archived

Evidence Summary

Colorectal Cancer: Screening

June 15, 2016

Recommendations made by the USPSTF are independent of the U.S. government. They should not be construed as an official position of the Agency for Healthcare Research and Quality or the U.S. Department of Health and Human Services.

By Jennifer S. Lin, MD; Margaret A. Piper, PhD; Leslie A. Perdue, MPH; Carolyn M. Rutter, PhD; Elizabeth M. Webber, MS; Elizabeth O'Connor, PhD; Ning Smith, PhD; and Evelyn P. Whitlock, MD

The information in this article is intended to help clinicians, employers, policymakers, and others make informed decisions about the provision of health care services. This article is intended as a reference and not as a substitute for clinical judgment.

This article may be used, in whole or in part, as the basis for the development of clinical practice guidelines and other quality enhancement tools, or as a basis for reimbursement and coverage policies. AHRQ or U.S. Department of Health and Human Services endorsement of such derivative products may not be stated or implied.

This article was first published in the Journal of the American Medical Association on June 15, 2016.

 

Return to Table of Contents

Importance: Colorectal cancer (CRC) remains a significant cause of morbidity and mortality in the United States.

Objective: To systematically review the effectiveness, diagnostic accuracy, and harms of screening for CRC.

Data Sources: Searches of MEDLINE, PubMed, and the Cochrane Central Register of Controlled Trials for relevant studies published from January 1, 2008, through December 31, 2014, with surveillance through February 23, 2016.

Study Selection: English-language studies conducted in asymptomatic populations at general risk of CRC.

Data Extraction and Synthesis: Two reviewers independently appraised the articles and extracted relevant study data from fair- or good-quality studies. Random-effects meta-analyses were conducted.

Main Outcomes and Measures: Colorectal cancer incidence and mortality, test accuracy in detecting CRC or adenomas, and serious adverse events.

Results: Four pragmatic randomized clinical trials (RCTs) evaluating 1-time or 2-time flexible sigmoidoscopy (n = 458,002) were associated with decreased CRC-specific mortality compared with no screening (incidence rate ratio, 0.73; 95% CI, 0.66-0.82). Five RCTs with multiple rounds of biennial screening with guaiac-based fecal occult blood testing (n = 419,966) showed reduced CRC-specific mortality (relative risk [RR], 0.91; 95% CI, 0.84-0.98, at 19.5 years to RR, 0.78; 95% CI, 0.65-0.93, at 30 years). Seven studies of computed tomographic colonography (CTC) with bowel preparation demonstrated per-person sensitivity and specificity to detect adenomas 6 mm and larger comparable with colonoscopy (sensitivity from 73% [95% CI, 58%-84%] to 98% [95% CI, 91%-100%]; specificity from 89% [95% CI, 84%-93%] to 91% [95% CI, 88%-93%]); variability and imprecision may be due to differences in study designs or CTC protocols. Sensitivity of colonoscopy to detect adenomas 6 mm or larger ranged from 75% (95% CI, 63%-84%) to 93% (95% CI, 88%-96%). On the basis of a single stool specimen, the most commonly evaluated families of fecal immunochemical tests (FITs) demonstrated good sensitivity (range, 73%-88%) and specificity (range, 90%-96%). One study (n = 9989) found that FIT plus stool DNA test had better sensitivity in detecting CRC than FIT alone (92%) but lower specificity (84%). Serious adverse events from colonoscopy in asymptomatic persons included perforations (4/10,000 procedures, 95% CI, 2-5 in 10,000) and major bleeds (8/10,000 procedures, 95% CI, 5-14 in 10,000). Computed tomographic colonography may have harms resulting from low-dose ionizing radiation exposure or identification of extracolonic findings.

Conclusions and Relevance: Colonoscopy, flexible sigmoidoscopy, CTC, and stool tests have differing levels of evidence to support their use, ability to detect cancer and precursor lesions, and risk of serious adverse events in average-risk adults. Although CRC screening has a large body of supporting evidence, additional research is still needed.

Return to Table of Contents

Although colorectal cancer (CRC) incidence has been declining over the past 20 years in the United States, it still causes significant morbidity and mortality.1 Despite increases in screening rates over the past 30 years, in 2012 an estimated 28% of eligible US adults had never been screened for CRC.2 A variety of tests are available for screening, including stool-based tests (eg, guaiac-based fecal occult blood testing [gFOBT], immunochemical-based fecal occult blood testing [FIT], stool DNA [sDNA] testing), endoscopy (eg, flexible sigmoidoscopy [SIG], colonoscopy), and imaging (eg, double-contrast barium enema, computed tomographic colonography [CTC]).

Currently, most US guideline organizations, including the US Preventive Services Task Force (USPSTF), recommend that options for CRC screening include colonoscopy every 10 years, an annual high-sensitivity gFOBT or FIT, and SIG every 5 years with high-sensitivity gFOBT or FIT.3, 4 In 2008, the USPSTF recommended CRC screening using fecal occult blood testing, sigmoidoscopy, or colonoscopy beginning at age 50 years and continuing until age 75 years (A recommendation); selectively offering screening in adults aged 76 to 85 years (C recommendation); and against screening for colorectal cancer in adults older than 85 years (D recommendation). At that time, the USPSTF had insufficient evidence to assess the benefits and harms of CTC and sDNA testing as screening modalities. A systematic review was conducted to update relevant evidence since 2008 and to help inform a separate modeling exercise, which together were used by the USPSTF in its process of updating the 2008 CRC screening recommendations.

Return to Table of Contents

Scope of Review

This review addressed 3 key questions (KQs) as shown in Figure 1. Additional methodological details regarding search strategies, detailed study inclusion criteria, quality assessment, excluded studies, and description of data analyses are publicly available in the full evidence report at: http://www.uspreventiveservicestaskforce.org/Page/Document/UpdateSummaryFinal/colorectal-cancer-screening2.5

Data Sources and Searches

MEDLINE, PubMed, and the Cochrane Central Register of Controlled Trials were searched to locate primary studies informing the key questions (eMethods in the Supplement) that were published from the end of the previous review6 (January 1, 2008) through December 31, 2014. The database searches were supplemented with expert suggestions and by reviewing reference lists from all other relevant systematic reviews, including the 2008 USPSTF evidence report. The search also included selected gray literature sources, including ClinicalTrials.gov and the World Health Organization International Clinical Trials Registry Platform, for ongoing trials. Since December 2014, we continued to conduct ongoing surveillance through article alerts and targeted searches of high-impact journals to identify major studies published in the interim that may affect the conclusions or understanding of the evidence and therefore the related USPSTF recommendation. The last surveillance was conducted on February 23, 2016. Although several potentially relevant new studies were identified,7-9 none of these studies would substantively change the review's interpretation of findings or conclusions.

Study Selection

Two investigators independently reviewed 8492 titles and abstracts and 696 articles against the specified inclusion criteria (Figure 2). Discrepancies were resolved through consensus and consultation with a third investigator. Inclusion criteria were fair- and good-quality English-language studies of asymptomatic screening populations of individuals who were 40 years or older, either at average risk for CRC or not selected for inclusion based on CRC risk factors. Studies were included that evaluated the following screening tests: colonoscopy, SIG, CTC, gFOBT, FIT, FIT plus sDNA, or a blood test for methylated SEPT9 DNA (mSEPT9).

For KQ1, randomized clinical trials (RCTs) or otherwise controlled trials of CRC screening vs no screening, as well as trials comparing screening tests, that included outcomes of cancer incidence, CRC-specific mortality, or all-cause mortality were reviewed for inclusion. For tests without trial-level evidence (ie, colonoscopy, FIT), well-conducted prospective cohort or population-based nested case-control studies were examined.

For KQ2, diagnostic accuracy studies that used colonoscopy as a reference standard were included. Studies whose design was subject to a high risk of bias were generally excluded, including studies that did not apply colonoscopy to at least a random subset of screen-negative persons (verification bias)10 and studies without an adequate representation of a full spectrum of patients (spectrum bias), such as case-control studies.10-14 Selected well-conducted FIT diagnostic accuracy studies that used robust registry follow-up for screen-negative participants were included.

For KQ3, all trials and observational studies that reported serious adverse events requiring unexpected or unwanted medical attention or resulting in death were included. These events included, but were not limited to, perforation, major bleeding, severe abdominal symptoms, and cardiovascular events. Studies designed to assess for extracolonic findings (ie, incidental findings on CTC) and the resultant diagnostic yield and harms of workup were also included. Studies reporting extracolonic findings generally used the CT Colonography Reporting and Data System (C-RADS). Under C-RADS, extracolonic findings are categorized as E0 (limited examination), E1 (normal examination or normal variant), E2 (clinically unimportant finding in which no workup is required), E3 (likely unimportant or incompletely characterized in which workup may be required), or E4 (potentially important finding requiring follow-up).15

Data Extraction and Quality Assessment

Two reviewers each critically appraised all articles that met inclusion criteria using the USPSTF design-specific quality criteria16 supplemented by the National Institute for Health and Care Excellence methodology checklists,17 A Measurement Tool to Assess Systematic Reviews (AMSTAR) for systematic reviews,18 Newcastle Ottawa Scales for cohort and case-control studies,19 and Quality Assessment of Diagnostic Accuracy (QUADAS) and QUADAS-2 for studies of diagnostic accuracy (eTable 1 in the Supplement).20, 21 Poor-quality studies and those with a single fatal flaw or multiple important limitations that could invalidate results were excluded from this review. Disagreements about critical appraisal were resolved by consensus and, if needed, consultation with a third independent reviewer. One reviewer extracted key data from included studies; a second reviewer checked the data for accuracy.

Data Synthesis and Analysis

For each KQ, the number and design of included studies, overall results, consistency or precision of results, reporting bias, study quality, limitations of the body of evidence, and applicability of findings were summarized. The results were synthesized by KQ, type of screening test, and study design. Studies from the 2008 review that met the updated inclusion criteria were incorporated. The analyses for test performance focused primarily on per-person (ie, by individual patient rather than by lesion) test sensitivity and specificity to detect adenomas (by size, where reported, <6 mm, ≥6 mm, ≥10 mm), advanced adenomas (as defined by the study), and CRC. The studies used several kinds of FITs, which were grouped as qualitative (fixed cutoff) or quantitative (adjustable cutoff), as well as into families (tests produced by the same manufacturer, using the same components and method, or compatible with different automated analyzers). Tests were compared using similar cutoff values expressed in μg hemoglobin (Hb)/g feces.

Because of the limited number of studies and the clinical heterogeneity of studies, the analyses were largely descriptive. Random-effects meta-analyses were conducted using the profile likelihood method22 to estimate the effect of SIG based on the pooled incidence rate ratio (events/person-year) for CRC incidence and mortality across the 4 major SIG trials. Random-effects models were also conducted using the restricted maximum likelihood estimation method to estimate rates of serious adverse events for colonoscopy and SIG. The presence and magnitude of statistical heterogeneity were assessed among pooled studies using the I2 statistic. All tests were 2-sided with a P value less than 0.05 indicating statistical significance. Meta-analyses were performed using R version 3.0.2 (R Project for Statistical Computing).23, 24

Return to Table of Contents

Effectiveness of Screening

Key Question 1. What is the effectiveness of screening programs based on the prespecified screening tests (alone or in combination) in reducing incidence of and mortality from colorectal cancer?

Twenty-five unique fair- or good-quality studies25-49 (published in 47 articles25-71) were found that assessed the effectiveness or comparative effectiveness of screening tests on CRC incidence and mortality. These studies included 1 cohort study of screening colonoscopy,36 4 RCTs of SIG (in 7 articles),25, 39, 41, 50, 60, 66, 71 and 6 trials of Hemoccult II gFOBT (in 11 articles).29, 33-35, 40, 44, 59, 62-64, 67 In addition, 15 comparative effectiveness studies (in 22 articles)26-28, 30-32, 37, 38, 42, 43, 45-49, 54-58, 65, 69 were found that were primarily designed to assess the relative uptake and CRC yield between different screening modalities. Due to limitations in study designs, the observational colonoscopy study and comparative effectiveness studies are not discussed further in this article. Summarized below are the results for CRC-specific mortality, as results for CRC incidence were consistent with CRC mortality findings.

