Review Open Access
Copyright ©The Author(s) 2019. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Meta-Anal. May 31, 2019; 7(5): 184-208
Published online May 31, 2019. doi: 10.13105/wjma.v7.i5.184
Current state and future direction of screening tool for colorectal cancer
Ji Taek Hong, Department of Internal Medicine, Hallym University College of Medicine, Chuncheon 24253, South Korea
Eun Ran Kim, Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
ORCID number: JiTaek hong (0000-0002-6310-2958); Eun Ran Kim (0000-0002-0495-2565).
Author contributions: All authors equally contributed to this paper with conception and design of the study, literature review and analysis, drafting and critical revision and editing, and final approval of the final version.
Conflict-of-interest statement: No potential conflicts of interest. No financial support.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Corresponding author: Eun Ran Kim, MD, Staff Physician, Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, South Korea. er.kim@samsung.com
Telephone: +82-2-34103409 Fax: +82-2-34106983
Received: March 18, 2019
Peer-review started: March 19, 2019
First decision: May 18, 2019
Revised: May 25, 2019
Accepted: May 27, 2019
Article in press: May 28, 2019
Published online: May 31, 2019
Processing time: 74 Days and 15.1 Hours

Abstract

As the second-most-common cause of cancer death, colorectal cancer (CRC) has been recognized as one of the biggest health concerns in advanced countries. The 5-year survival rate for patients with early-stage CRC is significantly better than that for patients with CRC detected at a late stage. The primary target for CRC screening and prevention is advanced neoplasia, which includes both CRC itself, as well as benign but histologically advanced adenomas that are at increased risk for progression to malignancy. Prevention of CRC through detection of advanced adenomas is important. It is, therefore, necessary to develop more efficient detection methods to enable earlier detection and therefore better prognosis. Although a number of CRC diagnostic methods are currently used for early detection, including stool-based tests, traditional colonoscopy, etc., they have not shown optimal results due to several limitations. Hence, development of more reliable screening methods is required in order to detect the disease at an early stage. New screening tools also need to be able to accurately diagnose CRC and advanced adenoma, help guide treatment, and predict the prognosis along with being relatively simple and non-invasive. As part of such efforts, many proposals for the early detection of colorectal neoplasms have been introduced. For example, metabolomics, referring to the scientific study of the metabolism of living organisms, has been shown to be a possible approach for discovering CRC-related biomarkers. In addition, a growing number of high-performance screening methodologies could facilitate biomarker identification. In the present, evidence-based review, the authors summarize the current state as recognized by the recent guideline recommendation from the American Cancer Society, US Preventive Services Task Force and the United States Multi-Society Task Force and discuss future direction of screening tools for colorectal cancer. Further, we highlight the most interesting publications on new screening tools, like molecular biomarkers and metabolomics, and discuss these in detail.

Key Words: Colorectal cancer, Screening tool, Early detection, Biomarkers, Metabolomics

Core tip: A large proportion of colorectal cancer (CRC) cases and deaths could be prevented by screening with early detection and removal of colorectal adenomas or early stage CRC. Reliable and non-invasive screening tools for early stage CRC and precancerous lesions, such as adenoma is indispensable. However, current screening methods have limitations. Therefore, it is important to review the current literature on new screening tools such as molecular biomarkers and metabolomics for the development of new diagnostic tools.



INTRODUCTION

Colorectal cancer (CRC) remains a global health problem and currently is considered one of the leading causes of death in the world[1]. The patient’s survival is predicted by the tumor stage at the time of diagnosis. Early CRC diagnosis maximizes the benefit of treatment. Typically, it takes 7-10 years for an adenoma to become a carcinoma, which provides a timeframe allowing for early detection of CRC[2]. At present, the best available option for early detection and elimination of premalignant lesions is colonoscopy. However, it is invasive, expensive, and inconvenient for patients. Therefore, non-invasive and reliable methods for diagnosing CRC are valuable due to colonoscopy risks: Puncture of the colon, intraperitoneal bleeding, post-polypectomy, and infection. In particular, with regards to the detection of CRC precursor lesions, such as adenoma, the lack of sensitivity and specificity or an unacceptably wide range of the FOBT has hampered the clinical application in CRC screening[3]. Therefore, newer, non-invasive screening methods and biomarkers to permit identification of CRC and its precursors in easily accessible biospecimens are needed. Consequently, current screening methods have limitations, and it is necessary to find new screening methods that can detect CRC in the early phase to improve survival and quality of life for patients with CRC.

The following qualities are what an ideal screening method would possess: It should show high sensitivity and specificity, it must be safe and cost-effective to be widely used, it must be simple to measure, and readings must be consistent among patients of all genders and races. Acceptability of screening method is also important in target population[4]. In this review, we summarize the current status of screening tools for colorectal cancer and discuss the future direction of colon cancer screening, including metabolism and proteomics.

CURRENT STATE OF SCREENING TOOLS FOR CRC

The American Cancer Society (ACS), US Preventive Services Task Force (USPSTF) and the United States Multi-Society Task Force (USMSTF) Guideline recommends stool-based tests and structural examinations as options for colorectal cancer screening[5-7]. Stool-based tests consist of guaiac-based tests, immunochemical tests, and mt-sDNA tests. Structural examinations consist of colonoscopy, computed tomography colonography, and flexible sigmoidoscopy (FS). These screening tests are currently in use, and we will first discuss the screening value and limitations of currently-used tests. The characteristics, advantages and disadvantages of colorectal cancer screening tests currently in use are shown in Table 1.

Table 1 Characteristics of colorectal cancer screening tests currently in use in the United States.
Screening testIntervalEvidenceAdvantagesDisadvantagesOther considerations
Stool-based screening tests
FIT with high sensitivity123Every yearImproved performance compared with high-sensitivity gFOBT Mortality reduction: indirect evidence from RCTs of guaiac-based stool testsCan be performed at home Requires only a single specimen No diet or medication restrictions Does not require bowel preparation or anesthesia Inexpensive compared with structural examinations and mt-sDNAHigh nonadherence to yearly testing (especially without reminder systems) Less effective for advanced adenoma detection Few accessible tests have published peer-reviewed performance dataVaries in test performance due to brand and version Follow-up colonoscopy for positive test may charge extra costs
gFOBT with high sensitivity12 (HSgFOBT)Every yearGood RCT evidence for incidence and mortality reduction[112-116] Varies in test performance characteristics by version of the testInexpensive compared with structural examinations and mt-sDNA Can be done at home Does not require bowel preparation or anesthesiaHigh nonadherence to yearly testing (especially without reminder system) Less effective for advanced adenoma detection Difficulty in determining test performance among the many FDA-cleared tests Requires multiple samples Requires dietary and medication restriction Higher false-positive rate than FIT leads to more colonoscopiesFollow-up colonoscopy for positive test may charge extra costs
mt-sDNA1Every 3 yrMortality reduction: indirect evidence from RCTs of guaiac-based stool tests Improved sensitivity for cancer and AA and poorer specificity compared with FITCan be done at home Does not require bowel preparation or anesthesiaMore expensive than other stool-based tests Higher false-positive rate than FITFollow-up colonoscopy for positive test may charge extra costs A new test with limited data on screening outcomes. Uncertainty in management of positive results followed by a negative colonoscopy
FIT-DNA23Every 1 or 3 yrTest characteristic studiesImproved sensitivity compared with FIT per single screening test Does not require bowel preparation or anesthesia Can be done at homeHigher false-positive rate than FITUncertainty in management of positive results followed by a negative colonoscopy
Direct visualization screening tests
Colonoscopy123Every 10 yrNon-RCT evidence of incidence and mortality reduction Prospective cohort study with mortality end pointRequires less frequent screening Screening, diagnosis, treatment and prevention through polypectomy can be done at the same-session. Gross visualization of the entire colonPain and discomfort Lower tolerability and compliance than FS[117] Possibility of bowel perforation / bleeding and cardiopulmonary complications from anesthesia Requires full bowel cleansing Performance varies upon adequacy of bowel preparation, the cecal intubation rate, withdrawal time, and adenoma detection rate Lower sensitivity for neoplasia in the proximal than the distal colonPolypectomy and anesthesia may charge extra costs Most expensive test, but currently reimbursable with insurance Requires day-off (if sedation is used)
CTC123Every 5 yrTest characteristic studies Extrapolation from RCTs of sigmoidoscopy demonstrating mortality reductionRapid, non-invasive imaging method Well-tolerated by patients Does not require anesthesia Better tolerability and acceptance than colonoscopy and FS[118]Exposure to low-dose radiation Requires full bowel cleansing A second bowel cleansing will be required before Follow-up colonoscopy for positive testFollow-up colonoscopy for positive test may charge extra costs Insufficient evidence about the benefit-burden balance of additional tests on incidental extracolonic findings Relatively expensive and may not be covered by insurance
FS123Every 5 yrRCTs with mortality end points:Does not require anesthesia Requires more limited bowel cleansing Better acceptance than colonoscopy[117]Pain and discomfort Does not examine the proximal Colon Requires enema prior to procedure Abnormal findings require second colonoscopyFollow-up colonoscopy for positive test may charge extra costs Concerns about lack of quality standards, limited availability, failure to achieve a complete examination
FS with FIT2FS every 10 yr plus FIT every yearRCT with mortality end point (subgroup analysis)More benefits than when combined with FIT or compared with other strategies It may be an potentially option for patients who want endoscopy screening but do not want colonoscopyTest declined in the US
Stool-based CRC screening tests

Stool-based tests are conventionally known as fecal occult blood tests (FOBTs) because they aim to discover the presence of occult blood in stool, which may derive from colorectal cancer or lager polyps of at least 2 cm in size. FOBT are divided into two primary categories according to the detected analyte: Guaiac-based fecal occult blood tests (gFOBT) and fecal immunochemical tests (FITs). gFOBT, which detects the peroxidase activity of hemoglobin, was first recognized as an effective screening for CRC. The use of this stool testing for CRC screening has been supported by multiple consistent randomized clinical trials[8]. However, the use of gFOBT is complicated by its poor sensitivity and specificity as the test shows false negatives when a patient uses antioxidants, like vitamin C, whereas false positives occur when a patient has upper GI bleeding from NSAIDS intake, or consumes red meat or dietary peroxidase from certain vegetables and fruits[9]. On the other hand, FITs specifically detect antibodies against human globin. Thus, it is not influenced by upper GI bleeding since globin is degraded in the upper GI tract, and the test result is well protected from the influence of medications, red meat, or peroxidases from foods, eliminating the need for pre-testing food restrictions[9].

Individual gFOBT and FIT versions show various performance characteristics. Although non-rehydrated, low-sensitivity gFOBT variants are still commercially available, it is not recommended or used for CRC screening test due to the poor performance of these gFOBT. Though there may be other tests having higher sensitivity, at the time of publication, only Hemoccult II Sensa (Beckman Coulter Inc., Brea, CA) performed as a high-sensitivity gFOBT (HSgFOBT) among the many guaiac-based tests evaluated in population-based studies. The ranges of sensitivity and specificity of HSgFOBT are from 62% to 79% and 87% to 96%, respectively[8,10,11]. The fact that gFOBT testing is often carried out in the physician's office in the form of a single-panel test after a digital rectal exam is a potential limitation of this testing. According to Collins et al[12], for advanced neoplasia, the gFOBT testing sensitivity was merely 4.9%, and for cancer, it was just 9%. The accuracy of this method is so low that it cannot, under any circumstances or rationale of convenience, be endorsed as a method for CRC screening. The sensitivity of many individual tests requires optimum situations, and this is another limitation of gFOBT, which can be even more compromised by insufficient and imperfect specimen collection together with absent or inappropriate processing and interpretation. Meanwhile, FITs present higher sensitivity and slightly lower specificity for cancer and advanced neoplasia when compared with low-sensitivity gFOBTs, while demonstrating similar or higher sensitivity and specificity than HSgFOBTs. The ranges of sensitivity and specificity of single-sample FIT are 73% to 92% and 91% to 97%, respectively[9,13-16]. FIT is a non-invasive test. Moreover, in one meta-analysis it showed a one-time sensitivity for cancer of 79% and also had a reasonable sensitivity for advanced adenomas (about 30%), and it is very inexpensive (about $20)[17]. Despite these numbers and advantages, the majority of the brands of FIT do not have sufficient data to show the accuracy of the test in identifying the presence of CRC, as Daly et al[18] were only able to review validation data for less than half of the versions of FIT available in the United States. The FITs were not tested in randomized studies and most of the evidence for the effectiveness of FIT test was based on indirect evidence of reduced mortality due to randomized controlled trials (RCTs) of the guaiac-based stool tests. Studies also used different versions of FITs tests to analyse their outcomes. As the FIT is intended to be repeated, the results for single-sample sensitivity and specificity alone are not sufficient. Although some evidence has started being published recently, the data for the long-term performance of FIT is still lacking[19]; thus, these studies should not be the basis for determining the performance qualities of FITs because they do not yet have adequate data.

Based on the general-population MISCAN modeling analysis that was conducted in 2018, annual FIT from 45–75 year-old adults became a recommendable strategy by providing 94% of the life-year gains (LYGs) compared with that of the standard screening test, a colonoscopy every 10 years for 45–75 year-old adults[20]. Compared to annual FIT, annual HSgFOBT from adults 45 to 75 years of age presented a higher rate of false positives, requiring more colonoscopies; thus, it was not considered to be a model-recommendable strategy, though LYGs of HSgFOBT were same as that of FIT (403 LYGs)[20]. Despite such limitations, HSgFOBT (i.e., Hemoccult II Sensa) is considered to be an option for CRC screening in the updated ACS guidelines because of its high sensitivity and low cost. These benefits can be advantageous when FIT is not available.

