
Sandler
RS
,
Snover
DC
, et al
.
A trial of calcium and vitamin D for the prevention of colorectal adenomas
.
N Engl J Med
2015
;
373
:
1519
30
.
8.
Snover
DC
,
Ahnen
D
,
Burt
R
,
Odze
RD
.
Serrated polyps of the colon and rectum and serrated polyposis
.
WHO Classification of Tumours of the Digestive System
. 4th ed.
Lyon, France
:
IARC
;
2010
.
9.
Bouwens
MW
,
Winkens
B
,
Rondagh
EJ
,
Driessen
AL
,
Riedl
RG
,
Masclee
AA
, et al
.
Simple clinical risk score identifies patients with serrated polyps in routine practice
.
Cancer Prev Res
2013
;
6
:
855
63
.
10.
Wallace
K
,
Grau
MV
,
Ahnen
D
,
Snover
DC
,
Robertson
DJ
,
Mahnke
D
, et al
.
The association of lifestyle and dietary factors with the risk for serrated polyps of the colorectum
.
Cancer Epidemiol Biomarkers Prev
2009
;
18
:
2310
7
.
11.
Davenport
JR
,
Su
T
,
Zhao
Z
,
Coleman
HG
,
Smalley
WE
,
Ness
RM
, et al
.
Modifiable lifestyle factors associated with risk of sessile serrated polyps, conventional adenomas and hyperplastic polyps
.
Gut
2018
;
67
:
456
65
.
12.
Haque
TR
,
Bradshaw
PT
,
Crockett
SD
.
Risk factors for serrated polyps of the colorectum
.
Dig Dis Sci
2014
;
59
:
2874
89
.
13.
O'Connell
BM
,
Crockett
SD
.
The clinical impact of serrated colorectal polyps
.
Clin Epidemiol
2017
;
9
:
113
25
.
14.
Tie
J
,
Gibbs
P
,
Lipton
L
,
Christie
M
,
Jorissen
RN
,
Burgess
AW
, et al
.
Optimizing targeted therapeutic development: analysis of a colorectal cancer patient population with the BRAF(V600E) mutation
.
Int J Cancer
2011
;
128
:
2075
84
.
15.
Gonsalves
WI
,
Mahoney
MR
,
Sargent
DJ
,
Nelson
GD
,
Alberts
SR
,
Sinicrope
FA
, et al
.
Patient and tumor characteristics and BRAF and KRAS mutations in colon cancer, NCCTG/Alliance N0147
.
J Natl Cancer Inst
2014
;
106
:
dju106
.
16.
Bailie
L
,
Loughrey
MB
,
Coleman
HG
.
Lifestyle risk factors for serrated colorectal polyps: a systematic review and meta-analysis
.
Gastroenterology
2017
;
152
:
92
104
.
17.
He
X
,
Wu
K
,
Ogino
S
,
Giovannucci
EL
,
Chan
AT
,
Song
M
.
Association between risk factors for colorectal cancer and risk of serrated polyps and conventional adenomas
.
Gastroenterology
2018
;
155
:
355
73
.
18.
Figueiredo
JC
,
Crockett
SD
,
Snover
DC
,
Morris
CB
,
McKeown-Eyssen
G
,
Sandler
RS
, et al
.
Smoking-associated risks of conventional adenomas and serrated polyps in the colorectum
.
Cancer Causes Control
2015
;
26
:
377
86
.
19.
Amitay
EL
,
Carr
PR
,
Jansen
L
,
Roth
W
,
Alwers
E
,
Herpel
E
, et al
.
Smoking, alcohol consumption and colorectal cancer risk by molecular pathological subtypes and pathways
.
Br J Cancer
2020
;
122
:
1604
10
.
20.
Botteri
E
,
Borroni
E
,
Sloan
EK
,
Bagnardi
V
,
Bosetti
C
,
Peveri
G
, et al
.
Smoking and colorectal cancer risk, overall and by molecular subtypes: a meta-analysis
.
Am J Gastroenterol
2020
;
115
:
1940
9
.
21.
Qin
Z
,
Wang
PY
,
Wan
JJ
,
Zhang
Y
,
Wei
J
,
Sun
Y
, et al
.
MicroRNA124-IL6R mediates the effect of nicotine in inflammatory bowel disease by shifting Th1/Th2 balance toward Th1
.
Front Immunol
2020
;
11
:
235
.
22.
Tejero
JD
,
Armand
NC
,
Finn
CM
,
Dhume
K
,
Strutt
TM
,
Chai
KX
, et al
.
Cigarette smoke extract acts directly on CD4 T cells to enhance Th1 polarization and reduce memory potential
.