Flexible Sigmoidoscopy

Four large, fair-quality, pragmatic RCTs (n = 458,002) evaluated the effectiveness of 1 or 2 rounds of SIG in average-risk adults aged 50 to 74 years (Table 1).25, 39, 41, 50, 60, 66, 71 Adherence to SIG in these trials ranged from 58% to 84%, and rates of diagnostic colonoscopy ranged from 5% to 33% due to differences in referral criteria. Based on pooled intention-to-treat analyses, SIG was associated with lower CRC-specific mortality compared with no screening at 11 to 12 years of follow-up (incidence rate ratio, 0.73; 95% CI, 0.66-0.82; I2 = 0%) (Figure 3); however, the association with mortality benefit was limited to distal CRC (incidence rate ratio, 0.63; 95% CI, 0.49-0.84; I2 = 44%) (eFigure 1 in the Supplement). In 1 trial, conducted in Norway, half of the participants randomized to SIG also received a single FIT test; the SIG-plus-FIT group had lower CRC mortality than the SIG-only group did (hazard ratio, 0.62; 95% CI, 0.42-0.90).60

gFOBT

Five older, large, pragmatic RCTs (n = 419,966) with 11 to 30 years of follow-up evaluated the effectiveness of annual or biennial screening programs with Hemoccult II29, 33, 34, 40, 44, 59, 63, 64, 67 (Table 1). Based on intention-to-treat analyses, compared with no screening, biennial screening with Hemoccult II resulted in a reduction in CRC-specific mortality after 2 to 9 rounds of screening (relative risk [RR], 0.91; 95% CI, 0.84-0.98, at 19.5 years to RR, 0.78; 95% CI, 0.65-0.93, at 30 years). Based on 1 trial, conducted in the United States, annual screening with Hemoccult II after 11 rounds of screening resulted in greater reductions (RR, 0.68; 95% CI, 0.56-0.82) than biennial screening at 30 years did (RR, 0.78; 95% CI, 0.65-0.93).44

Diagnostic Accuracy of Screening

Key Question 2. What are the test performance characteristics of the prespecified screening tests (alone or in combination) for detecting colorectal cancer, advanced adenomas, or adenomatous polyps based on size?

Thirty-three unique diagnostic accuracy studies72-104 (published in 44 articles72-115) were found that evaluated the 1-time test performance of a screening test compared with an adequate reference standard, including 9 studies of screening CTC (in 10 articles81, 82, 85-87, 89, 93, 99, 101, 114), 3 studies of gFOBT Hemoccult Sensa,72, 73, 90 20 studies of various FITs72-78, 80, 82-84, 88, 90, 91, 94-98, 100, 102-104 (1 of which evaluated a FIT plus sDNA test83), and 1 study of a blood test to detect circulating mSEPT9.79 The study of mSEPT9 (not approved by the US Food and Drug Administration [FDA] for screening) and studies evaluating Hemoccult Sensa and FITs that only applied the colonoscopy reference standard to positive stool tests are not discussed further in this article.

Direct Visualization Tests

Nine fair- or good-quality studies (n = 6497) evaluated the diagnostic accuracy of multidetector CTC in average-risk screening populations (Table 2).81, 82, 85-87, 89, 93, 99, 101, 114) The 2 largest and best-quality studies were multicenter trials conducted in the United States evaluating CTC with bowel preparation and fecal tagging.85, 99 Overall, the studies were not powered to estimate test performance to detect CRC. Based on 7 studies of CTC with bowel preparation (n = 5328), the per-person sensitivity to detect adenomas 10 mm and larger ranged from 67% (95% CI, 45%-84%) to 94% (95% CI, 84%-98%), and specificity ranged from 98% (95% CI, 96%-99%) to 96% (95% CI, 95%-97%). The per-person sensitivity to detect adenomas 6 mm and larger ranged from 73% (95% CI, 58%-84%) to 98% (95% CI, 91%-100%), and specificity ranged from 89% (95% CI, 84%-93%) to 91% (95% CI, 88%-93%). Two studies (N = 1169) evaluated CTC without bowel preparation.81, 101 Although the data were limited, the sensitivity of CTC without bowel preparation to detect adenomas 6 mm and larger appeared to be lower than the sensitivity of CTC protocols including bowel preparation.

Four (n = 4821) of the 9 CTC studies allowed for the estimation of sensitivity of colonoscopy generalizable to community practice.85, 86, 99, 101 Compared with CTC or colonoscopy plus CTC (eg, segmental unblinding), the sensitivity for colonoscopy to detect adenomas 10 mm and larger ranged from 89% (95% CI, 78%-96%) to 98% (95% CI, 74%-100%) and for adenomas 6 mm and larger ranged from 75% (95% CI, 63%-84%) to 93% (95% CI, 88%-96%) (see full report5). Therefore, CTC with bowel preparation had sensitivity to detect adenomas 6 mm and larger comparable with colonoscopy, albeit with wider variability in estimated performance. It is unclear whether the observed variation in CTC performance was due to differences in study design, populations, bowel preparation, CTC technologies, or differences in reader experience or reading protocols.

Stool Tests

Fourteen fair- or good-quality studies (n = 59,425) that used colonoscopy reference standard in all participants reported sensitivity and specificity for 19 different types of qualitative or quantitative FITs, including 1 FIT plus sDNA test (Table 3).74, 77, 78, 80, 82, 83, 91, 94, 95, 97, 98, 100, 103, 104, 107, 108, 115 Overall, the sensitivity for CRC and advanced adenomas varied widely, including a discontinued test with very low sensitivity.100 Given the heterogeneity among FITs and their test performance, focus was placed on the performance characteristics of currently available tests evaluated in more than 1 study. Two families of FDA-cleared tests, OC-Light (qualitative, No. of studies = 3, n = 25,924) and OC FIT-CHEK (eg, OC-Sensor Diana, OC-Micro, OC-Auto) (quantitative, No. of studies = 5, n = 12,794), had relatively high sensitivity and specificity. With a single stool specimen, the lowest sensitivity demonstrated for CRC was 73% (95% CI, 48%-90%) and specificity was 96% (95% CI, 95%-96%). Similarly, the highest sensitivity with paired specificity for CRC was 88% (95% CI, 55%-99%) and 91% (95% CI, 89%-92%), respectively. In the largest studies, sensitivity ranged from 74% (95% CI, 62%-83%) for quantitative test categories (n = 9989) to 79% (95% CI, 61%-90%) for qualitative test categories (n = 18,296). In a small study (n = 770) that tested 3 stool specimens, sensitivity was 92% (95% CI, 69%-99%), but specificity was 87% (95% CI, 85%-89%). OC-Light or OCFIT-CHEK test sensitivity and specificity for advanced adenomas ranged from 22% (95% CI, 17%-28%) to 40% (95% CI, 30%-51%), and specificity ranged from 97% (95% CI, 97%-98%) to 91% (95% CI, 91%-92%). Although higher sensitivities to detect advanced adenomas were obtained for certain other FITs or by using 3 stool specimens, the corresponding specificities were lower.

Cologuard (Exact Sciences) is an FDA-approved stool test that combines stool DNA with a proprietary FIT component. One fair-quality diagnostic accuracy study (n = 9989) evaluated Cologuard compared with OC FIT-CHEK.83 In that study, Cologuard had a statistically significant higher sensitivity to detect CRC and advanced adenoma compared with OC FIT-CHEK. The higher sensitivity for CRC (92%; 95% CI, 84%-97%) and for advanced adenoma (42%; 95% CI, 39%-46%) was accompanied by lower specificity (84%; 95% CI, 84%-85% for CRC and 87%; 95% CI, 86%-87% for advanced adenoma). In our active surveillance of the literature, we identified 1 additional diagnostic accuracy study of FIT plus sDNA (n = 661) in asymptomatic Alaska Native adults.9 This study was not powered to find a difference in detection of CRC; nonetheless, findings were generally consistent with the included study on FIT plus sDNA.83

Harms of Screening

Key Question 3a. What are the adverse effects of the different screening tests (either as single application or in a screening program)?
Key Question 3b. Do adverse effects vary by important subpopulations (eg, age)?

Ninety-eight fair- or good-quality studies27, 29, 37-39, 45, 48, 64, 66, 77, 78, 81, 82, 85-87, 89, 92, 93, 97, 99, 101, 116-191 in 113 articles27, 29, 34, 37-39, 44, 45, 48, 50, 53, 64, 66, 70, 77, 78, 81, 82, 85-87, 89, 92, 93, 97, 99, 101, 114, 116-200 were included that evaluated the harms of CRC screening. These studies included 14 studies of screening programs using stool testing or SIG, 55 studies of colonoscopy in asymptomatic adults,37, 45, 77, 78, 85, 97, 101, 116, 117, 119-124, 126, 128-131, 133, 136, 140, 142, 144, 147, 148, 150, 151, 153-156, 158, 159, 161-163, 170-178, 180-183, 187-190 18 studies of screening SIG,27, 38, 39, 43, 48, 50, 66, 143, 146, 151, 157, 162, 176, 183, 185, 186, 191-194, 200 and 15 studies of screening CTC in asymptomatic adults.45, 81, 82, 85, 87, 89, 101, 118, 135, 145, 150, 162, 169, 179 Twelve CTC studies provided estimates of radiation exposure per examination,81, 82, 85-87, 89, 93, 99, 101, 118, 135, 162 and another 21 CTC studies reported information on extracolonic findings.45, 85, 99, 101, 114, 125, 127, 134, 137-139, 141, 150, 152, 160, 164, 166-168, 184, 195, 198

Endoscopy Harms

Approximately half of colonoscopy harms studies (29/55 studies) were in explicitly screening or asymptomatic populations (eTable 2 in the Supplement). By pooling 26 studies (n = 3,414,108) in screening populations or generally asymptomatic persons,37, 45, 77, 78, 85, 97, 101, 120, 121, 124, 126, 130, 131, 136, 150, 156, 163, 170, 174, 176, 180-182, 188-190 it was estimated that the risk of perforations from colonoscopy was 4 in 10,000 procedures (95% CI, 2-5 in 10,000; I2 = 86%) (Figure 4). On the basis of 22 of those studies (n = 3,347,101),37, 45, 77, 85, 97, 101, 120, 121, 124, 126, 130, 131, 156, 163, 170, 174, 180-182, 188-190 it was estimated that the risk of major bleeding from colonoscopy was 8 in 10,000 procedures (95% CI, 5-14 in 10,000; I2 = 97%) (Figure 5). Only eight studies (n = 204,614) explicitly reported if perforation or major bleeding was related to polypectomy or biopsy.45, 85, 120, 136, 158, 173, 177, 178 Based on this limited subset of studies reporting adequate information, 36% (15/42) of perforations and 96% (49/51) of major bleeding events were from polypectomy.

All 18 SIG harms studies were conducted in general-risk screening populations (eTable 3 in the Supplement). Based on the results of 16 studies (n = 329,698),38, 39, 43, 48, 50, 66, 143, 146, 151, 157, 176, 183, 185, 186, 191, 192 perforations from SIG in average-risk screening populations were relatively uncommon: the pooled point estimate was 1 in 10,000 procedures (95% CI, 0.4-1.4 in 10,000; I2 = 18.4%). In 10 studies (n = 137,987),27, 38, 48, 50, 66, 143, 146, 157, 185, 186 major bleeding episodes from SIG were also relatively uncommon, with a pooled point estimate of 2 in 10,000 procedures (95% CI, 0.7-4 in 10,000; I2 = 52.5%) (Figure 6 and Figure 7). Flexible sigmoidoscopy, however, may require follow-up diagnostic or therapeutic colonoscopy. From 5 SIG screening trials, the pooled estimate was 14 perforations per 10,000 (95% CI, 9-26 in 10,000) and 34 major bleeds per 10,00 (95% CI, 5-63 in 10,000) for follow-up colonoscopy for positive screening SIG from 4 trials.

Other serious harms from endoscopy were not routinely reported or consistently defined. Only 2 studies compared harms other than perforation and bleeding in persons who had a colonoscopy vs those who had not.180, 187 Both of these studies found no statistically significant higher risks of serious harms (including myocardial infarction, cerebrovascular accident, other cardiovascular events, and mortality) attributable to colonoscopy. Because of reporting bias around serious harms other than perforation and bleeding, as well as the lack of evidence for other serious harms attributable to colonoscopy in the few studies with control groups, these data were not quantitatively pooled.

Nineteen studies examined differential harms of colonoscopy by age group.116, 119, 122, 123, 128, 129, 131, 136, 140, 154, 156, 159, 161, 170, 172, 174, 187, 189, 190 These studies generally found increasing rates of serious adverse events with increasing age, including perforation and bleeding.

CTC Harms

Fifteen fair- or good-quality studies addressed serious adverse effects of screening CTC (eTable 4 in the Supplement).45, 81, 82, 85, 87, 89, 101, 118, 135, 145, 150, 162, 169, 179 Evidence suggested little to no risk of serious adverse events, including perforation, from CTC based on 11 prospective studies (n = 10,272) performed in screening populations.45, 81, 82, 85, 87, 89, 101, 118, 135, 145, 150, 162, 169, 179

Many of the CTC studies in this review did not report actual radiation exposure or provide sufficient information to calculate it. Based on 4 included diagnostic accuracy studies of CTC,81, 82, 85, 101 the estimated radiation dose for 1 full-screening CTC examination (dual positioning supine and prone) was about 4.5 to 7 mSv. In 3 additional recent CTC screening studies118, 135, 162 (2004–2008), the estimated radiation dose decreased to a range of 1 to less than 5 mSv.

CTC Extracolonic Findings

Incidental extracolonic findings detected on CTC can be beneficial or harmful depending on the finding. Twenty-one studies (n = 38,293)45, 85, 99, 101, 125, 127, 134, 137-139, 141, 150, 152, 160, 164, 166-168, 184, 195, 198 in 22 articles45, 85, 99, 101, 114, 125, 127, 134, 137-139, 141, 150, 152, 160, 164, 166-168, 184, 195, 198 (7 studies with overlapping populations reported different types of extracolonic findings) reported on extracolonic findings in asymptomatic persons (eTable 5 in the Supplement). In general, these studies varied greatly in their ability to accurately assess follow-up and the duration of follow-up.