No direct injury is caused by HSgFOBT or FIT screening. However, special care must be taken to avoid physical injury when a practitioner performs a colonoscopy to confirm a positive HSgFOBT[8]. Lately, in screening programs for CRC, the original, low-sensitivity guaiac test has been used over HSgFOBT or FIT, with the Unites States similarly changing their screening programs in accordance with this trend.

A third stool test is the multi-targeted stool DNA (mt-sDNA) test that uses an immunochemical assay for human hemoglobin and assays for aberrantly methylated BMP3 and NDRG4, mutated K-ras, and β-Actin from exfoliated cells from colonic neoplasms[21]. Based on a manufacturer-supported, multi-center comparative study of mt-sDNA and FIT testing in average-risk individuals, the sensitivity of mt-sDNA and FIT were 92.3% and 73.8%, respectively[21]. Although the sensitivity of FIT could improve to 77% when the specificity of FIT was as high as that of mt-sDNA (86.6%), the sensitivity of FIT is significantly lower than that of mt-sDNA and did not show sufficient specificity for screening program. Compared with FIT, mt-sDNA was superior in detecting advanced adenomas, especially sessile serrated polyps that were larger than 1 cm. The sensitivity for serrated sessile polyps was 42.4% for mt-sDNA but 5.1% for FIT. However, mt-sDNA had a higher false positive rate, indicating significantly lower specificity (89.8%) compared with that of FIT (96.4%).

In the case of mt-sDNA testing for detecting large adenomas and CRC, the fact that the sensitivity of the test is based on a panel of markers, which seem to identify only a subset of CRC, presents an obvious drawback of mt-sDNA testing. Another possible drawback is the high expense per unit of the currently used test compared to other stool tests. It is also unclear how often the test should be performed. A benefit, though, is the lack of direct harm associated with mt-sDNA, but practitioners should, again, be careful in performing the necessary colonoscopy once a patient’s stool test is positive. Unlike other stool-based tests, mt-sDNA has issues with interpretation of false positive results because the reported results from the mt-sDNA test currently available in the United States cannot distinguish between a positive result originating from FIT versus mt-sDNA testing. A false positive from the mt-sDNA test could result from a failure to detect a visible lesion, invisible neoplastic changes, or from the presence of a non-colonic digestive tract neoplasm. Patients may proceed to more aggressive short-term surveillance to confirm a false positive result from mt-sDNA. Some follow-up studies that observed patients with false positive results from mt-sDNA for approximately 4 years showed that no patient developed CRC or aero-digestive malignancies[22,23]. According to the follow-up study of Cooper et al[24], only three out of 12 patients with previous false positive results on mt-sDNA had positive colonoscopy results upon follow-up. The study also emphasizes the importance of long-term follow-up, and high-quality colonoscopy, especially in the proximal colon, for patients presenting with positive mt-sDNA test results.

In the 2018 MISCAN modeling analysis, mt-sDNA was not a recommended test because it requires higher numbers of colonoscopies per LYGs[20]. Mt-sDNA done every 3 years resulted in 93% of the LYGs compared with annual FIT testing, but when compared with the LYGs of colonoscopy every 10 years, the percentage was 2% less than the a priori criterion of 90%[20].

The ACS’s 2018 guidelines decided to include mt-sDNA as one of the testing options for CRC screening at 3-year intervals, as mt-sDNA is superior in detecting advanced adenomas and serrated sessile polyps, and some adults prefer mt-sDNA over other screening tests. The USMSTF 2017 guidelines recommend that the combined FIT-fecal mt-sDNA test be performed at three-year intervals as a second-tier test (Table 1)[6].

Options for CRC structural (visual) examinations

Structural (visual) examinations, such as endoscopic and radiologic examinations [CT colonography (CTC)] are preferred options for CRC screening when bowel visualization is necessary. Since it directly visualizes the bowel, the screening interval is longer than for stool tests. Structural CRC screening tests require bowel preparation prior to implementing the test, which requires the patient’s active participation for self-administering an enema for FS or ingesting polyethylene glycol oral laxative for CTC. For CTC, patients are also restricted to a liquid diet for one day prior to CTC for bowel cleansing. Unlike FS and CTC, colonoscopy is often performed with anesthesia; hence, the patient must be accompanied by a caretaker[25].

Colonoscopy: Colonoscopy is a widely performed screening for CRC. Patients who are found to be positive for other CRC screening tests undergo colonoscopy for additional assessment. Colonoscopy has a high sensitivity to detect cancer and all classes of precancerous lesions, it allows single-session diagnosis and treatment, and the intervals between examinations are usually long (10 years) in subjects with normal findings. According to a large, prospective, observational cohort study by Nishihara et al[26], the CRC mortality hazard ratio was 0.32 [95% confidence interval (95%CI): 0.24–0.45], when the comparison was made between colonoscopy done at least once and none done at all over 24 years. In addition, distal cancers had a lower hazard ratio of 0.18 (95%CI: 0.10–0.31) compared to proximal cancers, which had a hazard ratio of 0.47 (95%CI: 0.29–0.76). Furthermore, a decrease in incidence was shown in participants who were found to have negative results for colonoscopy (hazard ratio: 0.53, 95%CI: 0.40–0.71)[26]. According to the study by Lin et al[8], the sensitivity and specificity of colonoscopy for identification of adenomas of at least 6 mm in size were 75%–93% and 94%, respectively, whereas those for identification of adenomas at least 1 cm were 89%–98% and 89%, respectively.

According to the three Cancer Intervention and Surveillance Modeling Network models that informed the USPSTF’s 2016 CRC screening guide, CRC incidence and mortality would decrease by 62%–88% and 79%–90%, respectively, if colonoscopy was done every ten years from 50 through 75 years of age[27]. According to the study by Knudsen et al[28], the median LYG for colonoscopy every 10 years was 270, which was higher than those of other exams. Based on the 2018 general-population MISCAN modeling, a large decrease in the incidence of CRC and the number of deaths from CRC along with an increase in LYG compared to other recommendable strategies were observed, although the frequency of lifetime colonoscopy is more than twice that of stool-based testing[20].

However, there are some drawbacks of colonoscopy. Although colonoscopy proved to be more effective as a screening tool than other models, its disadvantages include excessive detection and removal of minute low cancer risk polyps, increasing the risks associated with polypectomy as well as the possibility which could result in unnecessary follow-up evaluations. Thorough bowel cleansing is unavoidable, risk of bowel perforation is higher than for other screening methods, there is a greater risk of pneumonitis due to aspiration (especially when deep sedation is involved in the procedure), a slight risk of splenic injury necessitating splenectomy, and greater occurrence of bleeding after the procedure compared to other screening tests. Major complications of screening through colonoscopy are perforation and bleeding, amounting to approximately four cases per 10,000 screenings and eight cases per 10000 screenings, respectively, according to USPSTF[29]. The frequency of these dangers is increased when polypectomy is performed. There was a significantly greater rate of complications from performing colonoscopy after other positive non-colonoscopy screening tests than when performing an initial colonoscopy[8,30]. Injuries that result from undergoing colonoscopy increase significantly and nonlinearly with the comorbidity burden and age of the patients[31]. Colonoscopy has a greater proba-bility of not being able to detect serrated polyps compared to typical adenomas[32]. Colonoscopy’s performance is also operator-dependent. The skill of the operator affects the detection of cancer, adenomas, and serrated lesions, as well as the selection of appropriate screening and surveillance intervals after colonoscopy[33-42]. Despite these risks, colonoscopy is still the preferred approach to allow gross visualization of the entire colon and same-session detection, biopsy, and removal of polyps.

CTC: CTC, or virtual colonoscopy, produces multiple thin-slice CT images that can be printed on 2D film or compiled into 3D images, enabling examiners to observe internal organs without the use of colonoscopy. CRC detection rates with CTC were similar to that with colonoscopy based on two extensive studies[43,44]. A systemic review and meta-analysis of CTC and colonoscopy based on 49 studies calculated the sensitivity and specificity of CTC. The sensitivity of CTC for detecting CRC was 96.1%, and the sensitivity for detecting adenomas larger than 6 mm was 73%–98% with a specificity of 89%–91%[45]. In CTC, the chances of perforation are less than colonoscopy, and the 82%–92% sensitivity achieved for detecting adenomas larger than 1 cm is also an advantage of CTC[46-49]. Nevertheless, the sensitivity for detecting polyps less than 1 cm is inferior to colonoscopy, and a major deficiency of CTC is the difficulty in detecting flat and serrated lesions[50,51]. Patient who undergo CTC may experience some undesirable symptoms, such as abdominal pain resulting from bowel preparation, pain related to the examination, neuro-cardiogenic syncope and pre-syncope, and very rare worst adverse effects (i.e., GI perforation and radiation exposure-induced cancer). The detection of incidental extracolonic findings is also an unresolved problem. There is the limited evidence about the cost-benefit balance for additional tests necessary for these incidental extracolonic findings[8]. There is insufficient proof that CTC reduces the mortality or incidence of CRC. Both ACS and USPSTF (2016) guideline agreed that CTC every 5 years beginning from 45 to 50 years of age and continuing through 75 years of age is a recommended strategy for CRC screening.

FS: FS, a procedure to evaluate the lower half of the colon, is the first visual exa-mination used for CRC screening. Advantages of FS are that it is very cheap, has significantly lower risks than that of colonoscopy, does not require thorough bowel preparation, and sedation is unnecessary. A disadvantage of FS is that it provides less benefit in protecting against right-sided colon malignancy when compared with the amount of protection achieved by colonoscopy in case-control and cohort studies. In addition, since the procedure does not involve sedation, the patient might experience discomfort, could be dissatisfied by the procedure, and be more hesitant to repeat the examination compared to colonoscopy[52].

By analyzing four RCTs of FS with one or two screening examinations at intervals of every 3-5 years, there was significant decrease in CRC incidence and mortality[53-56]. According to pooled analysis conducted for the USPSTF, patients who had regular follow-up over 11 or 12 years generally decreased their CRC mortality by 27% (relative risk: 0.73, 95%CI: 0.66–0.82), which is especially significant for distal CRC but not proximal CRC[8,27]. There was also a decrease in CRC incidence of 21%. According to the US-based Prostate, Lung, Colon, and Ovarian Cancer (PLCO) Screening Trial, proximal and distal CRC incidence were both significantly reduced with FS screening[27]. In a study by Atkin et al[57], though, there was significant reduction in the incidence of CRC by 26% and mortality by 30%. The study concluded that the result derived from the detection of distal CRC because there was no significant reduction in incidence or mortality of proximal CRC after FS screening. Overall, FS screening showed a 21% reduction in incidence and a 27% reduction in mortality for CRC when analyzing the pooled data from the three studies (PLCO, SCORE, and NORCCAP) that had an average follow-up of 10 to 12 years[58]. Since FS is not effective in screening proximal CRC, which disproportionately affects older women, the incidence and mortality of CRC was not reduced in women aged 60 years or older.

According to MISCAN modeling analysis adjusted for increased incidence, FS screening is recommended beginning at age 45 and repeated every 5 years until the age of 75 years; whereas, assuming stable incidence, USPSTF (2016) is not recommending FS alone for CRC screening[28]. Because the effectiveness of FS screening is mostly restricted to the rectum and distal colon, its use has been replaced by colonoscopy in the United States[20]. A recent report found that only 2.5% of adults who are recommended for CRC screening test underwent FS screening, while 60% of them received colonoscopy[59]. Even if there is solid evidence that FS is an effective CRC screening test, it is questionable whether community-based clinicians are receiving regular training or performing an adequate number of FSs to maintain their skill because it is less frequently used in the United States. This assumption is also supported by proposed FS screening standards that do not provide information about strict quality standards[60]. The reasons mentioned above are leading the ACS Guideline Development Group to remove FS from the list of recommended CRC screening tests. However, it is still considered to be one of the recommended tests for CRC screening in some countries where colonoscopy is not yet commonly performed due to its efficacy in reducing CRC mortality and its availability as a primary visual examination tool.

Emerging technologies not currently recommended for routine screening

Blood screening for methylated SEPT9 DNA (mSEPT9) and capsule endoscopy are not recommended procedures but are FDA approved for certain situations.

Blood screening test for methylated SEPT9 DNA (mSEPT9): The FDA recently cleared a blood test that identifies a CRC biomarker, mSEPT9[61]. This blood screening test is performed on patients with average CRC risk who have declined other screening tests listed in the USPSTF CRC guidelines.

An advantage of the mSEPT9 test is that, as a serum assay, it is more convenient for patients. A disadvantage of the mSEPT9 test is that the performance characteristics are inferior to FIT, that is, sensitivity for cancer is lower than that of FIT, detection of advanced adenoma is impossible, and the cost is more than for other screening tests[62,63]. The test seems to be more sensitive for later stage compared to earlier stage cancer[64]. Patients who receive a positive result from this blood screening test should be ready to have follow-up tests, such as colonoscopy, which they have refused to undergo previously. Whether patients positive for mSEPT9 would be willing to undergo colonoscopy is questionable. There is also limited evidence in asymptomatic populations who are the targeted candidates for screening. Furthermore, no microsimulation modeling for the newer version of the test was done to evaluate the benefit and benefit-to-harm ratio or to determine the optimal timing for screening. Because of these limitations, most guidelines discourage the use of mSEPT9 for screening.

