Abstract
The early detection of colorectal cancer with effective screening is essential for reduction of cancer-specific mortality. The addition of fecal DNA testing in the armamentarium of screening methods already in clinical use launches a new era in the noninvasive part of colorectal cancer screening and emanates from a large number of previous and ongoing clinical investigations and technological advancements. In this review, we discuss the molecular rational and most important genetic alterations hallmarking the early colorectal carcinogenesis process. Also, representative DNA targets-markers and key aspects of their testing at the clinical level in comparison or/and association with other screening methods are described. Finally, a critical view of the strengths and limitations of fecal DNA tests is provided, along with anticipated barriers and suggestions for further exploitation of their use.
Keywords: Colorectal cancer, Screening, Fecal DNA, Cologuard®, Adenoma
Core tip: The molecular DNA targets from genetic and epigenetic alterations hallmarking colorectal carcinogenesis are reviewed here in the context of fecal testing. Also, comparison with other screening methods in terms of limitations, advantages and future perspectives of fecal DNA tests are discussed.
INTRODUCTION
Colorectal cancer (CRC) is the third most common cancer in men and women and accounts for 8% of all cancer-related deaths[1]. The incidence of CRC varies within different geographic locations and racial/ethnic groups. These differences may be related with different dietary and environmental exposures in association with a different genotype-driven susceptibility[2]. Screening for CRC plays a key role in reduction of CRC-related mortality, and the observed decline in the incidence of CRC since the mid-1980s is a striking proof of this effect, along with changes in risk factors[1].
CRC screening may be divided into two main categories: (1) biological sample-based tests, including fecal, blood and urine tests, as well as (2) colon structure-based and image-based tests, including flexible sigmoidoscopy, total colonoscopy, CT colonography and double-contrast barium enema[3,4]. Stool-based tests, including guaiac-based fecal occult blood test (g-FOBT), and the newer ones, fecal immunochemical test (FIT) and stool DNA test are already included in the American Cancer Society recommendations for CRC screening[4].
MOLECULAR RATIONAL FOR FECAL DNA TESTING
The detection of altered DNA from cancerous and pre-cancerous lesions of the colonic mucosa is based on the natural exfoliation of these cells and is further facilitated by their high degree of “integrity” compared to DNA from stools of healthy patients. Accumulating data on key mutations occurring during the early stages of colon carcinogenesis including K-Ras, adenoma polyposis coli (APC), and p53, as well as epigenetic changes such as microsatellite instability (MSI), has guided the targeted development of clinically relevant detection tests[5].
The genetic heterogeneity of CRC is essentially the reason underlying the concept of targeting multiple DNA markers. K-Ras encodes a RAS family protein which is a GTPase involved in many downstream signal transduction pathways[6]. The mutation is found in 13%-95% of CRC patients and is one of the initial mutations in colon carcinogenesis[6]. APC is an important tumor suppressor gene product involved in the Wnt/β-catenin signaling pathway, which in turn is a transcription regulator of several growth-controlling genes, including the oncogene MYC[7]. Thus it is not surprising that mutation or inactivation of the APC protein is a driver of inherited (familial adenomatous polyposis) and sporadic forms of CRC, occuring in the early stages of transition from adenoma to carcinoma[7]. Another tumor suppressor gene, p53 is found deleted or mutated in 30%-60% of CRC tumors[8]. Given its critical role in cell cycle control, apoptosis, and DNA damage response, p53 aberrations ultimately promote the development of increased genomic instability which facilitates transformation of colorectal adenomas to cancer[7].
MSI is a condition of genetic hypermutability within tandem repeats of short nucleotide sequences, the microsatellites, that results from impaired DNA mismatch repair (MMR) and is a frequent event in cancers, including 15% of all CRC[9]. The most common cause of sporadic MSI is epigenetic silencing of MMR genes, such as MLH1 due to promoter hypermethylation[7] and there are several MSI markers (BAT25, BAT26, D2S123, D5S346, and D17S2720) for detection of MSI with polymerase chain reaction. The clinical relevance of MSI lies in the fact that patients with MSI positive tumors have better prognosis and longer overall survival compared with non-MSI tumors[9].
Epigenetic methylation of gene promoters is a central mechanism that can promote carcinogenesis in the appropriate context and several preclinical studies have identified hypermethylated genes in stool samples from CRC patients, which are strikingly un-methylated in normal epithelial cells[9]. Characteristic examples include the genes secreted frizzled-related protein (SFRP), vimentin, MGMT, FBN1, and p16[7]. In addition, the panel of methylated genes varies depending on the different stages of carcinogenesis, involving (1) SLC5A8, SFRP1, SFRP2, CDH13, CRBP1, RUNX3, MINT1 and MINT31 from normal colon mucosa to aberrant crypt focus formation; (2) p14, HLTF, ITGA4, p16, CDH1, and ESR1 from aberrant crypt focus to adenoma formation; and (3) TIMP3, CXCL12, ID4, and IRF8 from adenoma to carcinoma formation and metastatic progression of CRC[7].