Overall, extracolonic findings were common, occurring in 27% to 69% of examinations. Similarly, the studies suggested a very wide range of findings needing additional workup: 5% to 37% had E3 or E4 findings, and 1.7% to 12% had E4 findings. Among the studies that also reported medical follow-up of extracolonic findings, 1.4% to 11% went on to diagnostic evaluation, which is similar to the prevalence of E4 category findings. Among studies that adequately reported subsequent treatment, only up to 3% required definitive medical or surgical treatment. Extracolonic cancers were not common, occurring in 0.5% of persons undergoing CTC examinations. In the largest series of examinations (n = 10,286), which had about 4 years of follow-up, 0.35% of examinations revealed an extracolonic malignancy, 32 of which received definitive treatment.167 Abdominal aortic aneurysms were identified in 1.4% of persons or fewer. In our active surveillance of the literature, we identified 1 additional study evaluating extracolonic findings in screening CTC (n = 7952).7 This study's population overlapped with several already included studies and reported that 2.5% of examinations had E4 category findings, consistent with findings from included studies.150, 164, 166-168, 195, 198

Return to Table of Contents

Colorectal cancer screening continues to be a necessary and active field of research. Since the 2008 USPSTF recommendation was published, 95 new studies were identified, including more evidence on (1) the effectiveness of SIG for reducing CRC mortality, (2) the test performance of screening CTC and decreasing radiation exposure from CTC, and (3) the test performance of a number of FDA-approved FITs (including1 FIT plus sDNA test). Colonoscopy, SIG, CTC, and stool testing (gFOBT, FIT, and FIT plus sDNA test) each have differing levels of evidence to support their use, ability to detect cancer and precursor lesions, and risk of serious adverse events in screening average-risk adults for CRC (Table 4).

To date, no CRC screening modality has been shown to reduce all-cause mortality. Robust data from well-conducted population-based screening RCTs have demonstrated that both Hemoccult II and SIG can reduce CRC mortality, although neither of these tests is widely used for screening in the United States. Therefore, the empirical data on the performance of CRC screening programs using modalities used in clinical practice today are limited. Expensive, large population-based RCTs of newer stool tests may not always be necessary, as evidence-based reasoning supports that screening with stool tests with sensitivity and specificity that are both as good as, or better than, Hemoccult II would result in CRC mortality reductions similar to or better than reductions shown in existing trials.201 Based on this review, stool tests that meet those requirements are available, including specific single-stool sample FITs. However, FITs are not homogenous: they use different assays and have different diagnostic performance levels. The FDA-approved OC-Light and OC FIT-CHEK tests have the most evidence to support their use. Stool tests that maximize sensitivity (eg, FIT plus sDNA test, multiple sample FITs, or quantitative FIT using lower cutoffs) have lower specificity and therefore need new trials or modeling exercises to understand the trade-off of higher false-positive findings. In addition, stool tests vary in cost; for example, the Centers for Medicare & Medicaid Services reimbursement is $23 per FIT vs $493 per FIT plus sDNA test.202

Even though its superiority in a program of screening has not been empirically established, colonoscopy remains the criterion standard for assessing the test performance of other CRC screening tests. Moreover, colonoscopy is significantly more invasive than other available tests and thus carries a greater possibility of procedural complications, as well as harms of overdiagnosis and overtreatment of smaller lesions (ie, <10 mm). Three large RCTs of screening colonoscopy in average-risk adults are underway and will provide information about the long-term CRC incidence and mortality outcomes: the Northern European Initiative on Colorectal Cancer (NordICC) trial, comparing screening colonoscopy with usual care (estimated primary completion date, June 2026)203; COLONPREV, comparing colonoscopy with biennial FIT in Spain (estimated primary completion date, November 2021)37, 204, 205; and CONFIRM, comparing colonoscopy with annual FIT in the United States (estimated primary completion date, September 2027).206

Evidence continues to accrue that CTC adequately detects CRC and large potential precursor lesions. Although the risk of immediate harms from screening CTC (eg, bowel perforation from insufflation) is very low, it is unclear what (if any) true harm is posed by cumulative exposure to low-dose radiation or detection of extracolonic findings. Although the radiation dose appears to be decreasing over time due to technological and protocol advancements, it still ranges as high as 7 mSv per examination (dual positioning). Given that the average amount of radiation one is exposed to from background sources in the United States is about 3 mSv per year,207 ionizing radiation from a single CTC examination is low. However, current expert recommendations are to repeat CTC every 5 years, and even low doses of ionizing radiation could cumulatively convey a small excess risk of cancer.208, 209 From empirical evidence to date, it remains unclear whether detection of extracolonic findings represents a net benefit or harm.

This evidence report and systematic review did not address several important issues: screening in high-risk adults (ie, those with known family history of CRC), risk assessment to tailor screening, test acceptability, availability of or access to screening tests, methods to increase screening adherence, potential harms of overdiagnosis or unnecessary polypectomy, overuse or misuse of screening, and surveillance after adenoma detection. This review was commissioned along with a separate set of microsimulation decision models from the Cancer Intervention and Surveillance Modeling Network (CISNET) that addressed other important gaps in evidence, including ages to start and stop screening, screening intervals, and targeted or tailored screening.210 The review was limited to evidence conducted in countries with the highest applicability to US practice; in addition, only articles published in English were considered for inclusion.

Unlike other routinely recommended or conducted cancer screening, there are multiple viable options for CRC screening. These options have various levels of evidence to support their use, aims (eg, to detect cancers, potential precursor lesions, or both), test acceptability and adherence, intervals of time to repeat screening, need for follow-up testing (including surveillance incurred), associated serious harms, availability in practice, cost, and advocacy for their use. This complexity is compounded by testing whose quality is more operator-dependent (eg, colonoscopy, CTC), as well as rapid technologic advancements in improving existing tests or developing new tests.

Empirical studies, trials, or well-designed cohort studies with average-risk populations are still needed to evaluate programs of screening using colonoscopy, the best-performing stool tests, and effect of CTC on cancer mortality and cancer incidence. Also needed are studies of diagnostic accuracy to confirm the screening test performance of promising stool tests based on high sensitivity to detect CRC or advanced adenomas with thus far limited reproducibility (ie, only 1 study). Diagnostic accuracy studies, particularly those evaluating new or more complex technologies, should report percentages of inadequate or indeterminate results. It is also important to understand the contribution of technological advancements to existing technology (eg, enhancements to optical colonoscopy or CTC) on test performance in average-risk adults as well as on reducing harms (eg, decreasing radiation exposure, less aggressive bowel preparation). More complete and consistent reporting regarding downstream benefits and harms from initial detection (ie, subsequent workup and definitive treatment) of C-RADS E3 and E4 findings need to be published in observational studies or trials with longer-term follow-up. Data are still needed on the differential uptake of and adherence to screening modalities and on continued adherence to repeated rounds of screening and diagnostic follow-up to screening over longer periods.

Return to Table of Contents

Colonoscopy, flexible sigmoidoscopy, CTC, and various stool tests have differing levels of evidence to support their use in CRC screening, ability to detect CRC and precursor lesions, and risk of serious adverse events in average-risk adults. Although CRC screening has a large body of supporting evidence, additional research is still needed to weigh the relative benefits and harms of each test in within a program of screening.

Return to Table of Contents

Source: This article was first published in the Journal of the American Medical Association on June 15, 2016 (JAMA. 2016;315(23)2576-94).

Conflict of Interest Disclosures: All authors have completed and submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest and none were reported.

Role of the Funder/Sponsor: Investigators worked with USPSTF members and AHRQ staff to develop the scope, analytic framework, and key questions for this review. AHRQ staff provided project oversight; reviewed the report to ensure that the analysis met methodological standards; and distributed the draft for peer review. AHRQ reviewed and approved the manuscript before submission, but had no role in the design and conduct of the study including study selection, quality assessment, analysis, and interpretation of the data; preparation of the manuscript; and decision to submit the manuscript for publication.

Additional Contributions: We gratefully acknowledge the following individuals for their contributions to this project: Smyth Lai, MLS; Kevin Lutz, MFA; and Elizabeth Hess, MA, ELS(D), at the Kaiser Permanente Center for Health Research; Jennifer Croswell, MD, MPH, at the Agency for Healthcare Research and Quality; and current and former members of the US Preventive Services Task Force who contributed to topic deliberations. USPSTF members, peer reviewers, and federal partner reviewers did not receive financial compensation for their contributions.

Additional Information: A draft version of this evidence report underwent external peer review from 6 content experts (James Allison, MD, University of California, San Francisco; Samir Gupta, MD, MSCS, University of California, San Diego; Theodore R. Levin, MD, Kaiser Permanente; David Lieberman, MD, Oregon Health & Science University; Perry Pickhardt, MD, MPH, University of Wisconsin; David Ransohoff, MD, University of North Carolina at Chapel Hill) and 4 federal partners: Centers for Disease Control and Prevention, National Institutes of Health, US Department of Veterans Affairs, and Indian Health Service. Comments were presented to the USPSTF during its deliberation of the evidence and were considered in preparing the final evidence review.