Capsule endoscopy: Initially, capsule endoscopy was predominantly used for gross assessment of the small bowel, but later, there were attempts to use it as a tool to screen the large bowel for CRC. In a systematic review that studied patients with a high risk or who presented with signs or symptoms of CRC, the pooled sensitivity was 87% (95%CI: 77%–93%), and the pooled specificity was 76% (95%CI: 60%–87%) for capsule endoscopy in identification of colorectal polyps at least 6 mm in size[65]. Increased pooled sensitivity and specificity were observed (89%, 95%CI: 77%–95% and 91%, 95% CI: 86%–95%, respectively) in tests on lager colorectal polyps that were at least 10 mm in size[65]. However, the use of capsule endoscopy as a screening tool is limited due to its side effects. Adverse effects were found to include predominantly gastrointestinal problems such as nausea, vomiting, abdominal pain, and fatigue from the required bowel preparation, and these were found in less than 4% of patients[65]. The most severe problem was capsule retention (0.8% of patients with 95%CI: 0.2%–2.4%). Capsule endoscopy also necessitates sufficient colon preparation and further evaluation with colonoscopy if polyps are detected. Capsule endoscopy is not presently approved by the FDA for use in CRC screening.

FUTURE DIRECTION OF SCREENING TOOLS FOR CRC

Currently, finding a CRC-specific tumor marker for the development of a new, non-invasive screening method is a primary focus among researchers. CRC is a disease of a highly heterogeneous nature. To interpret the heterogeneous mechanisms that bring about tumorigenesis, “-omics” data derived from genomic, transcriptomic, epigenetic, and proteomic analysis through multi-omics is required. With a single “-omics” approach, the degree of internal and individual variability related to tumor composition and oncogenic signals may be misinterpreted. Therefore, in order to understand the occurrence of tumor, various approaches are required and being studied. We will review the molecular biomarker studies that have been carried out so far and identify new approaches and studies, including metabolomics for the discovery of new CRC biomarkers.

Molecular biomarkers

CRC is a multifactorial disease caused by genetic and epigenetic changes in oncogenes, mismatch repair genes, tumor suppressor genes, and cell cycle regulating genes of the colon mucosal cells. As these molecular changes provide indications for diagnosis, prognosis, and information on treatment response, they were considered possible CRC biomarkers. The three main molecular pathways contributing to the genetic alterations responsible for carcinogenesis are microsatellite instability (MSI), chromosomal instability (CIN), and the CpG island methylator phenotype. Recently, new methods of molecular detection are being evaluated. Nonetheless, the majority of these methods have not yet been validated in larger, preclinical research using randomized study designs. Studies characteristics, patients characteristics, major markers and diagnostic performance of various molecular biomarkers studies are shown in Table 2.

Table 2 Summary of the current and potential biomarkers for early diagnosis of colorectal cancer.
Characteristics of the studiesTraining set [test set] (if applicable)Diagnostic performance (if applicable)
Ref.Study type, countryStudy groupPopulation (n)Male (%)Age (mean / SD)Stage (0) / I/ II/ III/ IV/ (?)SampleMarkerSn / SpAUC / P-value

Microsatellites loci
Piñol et al[119], 2005Prospective, multicenter, nation-wide study/SpainCRC122259.870/11161/510/337/214BloodBethesda panel81.8/98N/A
Umar et al[71], 2004GuidelinesN/AN/AN/AN/AN/ABloodBethesda panel81.8/98N/A
Berg et al[120], 2009RecommendationsN/AN/AN/AN/AN/ABloodMicrosatellites instability (MSI)55-90/90N/A
Liang et al[67], 2013Meta‐analysis/ChinaN/AN/AN/AN/AN/ABloodAPC PolymorphismsN/AN/A
CRC-specific RNA markers
Wu et al[77], 2014Case-control ChinaNormal10945.960.4/7.0I + II/III + IV/(?) 24/76/4StoolMiRNA-135b78 (CRC) 73(Advanced adenoma) 65(any adenoma) /680.79 (CRC) 0.71 (adenoma) / <0.0001
Adenoma < 1cm11053.6
Advanced adenoma5950.7
CRC10457.7
IBD4261.9
Kalimutho et al[78], 2011Case-control, ItalyCRC284666(5)/2/6/3/0/(NA:12)StoolmiRNA-14874/87N/A
HGD126762
Cn392858
Koga et al[74], 2010Case-control, JapanCRC206676323/46/133/4StoolPTGS274.1/74.1N/A, <0.0001
Cn1344460
Methylation biomarkers
Luo et al[86], 2011Meta‐Analysis/ChinaN/AN/AN/AN/AN/AStoolVIM80/80N/A
Guo et al[88], 2013Case-control, ChinaCRC756158.5 (12.5)12/30/30/3StoolFBNI72/93.3N/A, < 0.001
Cn306758.4 (12.9)
Glockner et al[89], 2009Case-control, United StatesCRC26 [47]52 [45]69.33 [71.1]Stage I to IIIStoolTFP1289/93N/A
Adenoma[19][61.4]
Cn45 [30]46 [54]55 [52.3]
Oh et al[90], 2013Case-control, South KoreaCRC1316958.426/57/36/12BloodSDC287/950.927, < 0.0001
Cn1256451
Grützmann et al[121], 2008Case-control, GermanyCRC252[126]57 [60]61 [67]63/83/59/29/(NA:19)BloodSeptin 948/93N/A
Cn102[183]35 [41]59 [56][22/37/54/11/(NA:3)][58/90]
Warren et al[91], 2011Case-control, United StatesCRC505462I + II/III + IVBlood/StoolSeptin 990/88N/A
Cn94455838/12
Tóth et al[92], 2012Case-control, HungaryCRC935267.8 (9.8)25/14/36/18StoolSeptin9 (gFOBT)100/100N/A
Cn943862.6 (9.9)

Adenomatous polyposis coli mutation: Multifunctional proteins that control Wnt signaling, cell cycle regulation, cytoskeleton stabilization, intracellular adhesion, and apoptosis are encoded by the adenomatous polyposis coli (APC) gene. The APC gene mutation qualifies as a molecular biomarker for CRC diagnosis because appro-ximately 90% of patients with CRC show APC gene mutation[66]. Liang et al[67] have performed meta-analysis study between 1997 and 2010 to correlate APC polymorphisms and CRC risk. It was found that while E1317Q significantly increased the risk of adenoma, I1307K was linked to a high risk of CRC.

MSI: Commonly, MSI is diagnosed by estimating missing MMR gene products, amplification via polymerase chain reaction (PCR), or immunohistochemistry (IHC)[68]. Through meta-analysis and prospective studies, it was shown that MSI serves as an exclusive marker with significant prognostic value in early-stage CRC[69]. The prognosis for MSI CRC was found to be superior to that of microsatellite stable (MSS) CRC[70]. The Bethesda panel consists of five microsatellite loci (BAT25, BAT26, D17S250, D5S346, and D2S123)[71]. At present, most clinical laboratories use a panel of five mononucleotide markers (Bat-25, Bat-26, NR-21, NR-24, and mono-27) to detect MSI[72]. MSI is found to be highly prevalent in stage II CRC with an approximate 20% incidence and rare in stage IV CRC with about a 4% prevalence. Hence, MSI screening may aid in early detection of CRC[72,73].

Detection of CRC-specific RNA Markers in stool: While the fecal occult blood test (FOBts) is commonly used as a screening tool, it still has poor sensitivity and specificity. Many tools using protein, DNA and RNA to detect various markers in stool were recently developed[74]. The idea is such that miRNA not only regulates specific mRNAs and serves a fundamental role in oncogenesis, but also plays critical role in normal development or in tumor cell multiplication, division, and death[75,76]. To diagnose CRC early, several miRNAs were recently assessed. Wu et al[77] obtained 424 stool specimens from adenoma, CRC, and control patients to investigate miRNA. They found out that when compared with the control, expression of miRNA-135b was significantly increased in advanced adenoma and CRC stool specimens. According to a study performed by Kalimutho et al[78], investigating hypermethylated miR-148a in stool specimens may be capable of early CRC detection. Also, following examination of 648 miRNAs from stool specimens of CRC, Kalimutho et al[78] have determined that fecal miR-144 may be used as a tool for CRC diagnosis. With 74% sensitivity and 87% specificity, miR-144 expression was found to be highly significant in CRC stool specimens. Additionally, PTGS2, a transcript of a specific colorectal tumor gene, expression is extremely specific for early diagnosis of CRC[79]. Koga et al[80] acquired stool specimens from 206 patients with CRC and 134 normal individuals and performed a study on miRNA expression in desquamated colonocytes from stool specimens. The result showed a sensitivity of 74.1% and specificity of 74.1%. Although it did not show sufficient specificity to be used as a screening test, they proposed that the profile of miRNA expression may be useful as a CRC screening test from stool specimens.

Methylation biomarkers: A number of factors, including one's lifestyle, diet, aging, reduction of folate levels, exposure to arsenic, and health problems (such as colitis) can lead to colorectal mucosa’s abnormal DNA methylation[81-84]. One can detect patterns of aberrant DNA methylation from CRC cells in the DNA derived from blood or stool specimens from patients with colorectal cancer[85]. Along with the various levels of specificity and sensitivity, several abnormally methylated genes that have been identified in either blood or stool can be used as diagnostic biomarkers in CRC patients. In the United States, for example, vimentin (VIM) gene methylation analysis in a stool-based test is readily available, with about 80% specificity and sensitivity[86]. These abnormally methylated genes are also AIX4, SEPT9, FBNI, WiF-1, P53, PGR, MGMT, TIMP3, and GATA4[81,87]. Guo et al[88] used PCR to study hypermethylation of FBNI in patients with CRC. The study involved tissues and stool specimens from 75 patients with CRC and 30 normal individuals. FBNI hypermethylation was found in 78.7% of CRC tissue specimens and 72% in stool specimens compared to 6.7% of controls, showing a specificity of 93.3% and a sensitivity of 72%. According to Guo et al., estimating hypermethylated FBNI in stool specimen can be a useful non-invasive biomarker for identification of CRC. One of the genes, tissue factor pathway inhibitor 2 (TFPI2), was methylated in almost all patients with CRC of all stages with 97% in adenoma and 99% in CRC[89]. TFPI2 gene methylation in CRC patient’s stool specimens yielded up to a 93% specificity and a 89% sensitivity. Oh et al[90] conducted a study to measure methylation of the SDC2 gene in blood specimens. This study included 131 patients with CRC representing all stages and 125 normal individuals. The results showed a high level of specificity, 95.2%, and an 87.0% level of sensitivity. Also, the sensitivity for early-stage was 92.3%. Therefore, SDC2 methylation in blood was suggested to be a non-invasive, highly sensitive, and specific biomarker for CRC screening[90]. There are a number of CRC screening tests available on the market detecting aberrant gene methylation from either blood or stool. As described previously, the mSEPT9 assay is an example of these available tests. Warren et al[91] conducted a study on the efficacy of the blood-based mSEPT9 assay for CRC detection using blood specimens from 50 CRC patients and 94 healthy individuals. The results showed 90% sensitivity and 88% specificity for all stages. Accordingly, Tóth et al[92] studied the efficiency of detection of mSEPT9, gFOBT, and CEA from CRC and normal plasma. As mSEPT9 achieved high sensitivity and specificity levels of 100%, it is considered to be a superior screening test for CRC detection over CEA and gFOBT.

Despite the wide variety of molecular techniques, More research is needed to produce a new molecular biomarker or biomarker panel that could be used for a broad range of screening. In the future, studies should provide solutions to resolve the predictive and prognostic problems of the proposed and presently used molecular biomarkers. Developing effective molecular screening for CRC capable of detecting early-stage colorectal malignancies would be an innovation. In considering the molecular background of the tumor, molecular markers ensure that the field develops a more personalized approach. Identifying clinically-related, cost-effective and easily tested biomarkers to facilitate patient management decisions and provide direct benefits to the patient is, after all, the goal.

Metabolomics

One option for non-invasive screening is metabolomics, which is a potential tumor marker for CRC. It is important to have a comprehensive understanding of all small-molecule marker metabolites of CRC to accurately understand the tumor metabolic pathway that will assist diagnosis and become the basis for novel preventive and therapeutic methods.

Published studies that attracted large amounts of publicity have recently peaked interest in the possibilities of metabolomic analysis to identify biomarkers for advanced identification of disease progression from easily obtainable biofluids. Therefore, metabolomics analysis had only just started to join the conventional practices of cancer diagnosis and treatment.

One of newly rising “omics” studies, metabolomics investigates global, or system-wide, metabolic profiles, offering a dynamic portrait of the metabolic status of living systems. Being highly potent for diagnosing various cancers using advanced analytic techniques and biometric tools, this approach has been used for therapeutic monitoring and drug development. There are some metabolic markers always found in CRC; however, metabolic profiles of patients with early-stage CRC, including precancerous lesions, are not clearly understood. Due to the non-invasive nature of the approach, it warrants further investigation.

Characteristics of Colorectal Cancer Screening By Biofluid Sample Type (Blood, Urine, Stool): Novel diagnostics can be subdivided based on the type of biofluid sample to be analyzed, primarily blood, urine, or stool specimens. The pros and cons of each specimen are shown in Table 3.