Cell Immunol
2018
;
331
:
121
9
.
23.
Lee
G
,
Jung
KH
,
Shin
D
,
Lee
C
,
Kim
W
,
Lee
S
, et al
.
Cigarette smoking triggers colitis by IFN-gamma(+) CD4(+) T cells
.
Front Immunol
2017
;
8
:
1344
.
24.
Wang
GZ
,
Zhang
L
,
Zhao
XC
,
Gao
SH
,
Qu
LW
,
Yu
H
, et al
.
The Aryl hydrocarbon receptor mediates tobacco-induced PD-L1 expression and is associated with response to immunotherapy
.
Nat Commun
2019
;
10
:
1125
.
25.
Jackson
CS
,
Vega
KJ
.
Higher prevalence of proximal colon polyps and villous histology in African-Americans undergoing colonoscopy at a single equal access center
.
J Gastrointest Oncol
2015
;
6
:
638
43
.
26.
Parikh
MP
,
Muthukuru
S
,
Jobanputra
Y
,
Naha
K
,
Gupta
NM
,
Wadhwa
V
, et al
.
Proximal sessile serrated adenomas are more prevalent in caucasians, and gastroenterologists are better than nongastroenterologists at their detection
.
Gastroenterol Res Pract
2017
;
2017
:
6710931
.
27.
Peery
AF
,
Crockett
SD
,
Murphy
CC
,
Lund
JL
,
Dellon
ES
,
Williams
JL
, et al
.
Burden and cost of gastrointestinal, liver, and pancreatic diseases in the United States: Update 2018
.
Gastroenterology
2018
;
156
:
254
72
.
28.
Shaukat
A
,
Holub
J
,
Greenwald
D
,
Eisen
G
,
Schmitt
C
.
Variation over time and factors associated with detection rates of sessile serrated lesion across the United States: results form a national sample using the GIQuIC registry
.
Am J Gastroenterol
2021
;
116
:
95
9
.
29.
Wallace
K
,
Brandt
HM
,
Bearden
JD
,
Blankenship
BF
,
Caldwell
R
,
Dunn
J
, et al
.
Race and prevalence of large bowel polyps among the low-income and uninsured in South Carolina
.
Dig Dis Sci
2016
;
61
:
265
72
.
30.
Burnett-Hartman
AN
,
Passarelli
MN
,
Adams
SV
,
Upton
MP
,
Zhu
LC
,
Potter
JD
, et al
.
Differences in epidemiologic risk factors for colorectal adenomas and serrated polyps by lesion severity and anatomical site
.
Am J Epidemiol
2013
;
177
:
625
37
.
31.
Shinmura
K
,
Konishi
K
,
Yamochi
T
,
Kubota
Y
,
Yano
Y
,
Katagiri
A
, et al
.
Molecular features of colorectal polyps presenting Kudo's type II mucosal crypt pattern: are they based on the same mechanism of tumorigenesis?
Endosc Int Open
2014
;
2
:
E171
7
.
32.
Kang
M
,
Shen
XJ
,
Kim
S
,
Araujo-Perez
F
,
Galanko
JA
,
Martin
CF
, et al
.
Somatic gene mutations in African Americans may predict worse outcomes in colorectal cancer
.
Cancer Biomark
2013
;
13
:
359
66
.
33.
Guda
K
,
Veigl
ML
,
Varadan
V
,
Nosrati
A
,
Ravi
L
,
Lutterbaugh
J
, et al
.
Novel recurrently mutated genes in African American colon cancers
.
Proc Natl Acad Sci U S A
2015
;
112
:
1149
54
.
34.
Yoon
HH
,
Shi
Q
,
Alberts
SR
,
Goldberg
RM
,
Thibodeau
SN
,
Sargent
DJ
, et al
.
Racial differences in BRAF/KRAS mutation rates and survival in stage III colon cancer patients
.
J Natl Cancer Inst
2015
;
107
:
djv186
.
35.
Xicola
RM
,
Gagnon
M
,
Clark
JR
,
Carroll
T
,
Gao
W
,
Fernandez
C
, et al
.
Excess of proximal microsatellite-stable colorectal cancer in African Americans from a multiethnic study
.
Clin Cancer Res
2014
;
20
:
4962
70
.
36.
Sylvester
BE
,
Huo
D
,
Khramtsov
A
,
Zhang
J
,
Smalling
RV
,
Olugbile
S
, et al
.
Molecular analysis of colorectal tumors within a diverse patient cohort at a single institution
.
Clin Cancer Res
2012
;
18
:
350
9
.
37.
Fessler
E
,
Drost
J
,
van Hooff
SR
,
Linnekamp
JF
,
Wang
X
,
Jansen
M
, et al
.