Return to Table of Contents

1. Cancer Facts and Figures 2013. American Cancer Society. http://www.cancer.org/research/cancerfactsstatistics/cancerfactsfigures2013/. Accessed May 24, 2016.
2. Centers for Disease Control and Prevention (CDC). Vital signs: colorectal cancer screening test use: United States, 2012. MMWR Morb Mortal Wkly Rep. 2013;62(44):881-888.
3. US Preventive Services Task Force. Screening for colorectal cancer: US Preventive Services Task Force recommendation statement. Ann Intern Med. 2008;149(9):627-637.
4. Levin B, Lieberman DA, McFarland B, et al; American Cancer Society Colorectal Cancer Advisory Group; US Multi-Society Task Force; American College of Radiology Colon Cancer Committee. Screening and surveillance for the early detection of colorectal cancer and adenomatous polyps, 2008: a joint guideline from the American Cancer Society, the US Multi-Society Task Force on Colorectal Cancer, and the American College of Radiology. CA Cancer J Clin. 2008;58(3):130-160.
5. Lin JS, Piper MA, Perdue LA, et al. Screening for Colorectal Cancer: A Systematic Evidence Review for the US Preventive Services Task Force. Evidence Synthesis No. 135. AHRQ Publication 14-05203-EF-1. Rockville, MD: Agency for Healthcare Research and Quality; 2016.
6. Whitlock EP, Lin J, Liles E, et al. Screening for Colorectal Cancer: An Updated Systematic Review. Rockville, MD: Agency for Healthcare Research and Quality; 2008.
7. Pooler BD, Kim DH, Pickhardt PJ. Potentially important extracolonic findings at screening CT colonography: incidence and outcomes data from a clinical screening program. AJR Am J Roentgenol. 2016;206(2):313-318.
8. Sali L, Mascalchi M, Falchini M, et al; SAVE Study Investigators. Reduced and full-preparation CT colonography, fecal immunochemical test, and colonoscopy for population screening of colorectal cancer: a randomized trial. J Natl Cancer Inst. 2015;108(2):108.
9. Redwood DG, Asay ED, Blake ID, et al. Stool DNA testing for screening detection of colorectal neoplasia in Alaska Native people. Mayo Clin Proc. 2016;91(1):61-70.
10. Lijmer JG, Mol BW, Heisterkamp S, et al. Empirical evidence of design-related bias in studies of diagnostic tests. JAMA. 1999;282(11):1061-1066.
11. Whiting P, Rutjes AW, Reitsma JB, Glas AS, Bossuyt PM, Kleijnen J. Sources of variation and bias in studies of diagnostic accuracy: a systematic review. Ann Intern Med. 2004;140(3):189-202.
12. Leeflang MM, Bossuyt PM, Irwig L. Diagnostic test accuracy may vary with prevalence: implications for evidence-based diagnosis. J Clin Epidemiol. 2009;62(1):5-12.
13. Ransohoff DF, Feinstein AR. Problems of spectrum and bias in evaluating the efficacy of diagnostic tests. N Engl J Med. 1978;299(17):926-930.
I 14. Rutjes AW, Reitsma JB, Di Nisio M, Smidt N, van Rijn JC, Bossuyt PM. Evidence of bias and variation in diagnostic accuracy studies. CMAJ. 2006;174(4):469-476.
15. Zalis ME, Barish MA, Choi JR, et al; Working Group on Virtual Colonoscopy. CT colonography reporting and data system: a consensus proposal. Radiology. 2005;236(1):3-9.
16. Harris RP, Helfand M, Woolf SH, et al; Methods Work Group, Third US Preventive Services Task Force. Current methods of the US Preventive Services Task Force: a review of the process. Am J Prev Med. 2001;20(3 Suppl):21-35.
17. National Institute for Health and Care Excellence. The Guidelines Manual. London, UK: National Institute for Health and Care Excellence; 2006.
18. Shea BJ, Grimshaw JM, Wells GA, et al. Development of AMSTAR: a measurement tool to assess the methodological quality of systematic reviews. BMC Med Res Methodol. 2007;7:10.
19. Wells GA, Shea B, O'Connell D, et al. The Newcastle-Ottawa Scale (NOS) for assessing the quality of nonrandomised studies in meta-analysis. Ottawa Hospital Research Institute. http://www.ohri.ca/programs/clinical_epidemiology/oxford.asp. Accessed May 25, 2016.
20. Whiting P, Rutjes AW, Reitsma JB, Bossuyt PM, Kleijnen J. The development of QUADAS: a tool for the quality assessment of studies of diagnostic accuracy included in systematic reviews. BMC Med Res Methodol. 2003;3:25.
21. Whiting PF, Rutjes AW, Westwood ME, et al; QUADAS-2 Group. QUADAS-2: a revised tool for the quality assessment of diagnostic accuracy studies. Ann Intern Med. 2011;155(8):529-536.
22. Hardy RJ, Thompson SG. A likelihood approach to meta-analysis with random effects. Stat Med. 1996;15(6):619-629.
23. Thompson SG, Sharp SJ. Explaining heterogeneity in meta-analysis: a comparison of methods. Stat Med. 1999;18(20):2693-2708.
24. Guolo A, Varin C. Package metaLik. https://cran.r-project.org/web/packages/metaLik/index.html. Accessed May 25, 2016.
25. Atkin WS, Edwards R, Kralj-Hans I, et al; UK Flexible Sigmoidoscopy Trial Investigators. Once-only flexible sigmoidoscopy screening in prevention of colorectal cancer: a multicentre randomised controlled trial. Lancet. 2010;375(9726):1624-1633.
26. Berry DP, Clarke P, Hardcastle JD, Vellacott KD. Randomized trial of the addition of flexible sigmoidoscopy to faecal occult blood testing for colorectal neoplasia population screening. Br J Surg. 1997;84(9):1274-1276.
27. Brevinge H, Lindholm E, Buntzen S, Kewenter J. Screening for colorectal neoplasia with faecal occult blood testing compared with flexible sigmoidoscopy directly in a 55-56 years' old population. Int J Colorectal Dis. 1997;12(5):291-295.
28. Faivre J, Dancourt V, Denis B, et al. Comparison between a guaiac and three immunochemical faecal occult blood tests in screening for colorectal cancer. Eur J Cancer. 2012;48(16):2969-2976.
29. Faivre J, Dancourt V, Lejeune C, et al. Reduction in colorectal cancer mortality by fecal occult blood screening in a French controlled study. Gastroenterology. 2004;126(7):1674-1680.
30. Guittet L, Bouvier V, Mariotte N, et al. Comparison of a guaiac and an immunochemical faecal occult blood test for the detection of colonic lesions according to lesion type and location. Br J Cancer. 2009;100(8):1230-1235.
31. Hamza S, Dancourt V, Lejeune C, Bidan JM, Lepage C, Faivre J. Diagnostic yield of a one sample immunochemical test at different cut-off values in an organised screening programme for colorectal cancer. Eur J Cancer. 2013;49(12):2727-2733.
32. Hol L, van Leerdam ME, van Ballegooijen M, et al. Screening for colorectal cancer: randomised trial comparing guaiac-based and immunochemical faecal occult blood testing and flexible sigmoidoscopy. Gut. 2010;59(1):62-68.
33. Kronborg O, Jørgensen OD, Fenger C, Rasmussen M. Randomized study of biennial screening with a faecal occult blood test: results after nine screening rounds. Scand J Gastroenterol. 2004;39(9):846-851.
34. Lindholm E, Brevinge H, Haglind E. Survival benefit in a randomized clinical trial of faecal occult blood screening for colorectal cancer. Br J Surg. 2008;95(8):1029-1036.
35. Malila N, Palva T, Malminiemi O, et al. Coverage and performance of colorectal cancer screening with the faecal occult blood test in Finland. J Med Screen. 2011;18(1):18-23.
36. Nishihara R, Wu K, Lochhead P, et al. Long-term colorectal-cancer incidence and mortality after lower endoscopy. N Engl J Med. 2013;369(12):1095-1105.
37. Quintero E, Castells A, Bujanda L, et al; COLONPREV Study Investigators. Colonoscopy versus fecal immunochemical testing in colorectal-cancer screening. N Engl J Med. 2012;366(8):697-706.
38. Rasmussen M, Kronborg O, Fenger C, Jørgensen OD. Possible advantages and drawbacks of adding flexible sigmoidoscopy to Hemoccult-II in screening for colorectal cancer: a randomized study. Scand J Gastroenterol. 1999;34(1):73-78.
39. Schoen RE, Pinsky PF, Weissfeld JL, et al; PLCO Project Team. Colorectal-cancer incidence and mortality with screening flexible sigmoidoscopy. N Engl J Med. 2012;366(25):2345-2357.
40. Scholefield JH, Moss SM, Mangham CM, Whynes DK, Hardcastle JD. Nottingham trial of faecal occult blood testing for colorectal cancer: a 20-year follow-up. Gut. 2012;61(7):1036-1040.
41. Segnan N, Armaroli P, Bonelli L, et al; SCORE Working Group. Once-only sigmoidoscopy in colorectal cancer screening: follow-up findings of the Italian Randomized Controlled Trial: SCORE. J Natl Cancer Inst. 2011;103(17):1310-1322.
42. Segnan N, Senore C, Andreoni B, et al; SCORE3 Working Group-Italy. Comparing attendance and detection rate of colonoscopy with sigmoidoscopy and FIT for colorectal cancer screening. Gastroenterology. 2007;132(7):2304-2312.
43. Segnan N, Senore C, Andreoni B, et al; SCORE2 Working Group-Italy. Randomized trial of different screening strategies for colorectal cancer: patient response and detection rates. J Natl Cancer Inst. 2005;97(5):347-357.
44. Shaukat A, Mongin SJ, Geisser MS, et al. Long-term mortality after screening for colorectal cancer. N Engl J Med. 2013;369(12):1106-1114.
45. Stoop EM, de Haan MC, de Wijkerslooth TR, et al. Participation and yield of colonoscopy versus non-cathartic CT colonography in population-based screening for colorectal cancer: a randomised controlled trial. Lancet Oncol. 2012;13(1):55-64.
46. van Roon AH, Goede SL, van Ballegooijen M, et al. Random comparison of repeated faecal immunochemical testing at different intervals for population-based colorectal cancer screening. Gut. 2013;62(3):409-415.
47. van Rossum LG, van Rijn AF, Laheij RJ, et al. Random comparison of guaiac and immunochemical fecal occult blood tests for colorectal cancer in a screening population. Gastroenterology. 2008;135(1):82-90.
48. Verne JE, Aubrey R, Love SB, Talbot IC, Northover JM. Population based randomized study of uptake and yield of screening by flexible sigmoidoscopy compared with screening by faecal occult blood testing. BMJ. 1998;317(7152):182-185.
49. Zubero MB, Arana-Arri E, Pijoan JI, et al. Population-based colorectal cancer screening: comparison of two fecal occult blood test. Front Pharmacol. 2014;4:175.
50. Atkin WS, Cook CF, Cuzick J, Edwards R, Northover JM, Wardle J; UK Flexible Sigmoidoscopy Screening Trial Investigators. Single flexible sigmoidoscopy screening to prevent colorectal cancer: baseline findings of a UK multicentre randomised trial. Lancet. 2002;359(9314):1291-1300.
51. Atkin WS, Cuzick J, Northover JM, Whynes DK.Prevention of colorectal cancer by once-only sigmoidoscopy. Lancet. 1993;341(8847):736-740.
52. Bretthauer M, Gondal G, Larsen K, et al. Design, organization and management of a controlled population screening study for detection of colorectal neoplasia: attendance rates in the NORCCAP study (Norwegian Colorectal Cancer Prevention). Scand J Gastroenterol. 2002;37(5):568-573.
53. de Wijkerslooth TR, de Haan MC, Stoop EM, et al. Study protocol: population screening for colorectal cancer by colonoscopy or CT colonography: a randomized controlled trial. BMC Gastroenterol. 2010;10:47.
54. Denters MJ, Deutekom M, Bossuyt PM, Stroobants AK, Fockens P, Dekker E. Lower risk of advanced neoplasia among patients with a previous negative result from a fecal test for colorectal cancer. Gastroenterology. 2012;142(3):497-504.
55. Denters MJ, Deutekom M, Fockens P, Bossuyt PM, Dekker E. Implementation of population screening for colorectal cancer by repeated fecal occult blood test in the Netherlands. BMC Gastroenterol. 2009;9:28.
56. Faivre J, Dancourt V, Manfredi S, et al. Positivity rates and performances of immunochemical faecal occult blood tests at different cut-off levels within a colorectal cancer screening programme. Dig Liver Dis. 2012;44(8):700-704.
57. Guittet L, Bouvier V, Guillaume E, et al. Colorectal cancer screening: why immunochemical faecal occult blood test performs as well with either one or two samples. Dig Liver Dis. 2012;44(8):694-699.
58. Guittet L, Bouvier V, Mariotte N, et al. Performance of immunochemical faecal occult blood test in colorectal cancer screening in average-risk population according to positivity threshold and number of samples. Int J Cancer. 2009;125(5):1127-1133.
59. Hardcastle JD, Chamberlain JO, Robinson MH, et al. Randomised controlled trial of faecal-occult-blood screening for colorectal cancer. Lancet. 1996;348(9040):1472-1477.
60. Holme Ø, Løberg M, Kalager M, et al. Effect of flexible sigmoidoscopy screening on colorectal cancer incidence and mortality: a randomized clinical trial. JAMA. 2014;312(6):606-615.
61. Kewenter J, Brevinge H, Engarås B, Haglind E, Ahrén C. Results of screening, rescreening, and follow-up in a prospective randomized study for detection of colorectal cancer by fecal occult blood testing: results for 68,308 subjects. Scand J Gastroenterol. 1994;29(5):468-473.
62. Malila N, Oivanen T, Malminiemi O, Hakama M. Test, episode, and programme sensitivities of screening for colorectal cancer as a public health policy in Finland: experimental design. BMJ. 2008;337:a2261.
63. Mandel JS, Church TR, Bond JH, et al. The effect of fecal occult-blood screening on the incidence of colorectal cancer. N Engl J Med. 2000;343(22):1603-1607.
64. Mandel JS, Bond JH, Church TR, et al. Reducing mortality from colorectal cancer by screening for fecal occult blood: Minnesota Colon Cancer Control Study. N Engl J Med. 1993;328(19):1365-1371.
65. Parra-Blanco A, Nicolas-Perez D, Gimeno-Garcia A, et al. The timing of bowel preparation before colonoscopy determines the quality of cleansing, and is a significant factor contributing to the detection of flat lesions: a randomized study. World J Gastroenterol. 2006;12(38):6161-6166.
66. Segnan N, Senore C, Andreoni B, et al; SCORE Working Group-Italy. Baseline findings of the Italian multicenter randomized controlled trial of "once-only sigmoidoscopy": SCORE. J Natl Cancer Inst. 2002;94(23):1763-1772.
67. Thomas W, White CM, Mah J, Geisser MS, Church TR, Mandel JS. Longitudinal compliance with annual screening for fecal occult blood: Minnesota Colon Cancer Control Study. Am J Epidemiol. 1995;142(2):176-182.
68. Atkin WS, Cook CF, Cuzick J, Edwards R, Northover JM, Wardle J; UK Flexible Sigmoidoscopy Screening Trial Investigators. Single flexible sigmoidoscopy screening to prevent colorectal cancer: baseline findings of a UK multicentre randomised trial. Lancet. 2002;359(9314):1291-1300.