Table 3 Characteristics of colorectal cancer screening of bio fluidic sample types (blood, urine, stool).
Sample typesEvidence of efficacyAdvantageDisadvantage
Blood-based biomarkers (serum, plasma, and dried blood spot)A combination of 8 metabolites (99.3% sensitivity, 93.8% specificity, and AUC 0.996)[94] Gastrointestinal tract acid 446 (83.3% sensitivity, 84.8% specificity, 85.7%, and 52.1% , respectively)[96,97] Decanoic acid (87.87% sensitivity, 80.0% specificity, 71.0%, and 75.0%, respectively)[98,99]Easily accessible Less affected by diet than urine Less diurnal variation and Less inter- and intra-subject variability than urine Stable over a 4-mo period frozen at -80 °C except at room temperatureAffected by smoking status More invasive than urine and stool Analysis can be more complex than urine
UrineCross-validated panel of seven metabolites (97.5% sensitivity, 100% specificity, and AUC 0.998)[104] 10 different metabolites (100% sensitivity, 80% specificity but small sample size)[103] N1, N12-Diacetylspermine[105,106]Easily accessible Less invasive than bloodMore affected by diet than serum samples More diurnal variation and More inter- and intra-subject variability than serum A full day storing at room temperature or on cool packs altered metabolite concentration More than 2 freeze and thaw cycles affected the metabolic profile significantly
StoolA three metabolite panel (AUC 1.0 but very small sample size)[107] A metabolomics panel (AUC 0.94)[108]Easily accessible Less invasive than bloodInconvenient to collect of stool samples Low compliance

(1) Blood-based biomarkers: Blood-based markers can be found in either plasma or serum samples, as well as in dried blood spots, which only requires minimal amounts of blood. Moreover, blood-based markers from dried blood spots have particular advantages, such as easy transportation, convenient storage, and ability to delay processing[93].

In a study of blood-based biomarkers, a dried blood spot biomarker that was composed of four amino acids and four acylcarnitines resulted a quite reasonable sensitivity (81.2%) and specificity (84.0%)[93]. One issue of this study, however, was that the 62% of participants were already in a later stage (III or IV) of CRC. Among the available blood-based panels, the most effective biomarker was introduced by Nishiumi et al[94], who combined eight metabolites to detect early-stage CRC. The panel showed 99.3% sensitivity, 93.8% specificity, and an area under the curve (AUC) of 0.996. The highest sensitivity and specificity were reported for a single marker, but the study involved limitations, such as a small study population and relatively young age (18–22 years) of healthy controls[95]. Most of all, the study was not validated. Gastrointestinal tract acid 446 (GTA-446) is a rising biomarker that has been newly introduced by Hata et al[96] (83.3% sensitivity, 84.8% specificity) and Ritchie et al[97] (85.7% sensitivity, 52.1% specificity). In addition, two independent studies found that decanoic acid could be a promising biomarker candidate (87.87% and 71.0% sensitivity, 80.0% and 75.0% specificity)[98,99].

(2) Urine: Most studies of biomarkers found in urine have discovered that a panel is more suitable than solitary metabolites. The outcomes of three Canadian studies were based on identical study settings[100-102]. Among the studies, the assay with the highest sensitivity used ten distinct metabolites. However, no additional categorization was done for the latter[103]. The study showed 100% sensitivity and a specificity of 80%. However, it had a small sample size. A cross-validated panel that included seven metabolites had a sensitivity of 97.5% (AUC: 0.998) and a specificity of 100%, the highest percentage[104]. Two studies, one by Deng, Deng et al[101] and another by H. Wang et al[102] reported similarly high sensitivities. In addition, two separate studies detected N1, N12-diacetylspermine as a distinct biomarker that could be used for a future screening test[105,106].

(3) Stool: In a systematic review of studies on early identification of abnormal colorectal growths using biomarker detection, one study reported an AUC of 1.0 based on a three-metabolite panel[107]. However, the research only had a small population size. Participants from true screening study showed another metabolomics panel to identify advanced colorectal neoplasms. The panel demonstrated good performance (AUC: 0.94)[108].

Sample type, analytical techniques, major metabolites, outcomes, sensitivity, specificity and significant findings of various metabolomic studies are shown in Table 4. It seems that a panel of metabolites is superior to a single marker for advanced colorectal neoplasms. As for amino acids in blood specimens and nucleosides in urine samples, the findings were consistent.

Table 4 High-throughput metabolomic studies of potential biomarkers in CRC screening.
Sample typeRef.Analytical technique(s)Major metabolitesOut-comesSn / SpSignificant finding(s)
Dried bloodJing et al[93], 2017Direct infusion MSAA (4) FA (4)CRC81.2/84Establishing a reasonable diagnostic regression model with eight blood parameters
SERUMBPZhang et al[122], 2018UPLC-MS/MSFA(2): EicosanoidsCRCN/AIdentification of eicosanoids as potential biomarkers for identifying among health, enteritis and CRC
Guo et al[123], 2017FTICR MSFA(5): Male FA(2): FemaleCRC77.3/92.4 80.8/85.9Presenting the relationship between the change trends of six phospholipids and cancer stages
Farshidfar et al[124], 2016GC-MSAA (9) FA(7) CH (12) Others (13)CRC85.0/86.0Discovery of a suite of CRC biomarkers that provide early detection, prognostication and preliminary staging information
Zhang et al[125], 2016FTICR MSFA (6)CRC93.8/92.2Identification of Free Fatty Acids as diagnostic indicators of early-stage CRC patients
Gu et al[126], 2015LC-MS/MSAA (8)CRC65.0/95.0Performing a combined analysis of amino acids in three different domains: FAAs, FSPAAs, and IPAAs
Zhu et al[127], 2014LC-MSAA (7) FA (3) CH (3)CRC96.0/80.0Establishing Partial least-squares-discriminant analysis (PLS-DA) models for distinguishing CRC patients
Li et al[128], 2013DI-ESI (±) -FTICR MSFA (9)CRC86.5/96.2Emphasize that the facile loss of methyl chloride from the [M  +  Cl] (-) form of LPC (16:0) in its tandem mass spectrum
Tan et al[129], 2013UPLC-QTOFMSAA (6) FA (1) CH (3)CRC83.7/91.7Identification of serum metabolite markers as diagnostic indicators for the detection of CRC
Ma et al[130], 2012GC-MSAA (3) CH (3)CRC93.31/96.71Emphasize integrated network connectivity analysis for the diagnosis
Nishiumi et al[131], 2012GC-MSAA (3) CH (1)CRC83.1/81.0Establishing potential predictive model for early detection of colorectal cancer
Ritchie et al[132], 2010FTICR MSFA (3)CRC75.0/90.0IdentifIcation of a systemic metabolic dysregulation comprising previously unknown hydroxylated polyunsaturated ultra-long chain fatty acid metabolites in CRC patients
Ludwig et al[133], 2009Hadamard-encoded TOCSY spectraFA (1) CH (4)CRC70.0/95.0Showing the potential of fast Hadamard-encoded TOCSY spectra for improved classification of serum samples from colorectal cancer patients using a metabolomics approach
SHata et al[96], 2017FIA–MS/MSFA (1: GTA-446)CRC83.3/84.8Identification of GTA-446 as promising tool for primary colorectal cancer screening
Uchiyama et al[98], 2017CE-TOFMSFA (1): Benzoic FA (1): Octanoic FA (1): Decanoic AA (1): HistidineCRC89.0/82.0 76.0/71.0 71.0/75.0 63.0/82.0The first report to determine the correlation between serum metabolites and CRC stage using CE-TOFMS Identification of benzoic acid as diagnostic indicators
Ritchie et al[97], 2013TQ-MSFA (1)CRC85.7/~52.12Identification of low-serum GTA-446 as significant risk factor for CRC and sensitive predictor of early-stage disease
Ikeda et al[134], 2012GC-MSAA (1): Alanine CH (1): GluL AA(1): GlutamineCRC54.5/91.6 75.0/75.0 81.8/66.7Showing the potential of metabolomics as an early diagnostic tool for cancer
Leichtle et al[135], 2012TIS-MSAA (1)CRCN/AShowing serum glycine and tyrosine in combination with CEA are superior to CEA for the discrimination
PLASMABPNishiumi et al[94], 2017GC/QqQMSAA (3) FA (3) CH (2)Stage 0/I/II99.3/93.8Establishing potential predictive model of colorectal cancer that do not involve lymph node or distant metastasis
Li et al[136], 2013Lipid extraction MSFA (3)CRC88.3/80.0Identification of the plasma choline-containing phospholipid levels as potential biomarkers to distinguish between healthy controls, AP and CRC cases, implying their clinical usage in CRC and/or AP-CRC progression detection
Miyagi et al[137], 2011HLPC-ESI-MSAA (10)CRCN/AShowing the potential of plasma free amino acids profiling for improving cancer screening and diagnosis and understanding disease pathogenesis
Okamoto et al[138], 2009HLPC-ESI-MSAA (6)CRCN/APresenting the possibility of plasma free amino acids profiling
Zhao et al[139], 2007LC- MSFA (4)CRC82.0/93.0Identification of percentage of 18:1-LPC or 18:2-LPC plasma levels compared with total saturated LPC levels, either individually or in combination as potential biomarkers for CRC
SLiu et al[140], 2018N/AAA(1) :HomocysteineCRC/A43.5/98.8Presenting the possibility of using homocysteine with CEA in screening of early rectal cancer
Shen et al[95], 20172D LC-QToF/MSFA (1): PG FA (1): SMCRC1.00/1.00 1.00/1.00Presenting the possibility of 2D LC-QToF/MS-based lipidomics profiling
Crotti et al[99], 2016GC-MSFA (1)CRC87.8/80.0Identification of the C10 fatty acid as valuable early diagnostic biomarker of CRC
Cavia-Saiz et al[141], 2014high pressure-LCAA (1)CRC85.2/100Identification of the plasma levels of l-kynurenine as a potential biomarkers of CRC
URINEBPNakajima et al[105], 2018LC- MSAA (2)CRCN/APresenting the potential of polyamines and a machine-learning method as a screening tool of CRC
Deng,Fang et al[142], 20171-dimensional NMRAA (7) FA (2) CH (8)A82.6/42.4Presenting novel urine-based metabolomic diagnostic test for the detection of adenomatous polyps
Deng et al[101], 2017LC- MSFA (1) CH (2)A82.43/36.03Presenting a clinically scalable MS-based urine metabolomic test for the detection of adenomatous polyps
Wang et al[143],2017H-NMRAA (3) CH (1)Stage I/II87.5/91.3Supporting the utility of NMR-based urinary metabolomics fingerprinting in early diagnosis of CRC
Rozalski et al[144], 2015GC-MSCH (3)CRC78.6/75.0Identification of Urinary 5-hydroxymethyluracil and 8-oxo-7,8-dihydroguanine as potential biomarkers
Wang et al[102], 20141-dimensional NMRAA (7) FA (2) CH (8)A82.7/51.2Presenting a proof-of-concept spot urine-based metabolomic diagnostic test
Hsu et al[145], 2013HPLC-MS/MSCH (6)CRC69.0/98.0Identification of a set of six targeted nucleosides as marker
Eisner et al[100], 2013H-NMRAA (2) CH (2)Polyps64.0/65.0Presenting a machine-learned predictor of colonic polyps based on urinary metabolomics
Yue et al[103], 2013RRLC-QTOF/MSFA (9) Others (1)CRC100/80.0Identification of CRC urinary metabolites as marker
Cheng et al[104], 2012GC/TOF-MS UPLC-QTOFMSAA (4) FA (1) CH (2)CRC97.5/100Reporting a second urinary metabonomic study on a larger cohort of CRC (n = 101) and healthy subjects (n = 103)
Chen[146], 2012CE-MSAA (8) CH (4)CRCN/APresenting the usefulness of the technique of CE-MS based on moving reaction boundary
Wang et al[147], 2010UPLC-MS SPE-HPLCAA(4) FA(5) / CH (7)CRCN/AIdentification of urinary metabolic biomarker based on UPLC-MS and SPE-HPLC
Feng[148], 2005RP-HPLCCH (2)CRC71.2/93.3Identification of Pseu and m1G as novel biomarkers for colorectal cancer diagnosis and surgery monitoring
Zheng et al[149], 2005Column switching HPLCCH (14)CRC71.0/96.0Identification of urinary nucleosides determined by column switching high performance liquid chromatography method
SJohnson et al[150], 2006LC- MSFA (1)ACN90.0/45.0Identification of urinary PGE-M as a potential biomarker of ACN
Hiramatsu et al[106], 2005ELISAAA (1)CRC75.8/96.0Indicating that urinary N(1), N(12)-Diacetylspermine is a more sensitive marker than CEA, CA19-9, and CA15-3
FECESBPAmiot et al[108], 2015H-NMRAA (2) FA (4) CH (1)ACNN/AIdentification of (1)H NMR Spectroscopy of Fecal Extracts as biomarker
Phua et al[107], 2014GC/TOF-MSFA (1) CH (2)CRCN/AEstablishing proof-of-principle for GC/TOFMS-based fecal metabonomic detection of CRC
Bezabeh et al[151], 2009(1)H-MRSAA (6) FA (1) CH (3)CRC85.2/86.9Detecting colorectal cancer by 1H magnetic resonance spectroscopy of fecal extracts
SLin et al[152], 2016H-NMRFA (1): Acetate FA (1): SuccinateEarly stage94.7/92.3 91.2/93.5Identification of the potential utility of NMR-based fecal metabolomics fingerprinting as predictors
Limitations of current studies on metabolic biomarkers and influences on metabolomics profiles

Due to some drawbacks, interpreting and implementing metabolomics studies becomes complicated, in particular, poor standardization is a major concern. The sample to be analyzed also has advantages and disadvantages depending on the type, and the results can be influenced by various situations (Table 3). For future, practical use, the Standard Metabolomics Reporting Structure Group attempted to standardize protocols for metabolomics studies beginning with the design of the study, collection and preparation of specimens[109]. Poor standardization could reduce the compa-rability of studies.