TGFbeta signaling directs serrated adenomas to the mesenchymal colorectal cancer subtype
.
EMBO Mol Med
2016
;
8
:
745
60
.
38.
De Sousa
EMF
,
Wang
X
,
Jansen
M
,
Fessler
E
,
Trinh
A
,
de Rooij
LP
, et al
.
Poor-prognosis colon cancer is defined by a molecularly distinct subtype and develops from serrated precursor lesions
.
Nat Med
2013
;
19
:
614
8
.
39.
Ye
CJ
,
Feng
T
,
Kwon
HK
,
Raj
T
,
Wilson
MT
,
Asinovski
N
, et al
.
Intersection of population variation and autoimmunity genetics in human T cell activation
.
Science
2014
;
345
:
1254665
.
40.
Quach
H
,
Rotival
M
,
Pothlichet
J
,
Loh
YE
,
Dannemann
M
,
Zidane
N
, et al
.
Genetic adaptation and neandertal admixture shaped the immune system of human populations
.
Cell
2016
;
167
:
643
56
.
41.
Nedelec
Y
,
Sanz
J
,
Baharian
G
,
Szpiech
ZA
,
Pacis
A
,
Dumaine
A
, et al
.
Genetic ancestry and natural selection drive population differences in immune responses to pathogens
.
Cell
2016
;
167
:
657
69
.
42.
Corley
DA
,
Jensen
CD
,
Marks
AR
,
Zhao
WK
,
de Boer
J
,
Levin
TR
, et al
.
Variation of adenoma prevalence by age, sex, race, and colon location in a large population: implications for screening and quality programs
.
Clin Gastroenterol Hepatol
2013
;
11
:
172
80
.
43.
Lieberman
DA
,
Williams
JL
,
Holub
JL
,
Morris
CD
,
Logan
JR
,
Eisen
GM
, et al
.
Race, ethnicity, and sex affect risk for polyps >9 mm in average-risk individuals
.
Gastroenterology
2014
;
147
:
351
8
;
quiz e14–5
.
44.
Schroy
PC
3rd
,
Coe
A
,
Chen
CA
,
O'Brien
MJ
,
Heeren
TC
.
Prevalence of advanced colorectal neoplasia in white and black patients undergoing screening colonoscopy in a safety-net hospital
.
Ann Intern Med
2013
;
159
:
13
20
.
45.
Anderson
JC
,
Butterly
LF
,
Robinson
CM
,
Weiss
JE
,
Amos
C
,
Srivastava
A
.
Risk of metachronous high-risk adenomas and large serrated polyps in individuals with serrated polyps on index colonoscopy: Data from the new Hampshire colonoscopy registry
.
Gastroenterology
2018
;
154
:
117
27
.
46.
Bensen
SP
,
Cole
BF
,
Mott
LA
,
Baron
JA
,
Sandler
RS
,
Haile
R
, et al
.
Colorectal hyperplastic polyps and risk of recurrence of adenomas and hyperplastic polyps
.
Lancet
1999
;
354
:
1873
4
.
47.
Crockett
SD
,
Barry
EL
,
Mott
LA
,
Ahnen
DJ
,
Robertson
DJ
,
Anderson
JC
, et al
.
Calcium and vitamin D supplementation and increased risk of serrated polyps: results from a randomised clinical trial
.
Gut
2019
;
68
:
475
86
.
48.
Crockett
SD
,
Gourevitch
RA
,
Morris
M
,
Carrell
DS
,
Rose
S
,
Shi
Z
, et al
.
Endoscopist factors that influence serrated polyp detection: a multicenter study
.
Endoscopy
2018
;
50
:
984
92
.
49.
Copeland
KT
,
Checkoway
H
,
McMichael
AJ
,
Holbrook
RH
.
Bias due to misclassification in the estimation of relative risk
.
Am J Epidemiol
1977
;
105
:
488
95
.
50.
Gourevitch
RA
,
Rose
S
,
Crockett
SD
,
Morris
M
,
Carrell
DS
,
Greer
JB
, et al
.
Variation in pathologist classification of colorectal adenomas and serrated polyps
.
Am J Gastroenterol
2018
;
113
:
431
9
.
51.
Symonds
E
,
Anwar
S
,
Young
G
,
Meng
R
,
Coats
M
,
Simpson
K
, et al
.
Sessile serrated polyps with synchronous conventional adenomas increase risk of future advanced Neoplasia
.
Dig Dis Sci
2019
;
64
:
1680
5
.
52.
Crockett
SD
.
Sessile serrated polyps and colorectal cancer
.
JAMA
2017
;
317
:
975
6
.

Supplementary data