69. van Roon AH, Wilschut JA, Hol L, et al. Diagnostic yield improves with collection of 2 samples in fecal immunochemical test screening without affecting attendance. Clin Gastroenterol Hepatol. 2011;9(4):333-339.
70. van Dam L, de Wijkerslooth TR, de Haan MC, et al. Time requirements and health effects of participation in colorectal cancer screening with colonoscopy or computed tomography colonography in a randomized controlled trial. Endoscopy. 2013;45(3):182-188.
71. Weissfeld JL, Schoen RE, Pinsky PF, et al; PLCO Project Team. Flexible sigmoidoscopy in the PLCO cancer screening trial: results from the baseline screening examination of a randomized trial. J Natl Cancer Inst. 2005;97(13):989-997.
72. Allison JE, Sakoda LC, Levin TR, et al. Screening for colorectal neoplasms with new fecal occult blood tests: update on performance characteristics. J Natl Cancer Inst. 2007;99(19):1462-1470.
73. Allison JE, Tekawa IS, Ransom LJ, Adrain AL. A comparison of fecal occult-blood tests for colorectal-cancer screening. N Engl J Med. 1996;334(3):155-159.
74. Brenner H, Tao S. Superior diagnostic performance of faecal immunochemical tests for haemoglobin in a head-to-head comparison with guaiac based faecal occult blood test among 2235 participants of screening colonoscopy. Eur J Cancer. 2013;49(14):3049-3054.
75. Castiglione G, Visioli CB, Ciatto S, et al. Sensitivity of latex agglutination faecal occult blood test in the Florence District population-based colorectal cancer screening programme. Br J Cancer. 2007;96(11):1750-1754.
76. Chen LS, Yen AM, Chiu SY, Liao CS, Chen HH. Baseline faecal occult blood concentration as a predictor of incident colorectal neoplasia: longitudinal follow-up of a Taiwanese population-based colorectal cancer screening cohort. Lancet Oncol. 2011;12(6):551-558.
77. Cheng TI, Wong JM, Hong CF, et al. Colorectal cancer screening in asymptomatic adults:comparison of colonoscopy, sigmoidoscopy and fecal occult blood tests. J Formos Med Assoc. 2002;101(10):685-690.
78. Chiu HM, Lee YC, Tu CH, et al. Association between early stage colon neoplasms and false-negative results from the fecal immunochemical test. Clin Gastroenterol Hepatol. 2013;11(7):832-8.e1, 2.
79. Church TR, Wandell M, Lofton-Day C, et al; PRESEPT Clinical Study Steering Committee, Investigators and Study Team. Prospective evaluation of methylated SEPT9 in plasma for detection of asymptomatic colorectal cancer. Gut. 2014;63(2):317-325.
80. de Wijkerslooth TR, Stoop EM, Bossuyt PM, et al. Immunochemical fecal occult blood testing is equally sensitive for proximal and distal advanced neoplasia. Am J Gastroenterol. 2012;107(10):1570-1578.
81. Fletcher JG, Silva AC, Fidler JL, et al. Noncathartic CT colonography: image quality assessment and performance and in a screening cohort. AJR Am J Roentgenol. 2013;201(4):787-794.
82. Graser A, Stieber P, Nagel D, et al. Comparison of CT colonography, colonoscopy, sigmoidoscopy and faecal occult blood tests for the detection of advanced adenoma in an average risk population. Gut. 2009;58(2):241-248.
83. Imperiale TF, Ransohoff DF, Itzkowitz SH, et al. Multitarget stool DNA testing for colorectal-cancer screening. N Engl J Med. 2014;370(14):1287-1297.
84. Itoh M, Takahashi K, Nishida H, Sakagami K, Okubo T. Estimation of the optimal cut off point in a new immunological faecal occult blood test in a corporate colorectal cancer screening programme. J Med Screen. 1996;3(2):66-71.
85. Johnson CD, Chen MH, Toledano AY, et al. Accuracy of CT colonography for detection of large adenomas and cancers. N Engl J Med. 2008;359(12):1207-1217.
86. Johnson CD, Fletcher JG, MacCarty RL, et al. Effect of slice thickness and primary 2D versus 3D virtual dissection on colorectal lesion detection at CT colonography in 452 asymptomatic adults. AJR Am J Roentgenol. 2007;189(3):672-680.
87. Kim YS, Kim N, Kim SH, et al. The efficacy of intravenous contrast-enhanced 16-raw multidetector CT colonography for detecting patients with colorectal polyps in an asymptomatic population in Korea. J Clin Gastroenterol. 2008;42(7):791-798.
88. Launoy GD, Bertrand HJ, Berchi C, et al. Evaluation of an immunochemical fecal occult blood test with automated reading in screening for colorectal cancer in a general average-risk population. Int J Cancer. 2005;115(3):493-496.
89. Lefere P, Silva C, Gryspeerdt S, et al. Teleradiology based CT colonography to screen a population group of a remote island; at average risk for colorectal cancer. Eur J Radiol. 2013;82(6):e262-e267.
90. Levi Z, Birkenfeld S, Vilkin A, et al. A higher detection rate for colorectal cancer and advanced adenomatous polyp for screening with immunochemical fecal occult blood test than guaiac fecal occult blood test, despite lower compliance rate: a prospective, controlled, feasibility study. Int J Cancer. 2011;128(10):2415-2424.
91. Levy BT, Bay C, Xu Y, et al. Test characteristics of faecal immunochemical tests (FIT) compared with optical colonoscopy. J Med Screen. 2014;21(3):133-143.
92. Lin JS, Webber EM, Beil TL, Goddard KA, Whitlock EP. Fecal DNA Testing in Screening for Colorectal Cancer in Average-Risk Adults. AHRQ Publication 12-EHC022-EF. Rockville, MD: Agency for Healthcare Research and Quality; 2012.
93. Macari M, Bini EJ, Jacobs SL, et al. Colorectal polyps and cancers in asymptomatic average-risk patients: evaluation with CT colonography. Radiology. 2004;230(3):629-636.
94. Morikawa T, Kato J, Yamaji Y, Wada R, Mitsushima T, Shiratori Y. A comparison of the immunochemical fecal occult blood test and total colonoscopy in the asymptomatic population. Gastroenterology. 2005;129(2):422-428.
95. Nakama H, Yamamoto M, Kamijo N, et al. Colonoscopic evaluation of immunochemical fecal occult blood test for detection of colorectal neoplasia. Hepatogastroenterology. 1999;46(25):228-231.
96. Nakama H, Kamijo N, Abdul Fattah AS, Zhang B. Validity of immunochemical faecal occult blood screening for colorectal cancer: a follow up study. J Med Screen. 1996;3(2):63-65.
97. Ng SC, Ching JY, Chan V, et al. Diagnostic accuracy of faecal immunochemical test for screening individuals with a family history of colorectal cancer. Aliment Pharmacol Ther. 2013;38(7):835-841.
98. Park DI, Ryu S, Kim YH, et al. Comparison of guaiac-based and quantitative immunochemical fecal occult blood testing in a population at average risk undergoing colorectal cancer screening. Am J Gastroenterol. 2010;105(9):2017-2025.
99. Pickhardt PJ, Choi JR, Hwang I, et al. Computed tomographic virtual colonoscopy to screen for colorectal neoplasia in asymptomatic adults. N Engl J Med. 2003;349(23):2191-2200.
100. Sohn DK, Jeong SY, Choi HS, et al. Single immunochemical fecal occult blood test for detection of colorectal neoplasia. Cancer Res Treat. 2005;37(1):20-23.
101. Zalis ME, Blake MA, Cai W, et al. Diagnostic accuracy of laxative-free computed tomographic colonography for detection of adenomatous polyps in asymptomatic adults: a prospective evaluation. Ann Intern Med. 2012;156(10):692-702.
102. Chiang TH, Chuang SL, Chen SL, et al. Difference in performance of fecal immunochemical tests with the same hemoglobin cutoff concentration in a nationwide colorectal cancer screening program. Gastroenterology. 2014;147(6):1317-1326.
103. Hernandez V, Cubiella J, Gonzalez-Mao MC, et al; COLONPREV Study Investigators. Fecal immunochemical test accuracy in average-risk colorectal cancer screening. World J Gastroenterol. 2014;20(4):1038-1047.
104. Lee YH, Hur M, Kim H, et al. Optimal cut-off concentration for a faecal immunochemical test for haemoglobin by Hemo Techt NS-Plus C15 system for the colorectal cancer screening. Clin Chem Lab Med. 2015;53(3):e69-e71.
105. Cologuard summary of safety and effectiveness data (SSED). US Food and Drug Administration. http://www.accessdata.fda.gov/cdrh_docs/pdf13/P130017b.pdf. Accessed May 25,2016.
106. Ahlquist DA, Sargent DJ, Loprinzi CL, et al. Stool DNA and occult blood testing for screen detection of colorectal neoplasia. Ann Intern Med. 2008;149(7):441-450, W81.
107. Brenner H, Haug U, Hundt S. Inter-test agreement and quantitative cross-validation of immunochromatographical fecal occult blood tests. Int J Cancer. 2010;127(7):1643-1649.
108. Brenner H, Haug U, Hundt S. Sex differences in performance of fecal occult blood testing. Am J Gastroenterol. 2010;105(11):2457-2464.
109. Grazzini G, Castiglione G, Ciabattoni C, et al. Colorectal cancer screening programme by faecal occult blood test in Tuscany: first round results. Eur J Cancer Prev. 2004;13(1):19-26.
110. Haug U, Kuntz KM, Knudsen AB, Hundt S, Brenner H. Sensitivity of immunochemical faecal occult blood testing for detecting left- vs right-sided colorectal neoplasia. Br J Cancer. 2011;104(11):1779-1785.
111. Haug U, Hillebrand T, Bendzko P, et al. Mutant-enriched PCR and allele-specific hybridization reaction to detect K-ras mutations in stool DNA: high prevalence in a large sample of older adults. Clin Chem. 2007;53(4):787-790.
112. Hundt S, Haug U, Brenner H. Comparative evaluation of immunochemical fecal occult blood tests for colorectal adenoma detection. Ann Intern Med. 2009;150(3):162-169.
113. Imperiale TF, Ransohoff DF, Itzkowitz SH, Turnbull BA, Ross ME; Colorectal Cancer Study Group. Fecal DNA versus fecal occult blood for colorectal-cancer screening in an average-risk population. N Engl J Med. 2004;351(26):2704-2714.
114. Johnson CD, Herman BA, Chen MH, et al. The National CT Colonography Trial: assessment of accuracy in participants 65 years of age and older. Radiology. 2012;263(2):401-408.
115. Morikawa T, Kato J, Yamaji Y, et al. Sensitivity of immunochemical fecal occult blood test to small colorectal adenomas. Am J Gastroenterol. 2007;102(10):2259-2264.
116. Adeyemo A, Bannazadeh M, Riggs T, Shellnut J, Barkel D, Wasvary H. Does sedation type affect colonoscopy perforation rates? Dis Colon Rectum. 2014;57(1):110-114.
117. Adler A, Wegscheider K, Lieberman D, et al. Factors determining the quality of screening colonoscopy: a prospective study on adenoma detection rates, from 12,134 examinations (Berlin Colonoscopy Project 3, BECOP-3). Gut. 2013;62(2):236-241.
118. An S, Lee KH, Kim YH, et al. Screening CT colonography in an asymptomatic average-risk Asian population: a 2-year experience in a single institution. AJR Am J Roentgenol. 2008;191(3):W100-W106.
119. Arora G, Mannalithara A, Singh G, Gerson LB, Triadafilopoulos G. Risk of perforation from a colonoscopy in adults: a large population-based study. Gastrointest Endosc. 2009;69(3 pt 2):654-664.
120. Bair D, Pham J, Seaton MB, Arya N, Pryce M, Seaton TL. The quality of screening colonoscopies in an office-based endoscopy clinic. Can J Gastroenterol. 2009;23(1):41-47.
121. Berhane C, Denning D. Incidental finding of colorectal cancer in screening colonoscopy and its cost effectiveness. Am Surg. 2009;75(8):699-703.
122. Bielawska B, Day AG, Lieberman DA, Hookey LC. Risk factors for early colonoscopic perforation include non-gastroenterologist endoscopists: a multivariable analysis. Clin Gastroenterol Hepatol. 2014;12(1):85-92.
123. Blotière PO, Weill A, Ricordeau P, Alla F, Allemand H. Perforations and haemorrhages after colonoscopy in 2010: a study based on comprehensive French health insurance data (SNIIRAM). Clin Res Hepatol Gastroenterol. 2014;38(1):112-117.
124. Bokemeyer B, Bock H, Hüppe D, et al. Screening colonoscopy for colorectal cancer prevention: results from a German online registry on 269,000 cases. Eur J Gastroenterol Hepatol. 2009;21(6):650-655.
125. Cash BD, Riddle MS, Bhattacharya I, et al. CT colonography of a Medicare-aged population: outcomes observed in an analysis of more than 1400 patients. AJR Am J Roentgenol. 2012;199(1):W27-W34.
126. Castro G, Azrak MF, Seeff LC, Royalty J. Outpatient colonoscopy complications in the CDC's Colorectal Cancer Screening Demonstration Program: a prospective analysis. Cancer. 2013;119(Suppl 15):2849-2854.
127. Chin M, Mendelson R, Edwards J, Foster N, Forbes G. Computed tomographic colonography: prevalence, nature, and clinical significance of extracolonic findings in a community screening program. Am J Gastroenterol. 2005;100(12):2771-2776.
128. Chukmaitov A, Bradley CJ, Dahman B, Siangphoe U, Warren JL, Klabunde CN. Association of polypectomy techniques, endoscopist volume, and facility type with colonoscopy complications. Gastrointest Endosc. 2013;77(3):436-446.
129. Cooper GS, Kou TD, Rex DK. Complications following colonoscopy with anesthesia assistance: a population-based analysis. JAMA Intern Med. 2013;173(7):551-556.
130. Cotterill M, Gasparelli R, Kirby E. Colorectal cancer detection in a rural community: development of a colonoscopy screening program. Can Fam Physician. 2005;51:1224-1228.
131. Crispin A, Birkner B, Munte A, Nusko G, Mansmann U. Process quality and incidence of acute complications in a series of more than 230,000 outpatient colonoscopies. Endoscopy. 2009;41(12):1018-1025.
132. Dancourt V, Lejeune C, Lepage C, Gailliard MC, Meny B, Faivre J. Immunochemical faecal occult blood tests are superior to guaiac-based tests for the detection of colorectal neoplasms. Eur J Cancer. 2008;44(15):2254-2258.
133. Dominitz JA, Baldwin LM, Green P, Kreuter WI, Ko CW. Regional variation in anesthesia assistance during outpatient colonoscopy is not associated with differences in polyp detection or complication rates. Gastroenterology. 2013;144(2):298-306.
134. Durbin JM, Stroup SP, Altamar HO, L'esperance JO, Lacey DR, Auge BK. Genitourinary abnormalities in an asymptomatic screening population: findings on virtual colonoscopy. Clin Nephrol. 2012;77(3):204-210.
135. Edwards JT, Mendelson RM, Fritschi L, et al. Colorectal neoplasia screening with CT colonography in average-risk asymptomatic subjects: community-based study. Radiology. 2004;230(2):459-464.
136. Ferlitsch M, Reinhart K, Pramhas S, et al. Sex-specific prevalence of adenomas, advanced adenomas, and colorectal cancer in individuals undergoing screening colonoscopy. JAMA. 2011;306(12):1352-1358.