Another limitation is that there is insufficient individual validation of the biomarkers in controlled clinical settings or in a true screening setting for early detection of malignancy in a cohort of asymptomatic individuals[110]. The majority of studies report biomarker panels used in their studies that have not been validated. Insufficient validation could lead to overestimation of the performance of biomarker panels because of overfitting. There are concerns of generalization in the case of studies that only used internal validation. Also, the ability to detect valid biomarkers is limited because most of the studies were performed with comparatively small sample sizes[111]. In clinical practice, before using metabolomics for early detection, significant effort should be devoted to screening large cohorts under standardized circumstances. Also, since the majority of subjects in these studies were Asian, there may be limited generalization and transferability to other races.

CONCLUSION

Herein, we provide a review of the literature on the current state and future direction of screening tools for colorectal cancer. Generally, detecting cancer and its precursors at an early stage and initiating treating can prevent unnecessary deaths from colorectal cancer. However, because of the limitations of the screening tools currently in use, the development of new screening tools is required, and studies on metabolomics and proteomics are currently underway. It may be possible to develop a new non-invasive diagnostic test based on biomarkers, which is simple, cost-effective, and highly specific and sensitive. Yet, due to heterogeneity of the biomarkers, more research on this topic needs to be conducted before implementing these potential screening biomarkers in clinical settings. Especially important for achieving better efficacy in colorectal cancer screening are establishing standardized protocols in research for metabolomics and proteomics, carrying out larger studies in true screening settings, and external validation of the outcomes. For better diagnostic performance of non-invasive tests in detecting CRC or its precursors, combining various approaches, such as metabolomics and proteomics, should also be considered.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Medicine, research and experimental

Country of origin: South Korea

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): D

Grade E (Poor): 0

P-Reviewer: Jeong KY, Zorzi M S-Editor: Gong ZM L-Editor: A E-Editor: Wu YXJ

References
1.  Diaz LA, Williams RT, Wu J, Kinde I, Hecht JR, Berlin J, Allen B, Bozic I, Reiter JG, Nowak MA, Kinzler KW, Oliner KS, Vogelstein B. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature. 2012;486:537-540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1259]  [Cited by in F6Publishing: 1302]  [Article Influence: 108.5]  [Reference Citation Analysis (0)]
2.  Bardhan K, Liu K. Epigenetics and colorectal cancer pathogenesis. Cancers (Basel). 2013;5:676-713.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 170]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
3.  Levin B, Lieberman DA, McFarland B, Andrews KS, Brooks D, Bond J, Dash C, Giardiello FM, Glick S, Johnson D, Johnson CD, Levin TR, Pickhardt PJ, Rex DK, Smith RA, Thorson A, Winawer SJ; 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. Gastroenterology. 2008;134:1570-1595.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1423]  [Cited by in F6Publishing: 1424]  [Article Influence: 89.0]  [Reference Citation Analysis (0)]
4.  Markowitz SD, Bertagnolli MM. Molecular origins of cancer: Molecular basis of colorectal cancer. N Engl J Med. 2009;361:2449-2460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1274]  [Cited by in F6Publishing: 1320]  [Article Influence: 88.0]  [Reference Citation Analysis (2)]
5.  Wolf AMD, Fontham ETH, Church TR, Flowers CR, Guerra CE, LaMonte SJ, Etzioni R, McKenna MT, Oeffinger KC, Shih YT, Walter LC, Andrews KS, Brawley OW, Brooks D, Fedewa SA, Manassaram-Baptiste D, Siegel RL, Wender RC, Smith RA. Colorectal cancer screening for average-risk adults: 2018 guideline update from the American Cancer Society. CA Cancer J Clin. 2018;68:250-281.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 945]  [Cited by in F6Publishing: 1123]  [Article Influence: 187.2]  [Reference Citation Analysis (0)]
6.  U.S. Preventive Services Task Force. Screening for colorectal cancer: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med. 2008;149:627-637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1005]  [Cited by in F6Publishing: 1053]  [Article Influence: 65.8]  [Reference Citation Analysis (1)]
7.  Rex DK, Boland CR, Dominitz JA, Giardiello FM, Johnson DA, Kaltenbach T, Levin TR, Lieberman D, Robertson DJ. Colorectal Cancer Screening: Recommendations for Physicians and Patients from the U.S. Multi-Society Task Force on Colorectal Cancer. Am J Gastroenterol. 2017;112:1016-1030.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 368]  [Cited by in F6Publishing: 420]  [Article Influence: 60.0]  [Reference Citation Analysis (0)]
8.  Lin JS, Piper MA, Perdue LA, Rutter CM, Webber EM, O'Connor E, Smith N, Whitlock EP. Screening for Colorectal Cancer: Updated Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA. 2016;315:2576-2594.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 506]  [Cited by in F6Publishing: 519]  [Article Influence: 64.9]  [Reference Citation Analysis (0)]
9.  Young GP, Symonds EL, Allison JE, Cole SR, Fraser CG, Halloran SP, Kuipers EJ, Seaman HE. Advances in Fecal Occult Blood Tests: the FIT revolution. Dig Dis Sci. 2015;60:609-622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 121]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
10.  Levi Z, Birkenfeld S, Vilkin A, Bar-Chana M, Lifshitz I, Chared M, Maoz E, Niv Y. 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;128:2415-2424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 76]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
11.  Allison JE, Tekawa IS, Ransom LJ, Adrain AL. A comparison of fecal occult-blood tests for colorectal-cancer screening. N Engl J Med. 1996;334:155-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 430]  [Cited by in F6Publishing: 387]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
12.  Collins JF, Lieberman DA, Durbin TE, Weiss DG; Veterans Affairs Cooperative Study #380 Group. Accuracy of screening for fecal occult blood on a single stool sample obtained by digital rectal examination: a comparison with recommended sampling practice. Ann Intern Med. 2005;142:81-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 148]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
13.  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;49:3049-3054.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in F6Publishing: 163]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
14.  Ou CH, Kuo FC, Hsu WH, Lu CY, Yu FJ, Kuo CH, Wang JY, Wu MT, Shiea J, Wu DC, Hu HM. Comparison of the performance of guaiac-based and two immunochemical fecal occult blood tests for identifying advanced colorectal neoplasia in Taiwan. J Dig Dis. 2013;14:474-483.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 5]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
15.  Rabeneck L, Rumble RB, Thompson F, Mills M, Oleschuk C, Whibley A, Messersmith H, Lewis N. Fecal immunochemical tests compared with guaiac fecal occult blood tests for population-based colorectal cancer screening. Can J Gastroenterol. 2012;26:131-147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 73]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
16.  Robertson DJ, Lee JK, Boland CR, Dominitz JA, Giardiello FM, Johnson DA, Kaltenbach T, Lieberman D, Levin TR, Rex DK. Recommendations on Fecal Immunochemical Testing to Screen for Colorectal Neoplasia: A Consensus Statement by the US Multi-Society Task Force on Colorectal Cancer. Gastroenterology. 2017;152:1217-1237.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in F6Publishing: 245]  [Article Influence: 35.0]  [Reference Citation Analysis (0)]
17.  Lee JK, Liles EG, Bent S, Levin TR, Corley DA. Accuracy of fecal immunochemical tests for colorectal cancer: systematic review and meta-analysis. Ann Intern Med. 2014;160:171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 389]  [Cited by in F6Publishing: 435]  [Article Influence: 43.5]  [Reference Citation Analysis (0)]
18.  Daly JM, Xu Y, Levy BT. Which Fecal Immunochemical Test Should I Choose? J Prim Care Community Health. 2017;8:264-277.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
19.  Zorzi M, Hassan C, Capodaglio G, Narne E, Turrin A, Baracco M, Dal Cin A, Fiore A, Martin G, Repici A, Rex D, Rugge M. Divergent Long-Term Detection Rates of Proximal and Distal Advanced Neoplasia in Fecal Immunochemical Test Screening Programs: A Retrospective Cohort Study. Ann Intern Med. 2018;169:602-609.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 32]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
20.  Peterse EFP, Meester RGS, Siegel RL, Chen JC, Dwyer A, Ahnen DJ, Smith RA, Zauber AG, Lansdorp-Vogelaar I. The impact of the rising colorectal cancer incidence in young adults on the optimal age to start screening: Microsimulation analysis I to inform the American Cancer Society colorectal cancer screening guideline. Cancer. 2018;124:2964-2973.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 144]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
21.  Imperiale TF, Ransohoff DF, Itzkowitz SH, Levin TR, Lavin P, Lidgard GP, Ahlquist DA, Berger BM. Multitarget stool DNA testing for colorectal-cancer screening. N Engl J Med. 2014;370:1287-1297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1015]  [Cited by in F6Publishing: 1067]  [Article Influence: 106.7]  [Reference Citation Analysis (1)]
22.  Cotter TG, Burger KN, Devens ME, Simonson JA, Lowrie KL, Heigh RI, Mahoney DW, Johnson DH, Ahlquist DA, Kisiel JB. Long-term Follow-up of Patients Having False-Positive Multitarget Stool DNA Tests after Negative Screening Colonoscopy: The LONG-HAUL Cohort Study. Cancer Epidemiol Biomarkers Prev. 2017;26:614-621.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
23.  Geenen DJ, Imperiale TF, Caskey SL, Anderson KA, Berger B. A 3-year observational study of persons with a negative colonoscopy and positive multi-target stool DNA test. Gastroenterology. 2017;152:S838.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Cooper GS, Markowitz SD, Chen Z, Tuck M, Willis JE, Berger BM, Brenner DE, Li L. Evaluation of Patients with an Apparent False Positive Stool DNA Test: The Role of Repeat Stool DNA Testing. Dig Dis Sci. 2018;63:1449-1453.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 11]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
25.  Wernli KJ, Brenner AT, Rutter CM, Inadomi JM. Risks Associated With Anesthesia Services During Colonoscopy. Gastroenterology. 2016;150:888-94; quiz e18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 140]  [Article Influence: 17.5]  [Reference Citation Analysis (1)]
26.  Nishihara R, Wu K, Lochhead P, Morikawa T, Liao X, Qian ZR, Inamura K, Kim SA, Kuchiba A, Yamauchi M, Imamura Y, Willett WC, Rosner BA, Fuchs CS, Giovannucci E, Ogino S, Chan AT. Long-term colorectal-cancer incidence and mortality after lower endoscopy. N Engl J Med. 2013;369:1095-1105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 968]  [Cited by in F6Publishing: 1073]  [Article Influence: 97.5]  [Reference Citation Analysis (0)]
27.  US Preventive Services Task Force; Bibbins-Domingo K, Grossman DC, Rutter CM, Curry SJ, Davidson KW, Epling JW Jr, García FAR, Gillman MW, Harper DM, Kemper AR, Krist AH, Kurth AE, Landefeld CS, Mangione CM, Owens DK, Phillips WR, Phipps MG, Pignone MP, Siu AL. Screening for Colorectal Cancer: US Preventive Services Task Force Recommendation Statement. JAMA. 2016;315:2564-2575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1249]  [Cited by in F6Publishing: 1310]  [Article Influence: 163.8]  [Reference Citation Analysis (1)]
28.  Knudsen AB, Zauber AG, Rutter CM, Naber SK, Doria-Rose VP, Pabiniak C, Johanson C, Fischer SE, Lansdorp-Vogelaar I, Kuntz KM. Estimation of Benefits, Burden, and Harms of Colorectal Cancer Screening Strategies: Modeling Study for the US Preventive Services Task Force. JAMA. 2016;315:2595-2609.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 366]  [Cited by in F6Publishing: 331]  [Article Influence: 41.4]  [Reference Citation Analysis (0)]
29.  Reumkens A, Rondagh EJ, Bakker CM, Winkens B, Masclee AA, Sanduleanu S. Post-Colonoscopy Complications: A Systematic Review, Time Trends, and Meta-Analysis of Population-Based Studies. Am J Gastroenterol. 2016;111:1092-1101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 206]  [Article Influence: 25.8]  [Reference Citation Analysis (0)]
30.  Levin TR, Zhao W, Conell C, Seeff LC, Manninen DL, Shapiro JA, Schulman J. Complications of colonoscopy in an integrated health care delivery system. Ann Intern Med. 2006;145:880-886.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 382]  [Cited by in F6Publishing: 382]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
31.  Warren JL, Klabunde CN, Mariotto AB, Meekins A, Topor M, Brown ML, Ransohoff DF. Adverse events after outpatient colonoscopy in the Medicare population. Ann Intern Med. 2009;150:849-857, W152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 303]  [Cited by in F6Publishing: 303]  [Article Influence: 20.2]  [Reference Citation Analysis (0)]
32.  de Wijkerslooth TR, Stoop EM, Bossuyt PM, Tytgat KM, Dees J, Mathus-Vliegen EM, Kuipers EJ, Fockens P, van Leerdam ME, Dekker E. Differences in proximal serrated polyp detection among endoscopists are associated with variability in withdrawal time. Gastrointest Endosc. 2013;77:617-623.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 110]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
33.  Kaminski MF, Regula J, Kraszewska E, Polkowski M, Wojciechowska U, Didkowska J, Zwierko M, Rupinski M, Nowacki MP, Butruk E. Quality indicators for colonoscopy and the risk of interval cancer. N Engl J Med. 2010;362:1795-1803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1287]  [Cited by in F6Publishing: 1366]  [Article Influence: 97.6]  [Reference Citation Analysis (0)]
34.  Corley DA, Jensen CD, Marks AR, Zhao WK, Lee JK, Doubeni CA, Zauber AG, de Boer J, Fireman BH, Schottinger JE, Quinn VP, Ghai NR, Levin TR, Quesenberry CP. Adenoma detection rate and risk of colorectal cancer and death. N Engl J Med. 2014;370:1298-1306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1251]  [Cited by in F6Publishing: 1397]  [Article Influence: 139.7]  [Reference Citation Analysis (0)]
35.  Shaukat A, Rector TS, Church TR, Lederle FA, Kim AS, Rank JM, Allen JI. Longer Withdrawal Time Is Associated With a Reduced Incidence of Interval Cancer After Screening Colonoscopy. Gastroenterology. 2015;149:952-957.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 160]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
36.  Barclay RL, Vicari JJ, Doughty AS, Johanson JF, Greenlaw RL. Colonoscopic withdrawal times and adenoma detection during screening colonoscopy. N Engl J Med. 2006;355:2533-2541.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 911]  [Cited by in F6Publishing: 907]  [Article Influence: 50.4]  [Reference Citation Analysis (0)]
37.  Chen SC, Rex DK. Endoscopist can be more powerful than age and male gender in predicting adenoma detection at colonoscopy. Am J Gastroenterol. 2007;102:856-861.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 284]  [Cited by in F6Publishing: 290]  [Article Influence: 17.1]  [Reference Citation Analysis (0)]
38.  Butterly L, Robinson CM, Anderson JC, Weiss JE, Goodrich M, Onega TL, Amos CI, Beach ML. Serrated and adenomatous polyp detection increases with longer withdrawal time: results from the New Hampshire Colonoscopy Registry. Am J Gastroenterol. 2014;109:417-426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 141]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
39.  Mysliwiec PA, Brown ML, Klabunde CN, Ransohoff DF. Are physicians doing too much colonoscopy? A national survey of colorectal surveillance after polypectomy. Ann Intern Med. 2004;141:264-271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 226]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
40.  Pohl H, Srivastava A, Bensen SP, Anderson P, Rothstein RI, Gordon SR, Levy LC, Toor A, Mackenzie TA, Rosch T, Robertson DJ. Incomplete polyp resection during colonoscopy-results of the complete adenoma resection (CARE) study. Gastroenterology. 2013;144:74-80.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 476]  [Cited by in F6Publishing: 510]  [Article Influence: 46.4]  [Reference Citation Analysis (0)]
41.  Abdeljawad K, Vemulapalli KC, Kahi CJ, Cummings OW, Snover DC, Rex DK. Sessile serrated polyp prevalence determined by a colonoscopist with a high lesion detection rate and an experienced pathologist. Gastrointest Endosc. 2015;81:517-524.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 132]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
42.  IJspeert JE, de Wit K, van der Vlugt M, Bastiaansen BA, Fockens P, Dekker E. Prevalence, distribution and risk of sessile serrated adenomas/polyps at a center with a high adenoma detection rate and experienced pathologists. Endoscopy. 2016;48:740-746.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 66]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
43.  Johnson CD, Chen MH, Toledano AY, Heiken JP, Dachman A, Kuo MD, Menias CO, Siewert B, Cheema JI, Obregon RG, Fidler JL, Zimmerman P, Horton KM, Coakley K, Iyer RB, Hara AK, Halvorsen RA, Casola G, Yee J, Herman BA, Burgart LJ, Limburg PJ. Accuracy of CT colonography for detection of large adenomas and cancers. N Engl J Med. 2008;359:1207-1217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 775]  [Cited by in F6Publishing: 673]  [Article Influence: 42.1]  [Reference Citation Analysis (0)]
44.  Pickhardt PJ, Choi JR, Hwang I, Butler JA, Puckett ML, Hildebrandt HA, Wong RK, Nugent PA, Mysliwiec PA, Schindler WR. Computed tomographic virtual colonoscopy to screen for colorectal neoplasia in asymptomatic adults. N Engl J Med. 2003;349:2191-2200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1495]  [Cited by in F6Publishing: 1260]  [Article Influence: 60.0]  [Reference Citation Analysis (0)]
45.  Pickhardt PJ, Hassan C, Halligan S, Marmo R. Colorectal cancer: CT colonography and colonoscopy for detection--systematic review and meta-analysis. Radiology. 2011;259:393-405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 294]  [Cited by in F6Publishing: 281]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
46.  Rosman AS, Korsten MA. Meta-analysis comparing CT colonography, air contrast barium enema, and colonoscopy. Am J Med. 2007;120:203-210.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 81]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
47.  Halligan S, Wooldrage K, Dadswell E, Kralj-Hans I, von Wagner C, Edwards R, Yao G, Kay C, Burling D, Faiz O, Teare J, Lilford RJ, Morton D, Wardle J, Atkin W; SIGGAR investigators. Computed tomographic colonography versus barium enema for diagnosis of colorectal cancer or large polyps in symptomatic patients (SIGGAR): a multicentre randomised trial. Lancet. 2013;381:1185-1193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 113]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
48.  Johnson CD, Herman BA, Chen MH, Toledano AY, Heiken JP, Dachman AH, Kuo MD, Menias CO, Siewert B, Cheema JI, Obregon R, Fidler JL, Zimmerman P, Horton KM, Coakley KJ, Iyer RB, Hara AK, Halvorsen RA, Casola G, Yee J, Blevins M, Burgart LJ, Limburg PJ, Gatsonis CA. The National CT Colonography Trial: assessment of accuracy in participants 65 years of age and older. Radiology. 2012;263:401-408.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 31]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