137. Flicker MS, Tsoukas AT, Hazra A, Dachman AH. Economic impact of extracolonic findings at computed tomographic colonography. J Comput Assist Tomogr. 2008;32(4):497-503.
138. Ginnerup Pedersen B, Rosenkilde M, Christiansen TE, Laurberg S. Extracolonic findings at computed tomography colonography are a challenge. Gut. 2003;52(12):1744-1747.
139. Gluecker TM, Johnson CD, Wilson LA, et al. Extracolonic findings at CT colonography: evaluation of prevalence and cost in a screening population. Gastroenterology. 2003;124(4):911-916.
140. Hamdani U, Naeem R, Haider F, et al. Risk factors for colonoscopic perforation: a population-based study of 80,118 cases. World J Gastroenterol. 2013;19(23):3596-3601.
141. Hara AK, Johnson CD, MacCarty RL, Welch TJ. Incidental extracolonic findings at CT colonography. Radiology. 2000;215(2):353-357.
142. Ho JM, Gruneir A, Fischer HD, et al. Serious events in older Ontario residents receiving bowel preparations for outpatient colonoscopy with various comorbidity profiles: a descriptive, population-based study. Can J Gastroenterol. 2012;26(7):436-440.
143. Hoff G, Thiis-Evensen E, Grotmol T, Sauar J, Vatn MH, Moen IE. Do undesirable effects of screening affect all-cause mortality in flexible sigmoidoscopy programmes? experience from the Telemark Polyp Study 1983-1996. Eur J Cancer Prev. 2001;10(2):131-137.
144. Hsieh TK, Hung L, Kang FC, Lan KM, Poon PW, So EC. Anesthesia does not increase the rate of bowel perforation during colonoscopy: a retrospective study. Acta Anaesthesiol Taiwan. 2009;47(4):162-166.
145. Iafrate F, Iussich G, Correale L, et al. Adverse events of computed tomography colonography: an Italian National Survey. Dig Liver Dis. 2013;45(8):645-650.
146. Jain A, Falzarano J, Jain A, Decker R, Okubo G, Fujiwara D. Outcome of 5,000 flexible sigmoidoscopies done by nurse endoscopists for colorectal screening in asymptomatic patients. Hawaii Med J. 2002;61(6):118-120.
147. Kamath AS, Iqbal CW, Sarr MG, et al. Colonoscopic splenic injuries: incidence and management. J Gastrointest Surg. 2009;13(12):2136-2140.
148. Kang HY, Kang HW, Kim SG, et al. Incidence and management of colonoscopic perforations in Korea. Digestion. 2008;78(4):218-223.
149. Kao KT, Jain A, Sheinbaum A. Ischemic colitis following routine screening colonoscopy: a case report. Endoscopy. 2009;41(Suppl 2):E100.
150. Kim DH, Pickhardt PJ, Taylor AJ, et al. CT colonography versus colonoscopy for the detection of advanced neoplasia. N Engl J Med. 2007;357(14):1403-1412.
151. Kim JS, Kim BW, Kim JI, et al. Endoscopic clip closure versus surgery for the treatment of iatrogenic colon perforations developed during diagnostic colonoscopy: a review of 115,285 patients. Surg Endosc. 2013;27(2):501-504.
152. Kim YS, Kim N, Kim SY, et al. Extracolonic findings in an asymptomatic screening population undergoing intravenous contrast-enhanced computed tomography colonography. J Gastroenterol Hepatol. 2008;23(7 pt 2):e49-e57.
153. Ko CW, Riffle S, Shapiro JA, et al. Incidence of minor complications and time lost from normal activities after screening or surveillance colonoscopy. Gastrointest Endosc. 2007;65(4):648-656.
154. Korman LY, Overholt BF, Box T, Winker CK. Perforation during colonoscopy in endoscopic ambulatory surgical centers. Gastrointest Endosc. 2003;58(4):554-557.
155. Layton JB, Klemmer PJ, Christiansen CF, et al. Sodium phosphate does not increase risk for acute kidney injury after routine colonoscopy, compared with polyethylene glycol. Clin Gastroenterol Hepatol. 2014;12(9):1514-1521.
156. Levin TR, Zhao W, Conell C, et al. Complications of colonoscopy in an integrated health care delivery system. Ann Intern Med. 2006;145(12):880-886.
157. Levin TR, Conell C, Shapiro JA, Chazan SG, Nadel MR, Selby JV. Complications of screening flexible sigmoidoscopy. Gastroenterology. 2002;123(6):1786-1792.
158. Loffeld RJ, Engel A, Dekkers PE. Incidence and causes of colonoscopic perforations: a single-center case series. Endoscopy. 2011;43(3):240-242.
159. Lorenzo-Zúñiga V, Moreno de Vega V, Doménech E, Mañosa M, Planas R, Boix J. Endoscopist experience as a risk factor for colonoscopic complications. Colorectal Dis. 2010;12(10):e273-e277.
160. Macari M, Nevsky G, Bonavita J, Kim DC, Megibow AJ, Babb JS. CT colonography in senior versus nonsenior patients: extracolonic findings, recommendations for additional imaging, and polyp prevalence. Radiology. 2011;259(3):767-774.
161. Mansmann U, Crispin A, Henschel V, et al. Epidemiology and quality control of 245,000 outpatient colonoscopies. Dtsch Arztebl Int. 2008;105(24):434-440.
162. Multicentre Australian Colorectal-neoplasia Screening (MACS) Group. A comparison of colorectal neoplasia screening tests: a multicentre community-based study of the impact of consumer choice. Med J Aust. 2006;184(11):546-550.
163. Nelson DB, McQuaid KR, Bond JH, Lieberman DA, Weiss DG, Johnston TK. Procedural success and complications of large-scale screening colonoscopy. Gastrointest Endosc. 2002;55(3):307-314.
164. O'Connor SD, Pickhardt PJ, Kim DH, Oliva MR, Silverman SG. Incidental finding of renal masses at unenhanced CT: prevalence and analysis of features for guiding management. AJR Am J Roentgenol. 2011;197(1):139-145.
165. Parente F, Boemo C, Ardizzoia A, et al. Outcomes and cost evaluation of the first two rounds of a colorectal cancer screening program based on immunochemical fecal occult blood test in northern Italy. Endoscopy. 2013;45(1):27-34.
166. Pickhardt PJ, Boyce CJ, Kim DH, Hinshaw LJ, Taylor AJ, Winter TC. Should small sliding hiatal hernias be reported at CT colonography? AJR Am J Roentgenol. 2011;196(4):W400-W404.
167. Pickhardt PJ, Kim DH, Meiners RJ, et al. Colorectal and extracolonic cancers detected at screening CT colonography in 10,286 asymptomatic adults. Radiology. 2010;255(1):83-88.
168. Pickhardt PJ, Kim DH, Taylor AJ, Gopal DV, Weber SM, Heise CP. Extracolonic tumors of the gastrointestinal tract detected incidentally at screening CT colonography. Dis Colon Rectum. 2007;50(1):56-63.
169. Pickhardt PJ. Incidence of colonic perforation at CT colonography: review of existing data and implications for screening of asymptomatic adults. Radiology. 2006;239(2):313-316.
170. Pox CP, Altenhofen L, Brenner H, Theilmeier A, Von Stillfried D, Schmiegel W. Efficacy of a nationwide screening colonoscopy program for colorectal cancer. Gastroenterology. 2012;142(7):1460-1467.
171. Quallick MR, Brown WR. Rectal perforation during colonoscopic retroflexion: a large, prospective experience in an academic center. Gastrointest Endosc. 2009;69(4):960-963.
172. Rabeneck L, Paszat LF, Hilsden RJ, et al. Bleeding and perforation after outpatient colonoscopy and their risk factors in usual clinical practice. Gastroenterology. 2008;135(6):1899-1906.
173. Rathgaber SW, Wick TM. Colonoscopy completion and complication rates in a community gastroenterology practice. Gastrointest Endosc. 2006;64(4):556-562.
174. Rutter CM, Johnson E, Miglioretti DL, Mandelson MT, Inadomi J, Buist DS. Adverse events after screening and follow-up colonoscopy. Cancer Causes Control. 2012;23(2):289-296.
175. Sagawa T, Kakizaki S, Iizuka H, et al. Analysis of colonoscopic perforations at a local clinic and a tertiary hospital. World J Gastroenterol. 2012;18(35):4898-4904.
176. Senore C, Ederle A, Fantin A, et al. Acceptability and side-effects of colonoscopy and sigmoidoscopy in a screening setting. J Med Screen. 2011;18(3):128-134.
177. Sieg A, Hachmoeller-Eisenbach U, Eisenbach T. Prospective evaluation of complications in outpatient GI endoscopy: a survey among German gastroenterologists. Gastrointest Endosc. 2001;53(6):620-627.
178. Singh H, Penfold RB, DeCoster C, et al. Colonoscopy and its complications across a Canadian regional health authority. Gastrointest Endosc. 2009;69(3 pt 2):665-671.
179. Sosna J, Blachar A, Amitai M, et al. Colonic perforation at CT colonography: assessment of risk in a multicenter large cohort. Radiology. 2006;239(2):457-463.
180. Stock C, Ihle P, Sieg A, Schubert I, Hoffmeister M, Brenner H. Adverse events requiring hospitalization within 30 days after outpatient screening and nonscreening colonoscopies. Gastrointest Endosc. 2013;77(3):419-429.
181. Strul H, Kariv R, Leshno M, et al. The prevalence rate and anatomic location of colorectal adenoma and cancer detected by colonoscopy in average-risk individuals aged 40-80 years. Am J Gastroenterol. 2006;101(2):255-262.
182. Suissa A, Bentur OS, Lachter J, et al. Outcome and complications of colonoscopy: a prospective multicenter study in northern Israel. Diagn Ther Endosc. 2012;2012:612542.
183. Tam MS, Abbas MA. Perforation following colorectal endoscopy: what happens beyond the endoscopy suite? Perm J. 2013;17(2):17-21.
184. Veerappan GR, Ally MR, Choi JH, Pak JS, Maydonovitch C, Wong RK. Extracolonic findings on CT colonography increases yield of colorectal cancer screening. AJR Am J Roentgenol. 2010;195(3):677-686.
185. Viiala CH, Olynyk JK. Outcomes after 10 years of a community-based flexible sigmoidoscopy screening program for colorectal carcinoma. Med J Aust. 2007;187(5):274-277.
186. Wallace MB, Kemp JA, Meyer F, et al. Screening for colorectal cancer with flexible sigmoidoscopy by nonphysician endoscopists. Am J Med. 1999;107(3):214-218.
187. Warren JL, Klabunde CN, Mariotto AB, et al. Adverse events after outpatient colonoscopy in the Medicare population. Ann Intern Med. 2009;150(12):849-857, W152.
188. Xirasagar S, Hurley TG, Sros L, Hebert JR. Quality and safety of screening colonoscopies performed by primary care physicians with standby specialist support. Med Care. 2010;48(8):703-709.
189. Zafar HM, Harhay MO, Yang J, Armstron K. Adverse events following computed tomographic colonography compared to optical colonoscopy in the elderly. Prev Med Rep. 2014;1:3-8.
190. Ko CW, Riffle S, Michaels L, et al. Serious complications within 30 days of screening and surveillance colonoscopy are uncommon. Clin Gastroenterol Hepatol. 2010;8(2):166-173.
191. Atkin WS, Hart A, Edwards R, et al. Uptake, yield of neoplasia, and adverse effects of flexible sigmoidoscopy screening. Gut. 1998;42(4):560-565.
192. Gondal G, Grotmol T, Hofstad B, Bretthauer M, Eide TJ, Hoff G. The Norwegian Colorectal Cancer Prevention (NORCCAP) screening study: baseline findings and implementations for clinical work-up in age groups 50-64 years. Scand J Gastroenterol. 2003;38(6):635-642.
193. Hoff G, Grotmol T, Skovlund E, Bretthauer M; Norwegian Colorectal Cancer Prevention Study Group. Risk of colorectal cancer seven years after flexible sigmoidoscopy screening: randomised controlled trial. BMJ. 2009;338:b1846.
194. Hoff G, Sauar J, Vatn MH, et al. Polypectomy of adenomas in the prevention of colorectal cancer: 10 years' follow-up of the Telemark Polyp Study I: a prospective, controlled population study. Scand J Gastroenterol. 1996;31(10):1006-1010.
195. Kim DH, Pickhardt PJ, Hanson ME, Hinshaw JL. CT colonography: performance and program outcome measures in an older screening population. Radiology. 2010;254(2):493-500.
196. Miles A, Wardle J, McCaffery K, Williamson S, Atkin W. The effects of colorectal cancer screening on health attitudes and practices. Cancer Epidemiol Biomarkers Prev. 2003;12(7):651-655.
197. Pickhardt PJ, Kim DH, Robbins JB. Flat (nonpolypoid) colorectal lesions identified at CT colonography in a US screening population. Acad Radiol. 2010;17(6):784-790.
198. Pickhardt PJ, Hanson ME, Vanness DJ, et al. Unsuspected extracolonic findings at screening CT colonography: clinical and economic impact. Radiology. 2008;249(1):151-159.
199. Regula J, Polkowski M. CT colonography versus colonoscopy for the detection of advanced neoplasia. N Engl J Med. 2008;358(1):88-89.
200. Thiis-Evensen E, Hoff GS, Sauar J, Langmark F, Majak BM, Vatn MH. Population-based surveillance by colonoscopy: effect on the incidence of colorectal cancer: Telemark Polyp Study I. Scand J Gastroenterol. 1999;34(4):414-420.
201. Lord SJ, Irwig L, Simes RJ. When is measuring sensitivity and specificity sufficient to evaluate a diagnostic test, and when do we need randomized trials? Ann Intern Med. 2006;144(11):850-855.
202. Exact Sciences: additional update on CMS reimbursement for Cologuard [press release]. http://investor.exactsciences.com/investor-relations/press-releases/press-release-details/2015/Exact-Sciences-Additional-Update-on-CMS-Reimbursement-for-Cologuard/default.aspx. Accessed May 25, 2016.
203. Kaminski MF, Bretthauer M, Zauber AG, et al. The NordICC Study: rationale and design of a randomized trial on colonoscopy screening for colorectal cancer. Endoscopy. 2012;44(7):695-702.
204. Castells A, Quintero E. Programmatic screening for colorectal cancer: the COLONPREV study. Dig Dis Sci. 2015;60(3):672-680.
205. Álvarez C, Andreu M, Castells A, et al; ColonPrev study investigators. Relationship of colonoscopy-detected serrated polyps with synchronous advanced neoplasia in average-risk individuals. Gastrointest Endosc. 2013;78(2):333-341.
206. Colonoscopy Versus Fecal Immunochemical Test in Reducing Mortality From Colorectal Cancer (CONFIRM). ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT01239082. Accessed May 25, 2016.
207. Committee to Assess Health Risks from Exposure to Low Levels of Ionizing Radiation, Board on Radiation Effects Research, Division on Earth and Life Studies, National Research Council of the National Academies. Health Risks From Exposure to Low Levels of Ionizing Radiation: BEIR VII Phase 2. Washington, DC: National Academies Press; 2006.
208. Cardis E, Vrijheid M, Blettner M, et al. Risk of cancer after low doses of ionising radiation: retrospective cohort study in 15 countries. BMJ. 2005;331(7508):77.
209. Brenner DJ, Hall EJ. Computed tomography: an increasing source of radiation exposure. N Engl J Med. 2007;357(22):2277-2284.
210. Knudsen AB, Zauber AG, Rutter CM, et al. Estimation of benefits, burden, and harms of colorectal cancer screening strategies: modeling study for the US Preventive Services Task Force. JAMA. doi:10.1001/jama.2016.6828.