49.  Stoop EM, de Haan MC, de Wijkerslooth TR, Bossuyt PM, van Ballegooijen M, Nio CY, van de Vijver MJ, Biermann K, Thomeer M, van Leerdam ME, Fockens P, Stoker J, Kuipers EJ, Dekker E. Participation and yield of colonoscopy versus non-cathartic CT colonography in population-based screening for colorectal cancer: a randomised controlled trial. Lancet Oncol. 2012;13:55-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 265]  [Cited by in F6Publishing: 266]  [Article Influence: 22.2]  [Reference Citation Analysis (0)]
50.  Sakamoto T, Mitsuzaki K, Utsunomiya D, Matsuda K, Yamamura S, Urata J, Kawakami M, Yamashita Y. Detection of flat colorectal polyps at screening CT colonography in comparison with conventional polypoid lesions. Acta Radiol. 2012;53:714-719.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
51.  IJspeert JE, Tutein Nolthenius CJ, Kuipers EJ, van Leerdam ME, Nio CY, Thomeer MG, Biermann K, van de Vijver MJ, Dekker E, Stoker J. CT-Colonography vs. Colonoscopy for Detection of High-Risk Sessile Serrated Polyps. Am J Gastroenterol. 2016;111:516-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 70]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
52.  Zubarik R, Ganguly E, Benway D, Ferrentino N, Moses P, Vecchio J. Procedure-related abdominal discomfort in patients undergoing colorectal cancer screening: a comparison of colonoscopy and flexible sigmoidoscopy. Am J Gastroenterol. 2002;97:3056-3061.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 40]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
53.  Atkin WS, Edwards R, Kralj-Hans I, Wooldrage K, Hart AR, Northover JM, Parkin DM, Wardle J, Duffy SW, Cuzick J; UK Flexible Sigmoidoscopy Trial Investigators. Once-only flexible sigmoidoscopy screening in prevention of colorectal cancer: a multicentre randomised controlled trial. Lancet. 2010;375:1624-1633.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1121]  [Cited by in F6Publishing: 1096]  [Article Influence: 78.3]  [Reference Citation Analysis (0)]
54.  Holme Ø, Løberg M, Kalager M, Bretthauer M, Hernán MA, Aas E, Eide TJ, Skovlund E, Schneede J, Tveit KM, Hoff G. Effect of flexible sigmoidoscopy screening on colorectal cancer incidence and mortality: a randomized clinical trial. JAMA. 2014;312:606-615.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 279]  [Cited by in F6Publishing: 283]  [Article Influence: 28.3]  [Reference Citation Analysis (0)]
55.  Schoen RE, Pinsky PF, Weissfeld JL, Yokochi LA, Church T, Laiyemo AO, Bresalier R, Andriole GL, Buys SS, Crawford ED, Fouad MN, Isaacs C, Johnson CC, Reding DJ, O'Brien B, Carrick DM, Wright P, Riley TL, Purdue MP, Izmirlian G, Kramer BS, Miller AB, Gohagan JK, Prorok PC, Berg CD; PLCO Project Team. Colorectal-cancer incidence and mortality with screening flexible sigmoidoscopy. N Engl J Med. 2012;366:2345-2357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 730]  [Cited by in F6Publishing: 697]  [Article Influence: 58.1]  [Reference Citation Analysis (1)]
56.  Segnan N, Armaroli P, Bonelli L, Risio M, Sciallero S, Zappa M, Andreoni B, Arrigoni A, Bisanti L, Casella C, Crosta C, Falcini F, Ferrero F, Giacomin A, Giuliani O, Santarelli A, Visioli CB, Zanetti R, Atkin WS, Senore C; 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;103:1310-1322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 428]  [Cited by in F6Publishing: 427]  [Article Influence: 32.8]  [Reference Citation Analysis (0)]
57.  Atkin W, Wooldrage K, Parkin DM, Kralj-Hans I, MacRae E, Shah U, Duffy S, Cross AJ. Long term effects of once-only flexible sigmoidoscopy screening after 17 years of follow-up: the UK Flexible Sigmoidoscopy Screening randomised controlled trial. Lancet. 2017;389:1299-1311.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 250]  [Cited by in F6Publishing: 238]  [Article Influence: 34.0]  [Reference Citation Analysis (0)]
58.  Holme Ø, Schoen RE, Senore C, Segnan N, Hoff G, Løberg M, Bretthauer M, Adami HO, Kalager M. Effectiveness of flexible sigmoidoscopy screening in men and women and different age groups: pooled analysis of randomised trials. BMJ. 2017;356:i6673.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 80]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
59.  Statistics NCfH. Health, United States, 2016, With chartbook on long-term trends in health: Government Printing Office, 2017. .  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Dachman AH, Barish MA. Structured reporting and quality control in CT colonography. Abdom Radiol (NY). 2018;43:566-573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
61.  US Food and Drug Administration (FDA). Epi ProColon. Silver Spring MFhwafgscccptcipA. .  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Parikh RB, Prasad V. Blood-Based Screening for Colon Cancer: A Disruptive Innovation or Simply a Disruption? JAMA. 2016;315:2519-2520.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
63.  Ladabaum U, Alvarez-Osorio L, Rösch T, Brueggenjuergen B. Cost-effectiveness of colorectal cancer screening in Germany: current endoscopic and fecal testing strategies versus plasma methylated Septin 9 DNA. Endosc Int Open. 2014;2:E96-E104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 32]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
64.  Church TR, Wandell M, Lofton-Day C, Mongin SJ, Burger M, Payne SR, Castaños-Vélez E, Blumenstein BA, Rösch T, Osborn N, Snover D, Day RW, Ransohoff DF; PRESEPT Clinical Study Steering Committee, Investigators and Study Team. Prospective evaluation of methylated SEPT9 in plasma for detection of asymptomatic colorectal cancer. Gut. 2014;63:317-325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 486]  [Cited by in F6Publishing: 518]  [Article Influence: 51.8]  [Reference Citation Analysis (0)]
65.  Palimaka S, Blackhouse G, Goeree R. Colon Capsule Endoscopy for the Detection of Colorectal Polyps: An Economic Analysis. Ont Health Technol Assess Ser. 2015;15:1-43.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Gonzalez-Pons M, Cruz-Correa M. Colorectal Cancer Biomarkers: Where Are We Now? Biomed Res Int. 2015;2015:149014.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 113]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
67.  Liang J, Lin C, Hu F, Wang F, Zhu L, Yao X, Wang Y, Zhao Y. APC polymorphisms and the risk of colorectal neoplasia: a HuGE review and meta-analysis. Am J Epidemiol. 2013;177:1169-1179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 58]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
68.  Gelsomino F, Barbolini M, Spallanzani A, Pugliese G, Cascinu S. The evolving role of microsatellite instability in colorectal cancer: A review. Cancer Treat Rev. 2016;51:19-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 174]  [Article Influence: 21.8]  [Reference Citation Analysis (0)]
69.  Hutchins G, Southward K, Handley K, Magill L, Beaumont C, Stahlschmidt J, Richman S, Chambers P, Seymour M, Kerr D, Gray R, Quirke P. Value of mismatch repair, KRAS, and BRAF mutations in predicting recurrence and benefits from chemotherapy in colorectal cancer. J Clin Oncol. 2011;29:1261-1270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 473]  [Cited by in F6Publishing: 498]  [Article Influence: 38.3]  [Reference Citation Analysis (0)]
70.  Ulamec M, Krušlin B. Colorectal cancer, novel biomarkers and immunohistochemistry-an overview. Rad Med Sci. 2014;520:41-49.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Umar A, Boland CR, Terdiman JP, Syngal S, de la Chapelle A, Rüschoff J, Fishel R, Lindor NM, Burgart LJ, Hamelin R, Hamilton SR, Hiatt RA, Jass J, Lindblom A, Lynch HT, Peltomaki P, Ramsey SD, Rodriguez-Bigas MA, Vasen HF, Hawk ET, Barrett JC, Freedman AN, Srivastava S. Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J Natl Cancer Inst. 2004;96:261-268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2154]  [Cited by in F6Publishing: 2122]  [Article Influence: 106.1]  [Reference Citation Analysis (0)]
72.  Roth AD, Tejpar S, Delorenzi M, Yan P, Fiocca R, Klingbiel D, Dietrich D, Biesmans B, Bodoky G, Barone C, Aranda E, Nordlinger B, Cisar L, Labianca R, Cunningham D, Van Cutsem E, Bosman F. Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: results of the translational study on the PETACC-3, EORTC 40993, SAKK 60-00 trial. J Clin Oncol. 2010;28:466-474.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 801]  [Cited by in F6Publishing: 886]  [Article Influence: 59.1]  [Reference Citation Analysis (0)]
73.  Koopman M, Kortman GA, Mekenkamp L, Ligtenberg MJ, Hoogerbrugge N, Antonini NF, Punt CJ, van Krieken JH. Deficient mismatch repair system in patients with sporadic advanced colorectal cancer. Br J Cancer. 2009;100:266-273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 291]  [Cited by in F6Publishing: 338]  [Article Influence: 22.5]  [Reference Citation Analysis (0)]
74.  Koga Y, Yamazaki N, Matsumura Y. New molecular diagnosis and screening methods for colorectal cancer using fecal protein, DNA and RNA. Expert Rev Mol Diagn. 2014;14:107-120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
75.  He L, Thomson JM, Hemann MT, Hernando-Monge E, Mu D, Goodson S, Powers S, Cordon-Cardo C, Lowe SW, Hannon GJ, Hammond SM. A microRNA polycistron as a potential human oncogene. Nature. 2005;435:828-833.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2729]  [Cited by in F6Publishing: 2785]  [Article Influence: 146.6]  [Reference Citation Analysis (0)]
76.  Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281-297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25833]  [Cited by in F6Publishing: 26942]  [Article Influence: 1347.1]  [Reference Citation Analysis (0)]
77.  Wu CW, Ng SC, Dong Y, Tian L, Ng SS, Leung WW, Law WT, Yau TO, Chan FK, Sung JJ, Yu J. Identification of microRNA-135b in stool as a potential noninvasive biomarker for colorectal cancer and adenoma. Clin Cancer Res. 2014;20:2994-3002.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 103]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
78.  Kalimutho M, Di Cecilia S, Del Vecchio Blanco G, Roviello F, Sileri P, Cretella M, Formosa A, Corso G, Marrelli D, Pallone F, Federici G, Bernardini S. Epigenetically silenced miR-34b/c as a novel faecal-based screening marker for colorectal cancer. Br J Cancer. 2011;104:1770-1778.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 76]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
79.  Young GP, Bosch LJ. Fecal Tests: From Blood to Molecular Markers. Curr Colorectal Cancer Rep. 2011;7:62-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 10]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
80.  Koga Y, Yasunaga M, Takahashi A, Kuroda J, Moriya Y, Akasu T, Fujita S, Yamamoto S, Baba H, Matsumura Y. MicroRNA expression profiling of exfoliated colonocytes isolated from feces for colorectal cancer screening. Cancer Prev Res (Phila). 2010;3:1435-1442.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 162]  [Article Influence: 11.6]  [Reference Citation Analysis (1)]
81.  Hashimoto Y, Zumwalt TJ, Goel A. DNA methylation patterns as noninvasive biomarkers and targets of epigenetic therapies in colorectal cancer. Epigenomics. 2016;8:685-703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
82.  Finkelstein JD. Methionine metabolism in mammals. J Nutr Biochem. 1990;1:228-237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 917]  [Cited by in F6Publishing: 911]  [Article Influence: 47.9]  [Reference Citation Analysis (0)]
83.  Hu Y, Su L, Snow ET. Arsenic toxicity is enzyme specific and its affects on ligation are not caused by the direct inhibition of DNA repair enzymes. Mutat Res. 1998;408:203-218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 136]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
84.  Abu-Remaileh M, Bender S, Raddatz G, Ansari I, Cohen D, Gutekunst J, Musch T, Linhart H, Breiling A, Pikarsky E, Bergman Y, Lyko F. Chronic inflammation induces a novel epigenetic program that is conserved in intestinal adenomas and in colorectal cancer. Cancer Res. 2015;75:2120-2130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 73]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
85.  Galanopoulos M, Tsoukalas N, Papanikolaou IS, Tolia M, Gazouli M, Mantzaris GJ. Abnormal DNA methylation as a cell-free circulating DNA biomarker for colorectal cancer detection: A review of literature. World J Gastrointest Oncol. 2017;9:142-152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 25]  [Cited by in F6Publishing: 22]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
86.  Luo YX, Chen DK, Song SX, Wang L, Wang JP. Aberrant methylation of genes in stool samples as diagnostic biomarkers for colorectal cancer or adenomas: a meta-analysis. Int J Clin Pract. 2011;65:1313-1320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 30]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
87.  Toiyama Y, Okugawa Y, Goel A. DNA methylation and microRNA biomarkers for noninvasive detection of gastric and colorectal cancer. Biochem Biophys Res Commun. 2014;455:43-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 122]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
88.  Guo Q, Song Y, Zhang H, Wu X, Xia P, Dang C. Detection of hypermethylated fibrillin-1 in the stool samples of colorectal cancer patients. Med Oncol. 2013;30:695.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 48]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
89.  Glöckner SC, Dhir M, Yi JM, McGarvey KE, Van Neste L, Louwagie J, Chan TA, Kleeberger W, de Bruïne AP, Smits KM, Khalid-de Bakker CA, Jonkers DM, Stockbrügger RW, Meijer GA, Oort FA, Iacobuzio-Donahue C, Bierau K, Herman JG, Baylin SB, Van Engeland M, Schuebel KE, Ahuja N. Methylation of TFPI2 in stool DNA: a potential novel biomarker for the detection of colorectal cancer. Cancer Res. 2009;69:4691-4699.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 167]  [Cited by in F6Publishing: 185]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
90.  Oh T, Kim N, Moon Y, Kim MS, Hoehn BD, Park CH, Kim TS, Kim NK, Chung HC, An S. Genome-wide identification and validation of a novel methylation biomarker, SDC2, for blood-based detection of colorectal cancer. J Mol Diagn. 2013;15:498-507.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 116]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
91.  Warren JD, Xiong W, Bunker AM, Vaughn CP, Furtado LV, Roberts WL, Fang JC, Samowitz WS, Heichman KA. Septin 9 methylated DNA is a sensitive and specific blood test for colorectal cancer. BMC Med. 2011;9:133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 323]  [Cited by in F6Publishing: 304]  [Article Influence: 23.4]  [Reference Citation Analysis (0)]
92.  Tóth K, Sipos F, Kalmár A, Patai AV, Wichmann B, Stoehr R, Golcher H, Schellerer V, Tulassay Z, Molnár B. Detection of methylated SEPT9 in plasma is a reliable screening method for both left- and right-sided colon cancers. PLoS One. 2012;7:e46000.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 129]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
93.  Jing Y, Wu X, Gao P, Fang Z, Wu J, Wang Q, Li C, Zhu Z, Cao Y. Rapid differentiating colorectal cancer and colorectal polyp using dried blood spot mass spectrometry metabolomic approach. IUBMB Life. 2017;69:347-354.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 25]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
94.  Nishiumi S, Kobayashi T, Kawana S, Unno Y, Sakai T, Okamoto K, Yamada Y, Sudo K, Yamaji T, Saito Y, Kanemitsu Y, Okita NT, Saito H, Tsugane S, Azuma T, Ojima N, Yoshida M. Investigations in the possibility of early detection of colorectal cancer by gas chromatography/triple-quadrupole mass spectrometry. Oncotarget. 2017;8:17115-17126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 54]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
95.  Shen S, Yang L, Li L, Bai Y, Cai C, Liu H. A plasma lipidomics strategy reveals perturbed lipid metabolic pathways and potential lipid biomarkers of human colorectal cancer. J Chromatogr B Analyt Technol Biomed Life Sci. 2017;1068-1069:41-48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 56]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
96.  Hata T, Takemasa I, Takahashi H, Haraguchi N, Nishimura J, Hata T, Mizushima T, Doki Y, Mori M. Downregulation of serum metabolite GTA-446 as a novel potential marker for early detection of colorectal cancer. Br J Cancer. 2017;117:227-232.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
97.  Ritchie SA, Tonita J, Alvi R, Lehotay D, Elshoni H, Myat S, McHattie J, Goodenowe DB. Low-serum GTA-446 anti-inflammatory fatty acid levels as a new risk factor for colon cancer. Int J Cancer. 2013;132:355-362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 31]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
98.  Uchiyama K, Yagi N, Mizushima K, Higashimura Y, Hirai Y, Okayama T, Yoshida N, Katada K, Kamada K, Handa O, Ishikawa T, Takagi T, Konishi H, Kuriu Y, Nakanishi M, Otsuji E, Itoh Y, Naito Y. Serum metabolomics analysis for early detection of colorectal cancer. J Gastroenterol. 2017;52:677-694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 68]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
99.  Crotti S, Agnoletto E, Cancemi G, Di Marco V, Traldi P, Pucciarelli S, Nitti D, Agostini M. Altered plasma levels of decanoic acid in colorectal cancer as a new diagnostic biomarker. Anal Bioanal Chem. 2016;408:6321-6328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 27]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
100.  Eisner R, Greiner R, Tso V, Wang H, Fedorak RN. A machine-learned predictor of colonic polyps based on urinary metabolomics. Biomed Res Int. 2013;2013:303982.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
101.  Deng L, Chang D, Foshaug RR, Eisner R, Tso VK, Wishart DS, Fedorak RN. Development and Validation of a High-Throughput Mass Spectrometry Based Urine Metabolomic Test for the Detection of Colonic Adenomatous Polyps. Metabolites. 2017;7:pii: E32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