Return to Table of Contents


CTC indicates computed tomographic colonography; FIT, fecal immunochemical test; SIG, flexible sigmoidoscopy; gFOBT, guaiac-based fecal occult blood test; KQ, key question; mSEPT9, circulating methylated septin 9 gene DNA.

Text Description.

Figure 1 is an analytic framework for the key questions of this systematic evidence review that depicts the events that adults age 40 years or older could experience while undergoing screening for colorectal cancer. In general, the figure illustrates how screening for colorectal cancer with colonoscopy, flexible sigmoidoscopy, CT colonography, fecal tests (gFOBT, FIT, fecal DNA), or a blood test (mSEPT9) could lead to detection of polyps or cancer and decrease colorectal cancer incidence and mortality and all-cause mortality.

Return to Table of Contents

KQ indicates key question; MA,meta-analysis; SER, systematic evidence review; SIG, flexible sigmoidoscopy; USPSTF, US Preventive Services Task Force.

a Details about reasons for exclusion are as follows. Relevance: study aim not relevant. Design: study did not use an included design. Setting: study was not conducted in a country relevant to US practice. Population: study was not conducted in an average-risk population. Outcomes: study did not have relevant outcomes or had incomplete outcomes. Intervention: study used an excluded intervention or screening approach. Quality: study did not meet criteria for fair or good quality. Simulated SIG: study used the distal colon results from a colonoscopy to simulate flexible sigmoidoscopy. SER-MA outdated: study was an existing systematic evidence review with an out-of-date meta-analysis.

Text Description.

Figure 2 displays the literature search results and article flow for the systematic evidence review. A total of 11,189 articles were identified through database searching, 372 through selected systematic evidence reviews and other sources, and 68 from the 2008 USPSTF evidence review. 8492 articles were screened and 696 full-text articles were assessed for eligibility. 47 articles (25 studies) were included for key question 1; 44 articles (33 studies) were included for key question 2; and 113 articles (98 studies) were included for key question 3.

Return to Table of Contents

Screening Tool and Reference Qualityb Country Patient
Age
Range, y
No. of
Participants
No. of
Screening
Rounds
Screening
Interval,
y
Follow-up
Period, yc
Positive
Screening
Results,
%d
CRC, %e No. of
CRC Deaths/
100,000
Person-Years
CRC
Mortality,
RR (95% CI)
Flexible Sigmoidoscopy
NORCCAP,60
2014
Fair Norway 50-64 Intervention: 20,572
Control: 78,220
1 NA 11.0 20.4 1.4 Intervention: 31
Control: 43
0.80 (0.62-1.04)f,g
PLCO,39, 71
2012
Fair United States 55-74 Intervention: 77,445
Control: 77,455
2 3-5 12.1 32.9 1.5 Intervention: 29
Control: 39
0.74 (0.63-0.87)
SCORE,41, 66
2011
Fair Italy 55-64 Intervention: 17,136
Control: 17,136
1 NA 11.4 8.6 1.6 Intervention: 35
Control: 44
0.78 (0.56-1.08)
UKFSST,25, 50
2010
Fair United
Kingdom
55-64 Intervention:
57,099
Control: 112,939
1 NA 11.2 5.2 1.5 Intervention: 30
Control: 44
0.69 (0.59-0.80)f
Hemoccult IIh
Minnesota Colon Cancer Control Study,44, 63, 64, 67
2013
Good United States 50-80 Intervention (biennial): 15,587
Control: 15,394
6 2 30 NRi 2.9j Intervention: 50
Control: 63
0.78 (0.65-0.93)
Intervention (annual): 15,570
Control: 15,394
11 1 30 NRi 2.9j Intervention: 42
Control: 63
0.68 (0.56-0.82)
Nottingham,40, 59
2012
Good United Kingdom 45-74 Intervention: 76,056
Control: 75,919
3-5 2 28 2.1 3.0 Intervention: 91
Control: 100
0.91 (0.84-0.98)
Göteborg,34
2008
Fair Sweden 60-64 Intervention: 34,144
Control: 34,164
2-3 1-9 19 3.8i 2.2 Intervention: 53
Control: 64
0.84 (0.71-0.99)
Burgundy,29
2004
Fair France 45-74 Intervention: 45,642
Control: 45,557
6 2 11 2.1 1.5 Intervention: 53
Control: 64
0.84 (0.71-0.99)
Funen,33
2004
Good Denmark 45-75 Intervention: 30,967
Control: 30,966
9 2 17 1.0 2.8 Intervention: 84
Control: 100
0.84 (0.73-0.96)

Abbreviations: CRC, colorectal cancer; NA, not applicable; NR, not reported; RCT, randomized clinical trial; RR, relative risk.

a The comparator for each of these RCTs was a control group that was not offered any CRC screening.
b Assessed using criteria from the US Preventive Services Task Force.16
c Median follow-up time for flexible sigmoidoscopy, longest follow-up time for Hemoccult II.
d For flexible sigmoidoscopy, this refers to the percentage of patients who were referred to colonoscopy out of those who received their flexible sigmoidoscopy. For Hemoccult II, it refers to the percentage of patients who tested positive out of those who took the test in round 1 only.
e The percentage of CRC cases that occurred during the follow-up period among those included in the study at baseline.
f Calculated RR (not study reported).
g NORCCAP reported a statistically significant decrease in CRC mortality for the screened group vs the control (hazard ratio, 0.73; 95% CI, 0.56-0.94; P = 0.02). To present consistent results across studies, we show unadjusted results here.
h One trial in Finland has not reported CRC mortality.35, 62
i Study included rehydrated tests: in Göteborg, 91.7% of all tests were rehydrated; in the Minnesota Colon Cancer Control Study, 82.5% of all tests were rehydrated.
j Refers to all 3 groups of the trial (annual, biennial, and control).

Return to Table of Contents

Control indicates no colorectal cancer screening; IRR, incidence rate ratio; NORCCAP, Norwegian Colorectal Cancer Prevention; PLCO, Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial; SCORE, Screening for Colon Rectum; UKFSST, UK Flexible Sigmoidoscopy Screening Trial.

Text Description.

Figure 3 displays a forest plot of the effect of screening with flexible sigmoidoscopy on colorectal cancer mortality. The pooled incidence rate ratio for the four studies (NORCCAP, PLCO, SCORE, UKFSST) was 0.73, with a 95% confidence interval of 0.66 to 0.82.

Return to Table of Contents

Study Qualitya Study Site Cohort Size Mean Patient Age, y Fecal Tagb No. of Readers,
Trainingc
Reading Strategyd Reference Standard Adenoma ≥6 mm, % (95% CI) Adenoma ≥10 mm, % (95% CI)
Sensitivity Specificity Sensitivity Specificity
With Bowel Preparation
Lefere et al,89
2013
Fair Portugal 496 60 Yes 1, >5000
exams
3D (with 2D) Repeat colonoscopy if indicated 98 (91-100) 91 (88-93) NR NR
Graser et al,82
2009
Good Germany 307 60 No 3, >300 exams 3D (with 2D Colonoscopy, segmental unblindinge 91 (80-97) 93 (90-96) 92 (76-98) 98 (96-99)
Johnson et al,85
2008 (ACRIN)
Good United States 2531 58 Yes 15, >500 exams 3D (with 2D Repeat colonoscopy if indicated 78 (72-83) 90 (88-91) 90 (83-95) 86 (85-87)
Kim et al,87 Fair South
Korea
241 58 No 2, >100 exams 2D (with 3D) Single colonoscopy 68 (55-80)g 89 (84-93)g 87 (64-97)h 97 (95-99)h
Johnson et al,86
2007
Fair United States 452 65 No 3, >1000
exams
3D (with 2D)i Single colonoscopy NR NR 67 (45-84) 98 (96-99)
Macari et al,93
2004
Fair United States 68 55 No 1, 5 y NR Single colonoscopy NR NR 100 (46-100)j 98 (93-100)j
Pickhardt et al,99
2003
Good United States 1233 58 Yes 6, >25 exams 3D (with 2D) Colonoscopy, segmental unblindinge 89 (83-93) 80 (77-82) 94 (84-98) 96 (95-97)
Without Bowel Preparation
Fletcher et al,81
2013
Good United States 564 NR Yes 2, >150 exams 2D and 3D Single colonoscopy 75 (59-87) 92 (90-94) 67 (42-86) 97 (96-98)
Zalis et al,101
2012
Good United States 605 60 Yes 3, >200 exams 2D and 3D Colonoscopy, segmental unblindinge 58 (46-69) 88 (85-91) 90 (70-98) 85 (82-88)

Abbreviations: CT, computed tomographic; exams, examinations; NR, not reported.

a Quality assessed using criteria from Quality Assessment of Diagnostic Accuracy Studies (QUADAS)20 and QUADAS 221 instrument.
b Oral ingestion of high-density oral contrast agent so that residual colonic contents can be differentiated from soft tissue density polyps.
c Number of examinations or years of training required by each reader or radiologist.
d Reader or radiologist procedure for using 2- and 3-dimensional images.
e CT colonography enhanced colonoscopy, in which endoscopist was shown respective segment of colon on CT colonography after examination of segment by colonoscopy.
f National CT Colonography Trial.
g Any histology ≥6 mm; sensitivity for adenomas ≥6 mm, 72.7% (95% CI, 58.4%-84.1%); specificity not reported.
h Any histology ≥10 mm; sensitivity for adenomas ≥10 mm, 90.0% (95% CI, 61.9%-99.0%); specificity not reported.
i Study evaluated different reading strategies; data shown reflect primary 3D strategy.
j Any histology ≥10 mm.

Return to Table of Contents

Study Qualitya Mean Patient Age, y Cohort Size Test Family Name Cutoff, μg
Hb/g Feces
No. of Stool
Samples per Person
CRC, % CRC Advanced Adenomas
Sensitivity,
% (95% CI)
Specificity,
% (95% CI)
Sensitivity,
% (95% CI)
Specificity,
% (95% CI)
Qualitative FIT Tests
Levy et al,91
2014b
Fair 56.9 308 Clearview (cassette) 6 NR NR NR NR NR NR
44 Clearview (test strip) 50 NR NR NR NR NR NR
217 OC-Light 10 NR NR NR NR NR NR
52 QuickVue 50 NR NR NR NR NR NR
Chiu et al,78
2013
Good   18,296 OC-Light 10 1 0.15 79 (61-90) 93 (92-93) 28 (25-32) 94 (93-94)
Ng et al,97
2013
Fair 57.7 4539 Hemosure 50 NR 0.48 54 (32-74) 89 (88-90) 37 (30-44) 91 (90-91)
Brenner
et al,107
2010
Good 63c 1319 Bionexia Hb NR NR 0.8 NR NR 52 (44-61) 80 (77-82)
1328 Bionexia Hb-Hp NR NR NR NR 72 (63-79) 56 (54-59)
1330 FOB advanced NR NR NR NR 27 (20-35) 91 (90-93)
1319 ImmoCARE-C 30 NR NR NR 25 (18-33) 96 (95-97)
1330 PreventID CC NR NR NR NR 49 (41-58) 81 (79-84)
1330 QuickVue NR NR NR NR 56 (48-64) 68 (65-70)
Cheng et al,77
2002
Fair 46.8 7411 OC-Light 10 NR 0.22 88 (66-97) 91 (90-92) 40 (30-51) 91 (91-92)
Nakama
et al,95
1999
Fair NR 4611 MonoHaem ~1000 1 0.39 56 (33-76) 97 (96-97) NR NR
~1000 2 83 (62-95) 95 (95-96) NR NR
~1000 3 89 (69-98) 93 (92-94) NR NR
Quantitative FIT Tests
Hernandez
et al,103
2014
Good 57.6 779 OC FIT-CHEK 10 1 0.6 100 (62-100) 92 (90-94) NR NR
20 1 100 (62-100) 94 (92-95) NR NR
10 2 100 (62-100) 88 (85-90) NR NR
20 2 100 (62-100) 90 (88-92) NR NR
Imperiale
et al,83
2014
Fair 64.2 9989 OC FIT-CHEK 20 1 0.65 74 (62-83) 93 (93-94) 24 (21-27) 95 (94-95)
Cologuard (FIT plus stool DNA test) NA 1 92 (84-97) 84 (84-85) 42 (39-46) 87 (86-87)
Lee et al,104
2014
Good 58c NR Hemo Techt NS-Plus C system 6.3 NR NR 86 (57-98) 94 (93-95) NR NR
Brenner
and Tao,74
2013
Good 62.7 2220 OC FIT-CHEK 20 1 0.67 73 (48-90) 96 (95-96) 22 (17-28) 97 (97-98)
2220 RIDASCREEN Hb 2 1 60 (35-81) 95 (94-96) 21 (16-27) 97 (96-98)
2235 RIDASCREEN Hb-Hp 2 1 53 (29-76) 95 (94-96) 18 (13-24) 97 (96-98)
de Wijkerslooth et al,80
2012
Good 60c 1256 OC FIT-CHEK 10 1 0.64 88 (55-99) 91 (89-92) 34 (26-43) 93 (92-95)
20 1 75 (41-94) 95 (93-96) 28 (20-37) 97 (96-98)
Park et al,98
2010
Fair 59.3 770 OC FIT-CHEK 10 3 1.7 92 (69-99) 87 (85-89) 44 (32-57) 89.8 (87.4-91.9)
757 20 3 92.3 (69.3-99.2) 90.1 (87.8-92.1) 33.9 (22.8-46.5) 92.1 (89.9-94.0)
Graser et al,82
2009
Good 60.5 285 FOB Gold NR 2 0.33 100 (14.7-100) NR 29.2 (14.1-48.9) 85.8 (81.1-89.6)
Morikawa et al,94
2005
Fair 48 21,805 Magstream/ HemeSelect 100-200 1 0.4 65.8 (54.9-75.6) 94.6 (94.3-94.9) NR NR
Sohn et al,100
2005
Fair 48.9 3794 OC Hemodia 20 1 0.3 25.0 NR 6.0 NR

Abbreviations: CRC, colorectal cancer; FIT, fecal immunochemical test; Hb, hemoglobin; NA, not applicable; NR, not reported.
a Quality assessed using criteria from Quality Assessment of Diagnostic Accuracy Studies (QUADAS)20 and QUADAS 221 instrument.
b Results reported for advanced neoplasia (composite of CRC and advanced adenoma) only.
c Median.

Return to Table of Contents


Note: 1 trial was excluded from the meta-analysis because of a very small number of participants (n = 63).159 There were no episodes of serious bleeding or perforation in the study.

Text Description.

Figure 4 displays a forest plot of perforations from colonoscopy. The pooled event rate from 26 studies in screening populations or generally asymptomatic persons was 3.62 perforations per 10,000 procedures, with a 95% confidence interval of 2.42 to 5.42.

Return to Table of Contents

Note: 1 trial was excluded from the meta-analysis because of a very small number of participants (n = 63).162 There were no episodes of serious bleeding or perforation in the study.

Text Description.

Figure 5 displays a forest plot of major bleeding from colonoscopy. The pooled event rate from 22 studies in screening populations or generally asymptomatic persons was 8.21 major bleeding events per 10,000 procedures, with a 95% confidence interval of 4.95 to 13.61.

Return to Table of Contents


Note: 1 trial was excluded from the meta-analysis because of a very small number of participants (n = 52).162 There were no episodes of serious bleeding or perforation in the study.

Text Description.

Figure 6 displays a forest plot of perforations from flexible sigmoidoscopy. The pooled event rate from 16 studies was 0.74 perforations per 10,000 procedures, with a 95% confidence interval of 0.40 to 1.35.

Return to Table of Contents

Note: 1 trial was excluded from the meta-analysis because of a very small number of participants (n = 52).162 There were no episodes of serious bleeding or perforation in the study.

Text Description.

Figure 7 displays a forest plot of major bleeding from flexible sigmoidoscopy. The pooled event rate from 10 studies is 1.76 major bleeding events per 10,000 procedures, with a 95% confidence interval of 0.70 to 4.41.

Return to Table of Contents

Test Name Study Design No. of Studies No. of
Participants
Summary of Findings (Includes Consistency, Precision) Applicabilitya Limitations (Includes Reporting Bias) Overall Quality
Key Question 1: Effectiveness of Screeningb
SIG RCT 4 458,002 SIG consistently decreased CRC-specific mortality compared with no screening at 11-12 y of follow-up (IRR, 0.73; 95% CI, 0.66-0.82). Mortality benefit was limited to distal CRC. Fair to poor. No longer widely used in the United States. Only 1 trial evaluated more than a single round of screening. Variation in referral criteria led to differing rates of follow-up colonoscopy. Fair to good
gFOBT, Hemoccult II RCT 5 419,966 Biennial screening with Hemoccult II compared with no screening (n=404,396) consistently resulted in reduction of CRC-specific mortality (ranging from 9%-22% after 2-9 rounds of screening with 11-30 y of follow-up). Poor. No longer widely used. Variation in number of screening rounds, use of rehydrated samples, definition of "test positive," and recommended diagnostic follow-up. Fair to good
Key Question 2: Diagnostic Accuracy of Screeningc
Colonoscopy Prospective diagnostic accuracy 4 4821 Comparing colonoscopy with CTC or CTC plus colonoscopy, per-person (or per-lesion) sensitivity for adenomas ≥10 mm was 89%-98%, and per-person sensitivity for adenomas ≥6 mm was 75%-93%. Fair. Colonoscopies were conducted or supervised by "experienced" specialists. Studies were not designed to assess diagnostic accuracy to detect cancers. Limited studies with large number of endoscopists that were applicable to community practice. Fair to good
CTC Prospective diagnostic accuracy 9 6497 The per-person sensitivity and specificity of CTC using bowel preparation to detect adenomas ≥10 mm ranged from 67%-94% and 86%-98%, respectively. The per-person sensitivity and specificity to detect adenomas ≥6 mm ranged from 73%-98% and 80%-93%, respectively. In 2 studies, sensitivity without bowel preparation to detect adenomas was lower than that of CTC protocols using bowel preparation. Fair. Mostly single-center studies, with ≤3 highly trained radiologists. Current practice may use different technologies and protocols. Studies were not designed to assess diagnostic accuracy to detect cancers. Unclear if the variation of test performance was due to differences in study design, populations, bowel preparation, CTC technology, reader experience, or reading protocols. Fair to good
FIT Prospective diagnostic accuracy 6 Qualitative 36,808 In studies with colonoscopy follow-up for all, FIT sensitivity varied considerably across assays for each outcome. OC-Light had the highest sensitivity and specificity for CRC, from 88% and 91%, respectively, to 79% and 93%, respectively. OC FIT-CHEK had the best sensitivity and specificity for CRC, from 73% and 96%, respectively, to 92% and 87%, respectively. Fair to good. There is a wide range in costs for specific tests (OC-Light, OC FIT-CHEK, Cologuard). Quantitative FITs included some that are older and now discontinued. Variation in test performance resulted from the use of 18 different FITs (FIT families), different numbers of stool samples, and to some extent different assay cutoff values. Sparse data on most individual tests limited comparisons. Fair to good
7 Quantitative 40,134
1 FIT plus sDNA 9989 A FIT plus sDNA assay (Cologuard) had better sensitivity but lower specificity, 92% (95% CI, 84-97) and 84% (95% CI, 84-85), respectively, compared with OC FIT-CHEK. FIT plus sDNA was limited to a single study with 6% inadequate stool samples.
Key Questions 3a, 3b: Harms of Screeningd
Endoscopy Prospective and retrospective studies 18 SIG 331,181 Harms from screening SIG were estimated at 1 perforation/10,000 procedures (95% CI, 0.4-1.4/10,000) (No. of studies = 16) and 2 major bleeds/10,000 procedures (95% CI, 0.7-4/10,000) (No. of studies = 10). Good. Reflects community practice. Only 2 studies reported serious adverse events in persons without colonoscopy (no difference in serious harms other than perforation and bleeding). Likely reporting bias of serious harms other than perforation and bleeding. Fair
55 Colonoscopy 10,398,876 Harms from screening colonoscopy or colonoscopy in asymptomatic persons was estimated at 4 perforations/10,000 procedures (95% CI, 2-5/10,000) (No. of studies = 26) and 8 major bleeds/10,000 procedures (95% CI, 5-14/10,000) (No. of studies = 22). Risk of perforations, bleeding, and other serious harms from colonoscopy increased with age.
CTC Prospective and retrospective studies 15 75,354 Harms from CTC in asymptomatic persons were uncommon. Risk of perforation for screening CTC was <2/10,000 examinations. The range of low-dose ionizing radiation per examination was 1-7 mSv. Fair to good. Radiation exposure per examination may be decreasing over time. No studies reported serious adverse events in persons without CTC. Limited evidence in true average-risk screening populations. Likely reporting bias of serious harms other than perforation. No studies reported differential harms by age group. No studies were able to quantify net benefits and harms of ECF findings. Varying levels of follow-up and few studies with final disposition of ECF. Very limited studies comparing ECF by age group. Fair
21 ECF 38,193 ECF was estimated to occur in up to 69% of examinations, and 5%-37% of examinations might necessitate diagnostic follow-up; however, ≤3% required any type of definitive treatment. Higher prevalence of ECF with increasing age.

Abbreviations: CRC, colorectal cancer; CTC, computer tomographic colonography; ECF, extracolonic findings; FIT, fecal immunochemical test; gFOBT, guaiac-based fecal occult blood test; IRR, incidence rate ratio; RCT, randomized clinical trial; sDNA, stool DNA; SIG, flexible sigmoidoscopy.
a Applicability or external validity to US practice.
b Key question 1: What is the effectiveness of screening programs in adults at average risk for colorectal cancer, compared with no screening, in reducing the incidence of or mortality from colorectal cancer?
c Key question 2: In adults at average-risk for colorectal cancer, what are the test performance characteristics (eg, sensitivity and specificity) of a 1-time application of a screening test, compared with an adequate reference standard, for detecting colorectal cancers, advanced adenomas, or adenomas based on size?
d Key question 3a: What are the serious adverse effects of colorectal cancer screening tests in asymptomatic adults? Key question 3b: Do adverse effects vary by important subpopulations (eg, age)?

Return to Table of Contents