102.  Wang H, Tso V, Wong C, Sadowski D, Fedorak RN. Development and validation of a highly sensitive urine-based test to identify patients with colonic adenomatous polyps. Clin Transl Gastroenterol. 2014;5:e54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 31]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
103.  Yue H, Wang Y, Zhang Y, Ren H, Wu J, Ma L, Liu S. A metabonomics study of colorectal cancer by RRLC-QTOF/MS. J Liq Chromatogr Relat Technol. 2013;36:428-438.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  Cheng Y, Xie G, Chen T, Qiu Y, Zou X, Zheng M, Tan B, Feng B, Dong T, He P, Zhao L, Zhao A, Xu LX, Zhang Y, Jia W. Distinct urinary metabolic profile of human colorectal cancer. J Proteome Res. 2012;11:1354-1363.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 159]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
105.  Nakajima T, Katsumata K, Kuwabara H, Soya R, Enomoto M, Ishizaki T, Tsuchida A, Mori M, Hiwatari K, Soga T, Tomita M, Sugimoto M. Urinary Polyamine Biomarker Panels with Machine-Learning Differentiated Colorectal Cancers, Benign Disease, and Healthy Controls. Int J Mol Sci. 2018;19:pii: E756.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 34]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
106.  Hiramatsu K, Takahashi K, Yamaguchi T, Matsumoto H, Miyamoto H, Tanaka S, Tanaka C, Tamamori Y, Imajo M, Kawaguchi M, Toi M, Mori T, Kawakita M. N(1),N(12)-Diacetylspermine as a sensitive and specific novel marker for early- and late-stage colorectal and breast cancers. Clin Cancer Res. 2005;11:2986-2990.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 60]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
107.  Phua LC, Chue XP, Koh PK, Cheah PY, Ho HK, Chan EC. Non-invasive fecal metabonomic detection of colorectal cancer. Cancer Biol Ther. 2014;15:389-397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 54]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
108.  Amiot A, Dona AC, Wijeyesekera A, Tournigand C, Baumgaertner I, Lebaleur Y, Sobhani I, Holmes E. (1)H NMR Spectroscopy of Fecal Extracts Enables Detection of Advanced Colorectal Neoplasia. J Proteome Res. 2015;14:3871-3881.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
109.  Lindon JC, Nicholson JK, Holmes E, Keun HC, Craig A, Pearce JT, Bruce SJ, Hardy N, Sansone SA, Antti H, Jonsson P, Daykin C, Navarange M, Beger RD, Verheij ER, Amberg A, Baunsgaard D, Cantor GH, Lehman-McKeeman L, Earll M, Wold S, Johansson E, Haselden JN, Kramer K, Thomas C, Lindberg J, Schuppe-Koistinen I, Wilson ID, Reily MD, Robertson DG, Senn H, Krotzky A, Kochhar S, Powell J, van der Ouderaa F, Plumb R, Schaefer H, Spraul M; Standard Metabolic Reporting Structures working group. Summary recommendations for standardization and reporting of metabolic analyses. Nat Biotechnol. 2005;23:833-838.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 208]  [Cited by in F6Publishing: 158]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
110.  Claudino WM, Goncalves PH, di Leo A, Philip PA, Sarkar FH. Metabolomics in cancer: a bench-to-bedside intersection. Crit Rev Oncol Hematol. 2012;84:1-7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 58]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
111.  O'Connell TM. Recent advances in metabolomics in oncology. Bioanalysis. 2012;4:431-451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 46]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
112.  Hardcastle JD, Chamberlain JO, Robinson MH, Moss SM, Amar SS, Balfour TW, James PD, Mangham CM. Randomised controlled trial of faecal-occult-blood screening for colorectal cancer. Lancet. 1996;348:1472-1477.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1858]  [Cited by in F6Publishing: 1806]  [Article Influence: 64.5]  [Reference Citation Analysis (0)]
113.  Kronborg O, Fenger C, Olsen J, Jørgensen OD, Søndergaard O. Randomised study of screening for colorectal cancer with faecal-occult-blood test. Lancet. 1996;348:1467-1471.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1640]  [Cited by in F6Publishing: 1582]  [Article Influence: 56.5]  [Reference Citation Analysis (0)]
114.  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;95:1029-1036.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 208]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
115.  Mandel JS, Church TR, Ederer F, Bond JH. Colorectal cancer mortality: effectiveness of biennial screening for fecal occult blood. J Natl Cancer Inst. 1999;91:434-437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 529]  [Cited by in F6Publishing: 569]  [Article Influence: 22.8]  [Reference Citation Analysis (0)]
116.  Hewitson P, Glasziou P, Irwig L, Towler B, Watson E. Screening for colorectal cancer using the faecal occult blood test, Hemoccult. Cochrane Database Syst Rev. 2007;CD001216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 183]  [Cited by in F6Publishing: 289]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
117.  Senore C, Ederle A, Fantin A, Andreoni B, Bisanti L, Grazzini G, Zappa M, Ferrero F, Marutti A, Giuliani O, Armaroli P, Segnan N. Acceptability and side-effects of colonoscopy and sigmoidoscopy in a screening setting. J Med Screen. 2011;18:128-134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 59]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
118.  Taylor SA, Halligan S, Saunders BP, Bassett P, Vance M, Bartram CI. Acceptance by patients of multidetector CT colonography compared with barium enema examinations, flexible sigmoidoscopy, and colonoscopy. AJR Am J Roentgenol. 2003;181:913-921.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 109]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
119.  Piñol V, Castells A, Andreu M, Castellví-Bel S, Alenda C, Llor X, Xicola RM, Rodríguez-Moranta F, Payá A, Jover R, Bessa X; Gastrointestinal Oncology Group of the Spanish Gastroenterological Association. Accuracy of revised Bethesda guidelines, microsatellite instability, and immunohistochemistry for the identification of patients with hereditary nonpolyposis colorectal cancer. JAMA. 2005;293:1986-1994.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 389]  [Cited by in F6Publishing: 424]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
120.  Evaluation of Genomic Applications in Practice and Prevention (EGAPP) Working Group. Recommendations from the EGAPP Working Group: genetic testing strategies in newly diagnosed individuals with colorectal cancer aimed at reducing morbidity and mortality from Lynch syndrome in relatives. Genet Med. 2009;11:35-41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 468]  [Cited by in F6Publishing: 496]  [Article Influence: 33.1]  [Reference Citation Analysis (0)]
121.  Grützmann R, Molnar B, Pilarsky C, Habermann JK, Schlag PM, Saeger HD, Miehlke S, Stolz T, Model F, Roblick UJ, Bruch HP, Koch R, Liebenberg V, Devos T, Song X, Day RH, Sledziewski AZ, Lofton-Day C. Sensitive detection of colorectal cancer in peripheral blood by septin 9 DNA methylation assay. PLoS One. 2008;3:e3759.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 281]  [Cited by in F6Publishing: 291]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
122.  Zhang J, Yang Q, Li J, Zhong Y, Zhang L, Huang Q, Chen B, Mo M, Shen S, Zhong Q, Liu H, Cai C. Distinct differences in serum eicosanoids in healthy, enteritis and colorectal cancer individuals. Metabolomics. 2017;14:4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 4]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
123.  Guo Y, Ren J, Li X, Liu X, Liu N, Wang Y, Li Z. Simultaneous Quantification of Serum Multi-Phospholipids as Potential Biomarkers for Differentiating Different Pathophysiological states of lung, stomach, intestine, and pancreas. J Cancer. 2017;8:2191-2204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 20]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
124.  Farshidfar F, Weljie AM, Kopciuk KA, Hilsden R, McGregor SE, Buie WD, MacLean A, Vogel HJ, Bathe OF. A validated metabolomic signature for colorectal cancer: exploration of the clinical value of metabolomics. Br J Cancer. 2016;115:848-857.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 99]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
125.  Zhang Y, He C, Qiu L, Wang Y, Qin X, Liu Y, Li Z. Serum Unsaturated Free Fatty Acids: A Potential Biomarker Panel for Early-Stage Detection of Colorectal Cancer. J Cancer. 2016;7:477-483.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 27]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
126.  Cowan MJ, Smith W, Ammann AJ. Interleukin 2 responsive lymphocytes in patients with adenosine deaminase deficiency. Clin Immunol Immunopathol. 1989;53:59-67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 33]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
127.  Zhu J, Djukovic D, Deng L, Gu H, Himmati F, Chiorean EG, Raftery D. Colorectal cancer detection using targeted serum metabolic profiling. J Proteome Res. 2014;13:4120-4130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 160]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
128.  Li F, Qin X, Chen H, Qiu L, Guo Y, Liu H, Chen G, Song G, Wang X, Li F, Guo S, Wang B, Li Z. Lipid profiling for early diagnosis and progression of colorectal cancer using direct-infusion electrospray ionization Fourier transform ion cyclotron resonance mass spectrometry. Rapid Commun Mass Spectrom. 2013;27:24-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 80]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
129.  Tan B, Qiu Y, Zou X, Chen T, Xie G, Cheng Y, Dong T, Zhao L, Feng B, Hu X, Xu LX, Zhao A, Zhang M, Cai G, Cai S, Zhou Z, Zheng M, Zhang Y, Jia W. Metabonomics identifies serum metabolite markers of colorectal cancer. J Proteome Res. 2013;12:3000-3009.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 145]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
130.  Ma Y, Zhang P, Wang F, Liu W, Yang J, Qin H. An integrated proteomics and metabolomics approach for defining oncofetal biomarkers in the colorectal cancer. Ann Surg. 2012;255:720-730.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 79]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
131.  Nishiumi S, Kobayashi T, Ikeda A, Yoshie T, Kibi M, Izumi Y, Okuno T, Hayashi N, Kawano S, Takenawa T, Azuma T, Yoshida M. A novel serum metabolomics-based diagnostic approach for colorectal cancer. PLoS One. 2012;7:e40459.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 194]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
132.  Ritchie SA, Ahiahonu PW, Jayasinghe D, Heath D, Liu J, Lu Y, Jin W, Kavianpour A, Yamazaki Y, Khan AM, Hossain M, Su-Myat KK, Wood PL, Krenitsky K, Takemasa I, Miyake M, Sekimoto M, Monden M, Matsubara H, Nomura F, Goodenowe DB. Reduced levels of hydroxylated, polyunsaturated ultra long-chain fatty acids in the serum of colorectal cancer patients: implications for early screening and detection. BMC Med. 2010;8:13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 111]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
133.  Ludwig C, Ward DG, Martin A, Viant MR, Ismail T, Johnson PJ, Wakelam MJ, Günther UL. Fast targeted multidimensional NMR metabolomics of colorectal cancer. Magn Reson Chem. 2009;47 Suppl 1:S68-S73.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 54]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
134.  Ikeda A, Nishiumi S, Shinohara M, Yoshie T, Hatano N, Okuno T, Bamba T, Fukusaki E, Takenawa T, Azuma T, Yoshida M. Serum metabolomics as a novel diagnostic approach for gastrointestinal cancer. Biomed Chromatogr. 2012;26:548-558.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 102]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
135.  Leichtle AB, Nuoffer JM, Ceglarek U, Kase J, Conrad T, Witzigmann H, Thiery J, Fiedler GM. Serum amino acid profiles and their alterations in colorectal cancer. Metabolomics. 2012;8:643-653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 106]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
136.  Li S, Guo B, Song J, Deng X, Cong Y, Li P, Zhao K, Liu L, Xiao G, Xu F. Plasma choline-containing phospholipids: potential biomarkers for colorectal cancer progression. Metabolomics. 2013;9:202-212.  [PubMed]  [DOI]  [Cited in This Article: ]
137.  Miyagi Y, Higashiyama M, Gochi A, Akaike M, Ishikawa T, Miura T, Saruki N, Bando E, Kimura H, Imamura F, Moriyama M, Ikeda I, Chiba A, Oshita F, Imaizumi A, Yamamoto H, Miyano H, Horimoto K, Tochikubo O, Mitsushima T, Yamakado M, Okamoto N. Plasma free amino acid profiling of five types of cancer patients and its application for early detection. PLoS One. 2011;6:e24143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 279]  [Cited by in F6Publishing: 307]  [Article Influence: 23.6]  [Reference Citation Analysis (0)]
138.  Okamoto N, Miyagi Y, Chiba A, Akaike M, Shiozawa M, Imaizumi A, Yamamoto H, Ando T, Yamakado M, Tochikubo O. Diagnostic modeling with differences in plasma amino acid profiles between non-cachectic colorectal/breast cancer patients and healthy individuals. Int J Medicine Med Sci. 2009;1:001-008.  [PubMed]  [DOI]  [Cited in This Article: ]
139.  Zhao Z, Xiao Y, Elson P, Tan H, Plummer SJ, Berk M, Aung PP, Lavery IC, Achkar JP, Li L, Casey G, Xu Y. Plasma lysophosphatidylcholine levels: potential biomarkers for colorectal cancer. J Clin Oncol. 2007;25:2696-2701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 160]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
140.  Liu Z, Cui C, Wang X, Fernandez-Escobar A, Wu Q, Xu K, Mao J, Jin M, Wang K. Plasma Levels of Homocysteine and the Occurrence and Progression of Rectal Cancer. Med Sci Monit. 2018;24:1776-1783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
141.  Cavia-Saiz M, Muñiz Rodríguez P, Llorente Ayala B, García-González M, Coma-Del Corral MJ, García Girón C. The role of plasma IDO activity as a diagnostic marker of patients with colorectal cancer. Mol Biol Rep. 2014;41:2275-2279.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
142.  Deng L, Fang H, Tso VK, Sun Y, Foshaug RR, Krahn SC, Zhang F, Yan Y, Xu H, Chang D, Zhang Y, Fedorak RN. Clinical validation of a novel urine-based metabolomic test for the detection of colonic polyps on Chinese population. Int J Colorectal Dis. 2017;32:741-743.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 16]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
143.  Wang Z, Lin Y, Liang J, Huang Y, Ma C, Liu X, Yang J. NMR-based metabolomic techniques identify potential urinary biomarkers for early colorectal cancer detection. Oncotarget. 2017;8:105819-105831.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 40]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
144.  Rozalski R, Gackowski D, Siomek-Gorecka A, Starczak M, Modrzejewska M, Banaszkiewicz Z, Olinski R. Urinary 5-hydroxymethyluracil and 8-oxo-7,8-dihydroguanine as potential biomarkers in patients with colorectal cancer. Biomarkers. 2015;20:287-291.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
145.  Hsu WY, Chen CJ, Huang YC, Tsai FJ, Jeng LB, Lai CC. Urinary nucleosides as biomarkers of breast, colon, lung, and gastric cancer in Taiwanese. PLoS One. 2013;8:e81701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 40]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
146.  Chen JL, Fan J, Yan LS, Guo HQ, Xiong JJ, Ren Y, Hu JD. Urine Metabolite Profiling of Human Colorectal Cancer by Capillary Electrophoresis Mass Spectrometry Based on MRB. Gastroenterol Res Pract. 2012;2012:125890.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 25]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
147.  Wang W, Feng B, Li X, Yin P, Gao P, Zhao X, Lu X, Zheng M, Xu G. Urinary metabolic profiling of colorectal carcinoma based on online affinity solid phase extraction-high performance liquid chromatography and ultra performance liquid chromatography-mass spectrometry. Mol Biosyst. 2010;6:1947-1955.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
148.  Feng B, Zheng MH, Zheng YF, Lu AG, Li JW, Wang ML, Ma JJ, Xu GW, Liu BY, Zhu ZG. Normal and modified urinary nucleosides represent novel biomarkers for colorectal cancer diagnosis and surgery monitoring. J Gastroenterol Hepatol. 2005;20:1913-1919.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 57]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
149.  Zheng YF, Yang J, Zhao XJ, Feng B, Kong HW, Chen YJ, Lv S, Zheng MH, Xu GW. Urinary nucleosides as biological markers for patients with colorectal cancer. World J Gastroenterol. 2005;11:3871-3876.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 55]  [Cited by in F6Publishing: 44]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
150.  Johnson JC, Schmidt CR, Shrubsole MJ, Billheimer DD, Joshi PR, Morrow JD, Heslin MJ, Washington MK, Ness RM, Zheng W, Schwartz DA, Coffey RJ, Beauchamp RD, Merchant NB. Urine PGE-M: A metabolite of prostaglandin E2 as a potential biomarker of advanced colorectal neoplasia. Clin Gastroenterol Hepatol. 2006;4:1358-1365.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 69]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
151.  Bezabeh T, Somorjai R, Dolenko B, Bryskina N, Levin B, Bernstein CN, Jeyarajah E, Steinhart AH, Rubin DT, Smith IC. Detecting colorectal cancer by 1H magnetic resonance spectroscopy of fecal extracts. NMR Biomed. 2009;22:593-600.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 41]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
152.  Lin Y, Ma C, Liu C, Wang Z, Yang J, Liu X, Shen Z, Wu R. NMR-based fecal metabolomics fingerprinting as predictors of earlier diagnosis in patients with colorectal cancer. Oncotarget. 2016;7:29454-29464.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 87]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]