CLINICAL STUDIES OF FECAL DNA TESTS
An important limiting factor for developing a screening stool test with high sensitivity is the fact that only 0.01% of total fecal DNA is human and the tumor DNA is only a small percentage of the former[10].
K-RAS was the first gene tested for mutations in feces from CRC patients[11-13]. A comparative study assessed gFOBT and a fecal DNA test analyzing a panel of 21 gene mutations[14]. Imperiale et al[14] concluded that the multitarget fecal DNA test detected more invasive cancers plus adenomas with high-grade dysplasia than did gFOBT (40.8% vs 14.1%) without compromising specificity (94.4% vs 95.2%). In a blinded, multicenter, case-control study, with cases including CRC, advanced adenoma (AA), or sessile serrated adenoma ≥ 1 cm (SSA), an automated multitarget stool DNA assay was able to detect AA with high-grade dysplasia with 83% sensitivity[15]. Another blinded, multicenter, case-control study assessing a similar panel of DNA markers identified 85% of patients with CRC and 54% with AA, without sensitivity differences based on location, but with tumor size affecting detection rates[16].
More recently, Imperiale et al[17] reported their results from comparison of fecal DNA to FIT in a huge patient population who had a complete screening colonoscopy (n = 9989). The sensitivity of fecal DNA test including evaluation of KRAS mutations, aberrant NDRG4 and BMP3 methylation, B-actin and a hemoglobin assay was superior to that of FIT (92.3% vs 73.8%). However, in addition to a lower specificity of fecal DNA and the lack of comparison with repeated FIT applications over time, a far higher number of patients (n = 689) were excluded due to problematic fecal DNA testing, compared to those who underwent FIT (n = 34)[18].
A systematic review of the literature for studies of biomarkers for early detection of colorectal cancer and polyps since 2007, disclosed overall sensitivities for colorectal cancer detection by fecal DNA markers ranging from 53% to 87%, with varying specificities above 76%[19]. The diversity and combinations of various fecal DNA markers with the corresponding sensitivities and specificities per study[12-17,20-28] are summarized in Table 1.
Table 1.
Ref. | Marker |
Sensitivity |
Specificity | |
CRC | Adenoma > 1 cm | |||
[12] | Meth BMP3, hDNA, KRAS, APC | 67 (91) | 21 (78) | 85 (85) |
[13] | APC, KRAS, p53, long DNA | 3 (25) | 47 (8) | 2246 (96) |
[14] | APC, KRAS, p53, long DNA | 16 (52) | 84 (12) | 1344 (94) |
[15] | β-actin, KRAS, meth BMP3 and NDRG4, fecal hemoglobin | 91 (98) | 48 (57) | 139 (90) |
[16] | KRAS, a actina Meth NDRG4, BMP3, vimentin, TFPI2 | 214 (85) | 72 (54) | 264 (90) |
[17] | KRAS, NDRG4, BMP3, β-actin, fecal hemoglobin | 60 (92) | 321 (42) | 4457 (90) |
[20] | Meth vimentin | 9 (41) | 9 (45) | 63 (95) |
[21] | Meth SFRP2 | 60 (87) | 21 (62) | 28 (93) |
[22] | Meth TFPI2, long DNA | 52 (87) | 4 (44) | 25 (83) |
[23] | Meth SFRP2, HPPI, MGMT | 50 (96) | 15 (71) | 23 (96) |
[24] | Meth APC, ATM, hMLH1, sFRP2, HLTF, MGMT, and GSTP1 | 15 (75) | 17 (68) | 27 (90) |
[25] | Meth vimentin, long DNA | 68 (83) | 6 (86) | 298 (82) |
[26] | Meth RASSF2 or SFRP2 | 63 (75) | 25 (44) | 101 (89) |
[27] | Meth vimentin, MLH1, MGMT | 45 (75) | 31 (60) | 32 (87) |
[28] | Meth RARB2, p16INK4a, MGMT, APC | 16 (62) | 8 (40) | 20 (100) |
Adapted from Ref.[38]. Copyright 2014 by Baishideng Publishing Group Inc. Adapted with permission. CRC: Colorectal cancer.
EVOLUTION OF FECAL DNA TESTING METHODOLOGY AND TECHNIQUES
Initially, the first fecal DNA tests were performed without stabilizing buffers, resulting in low sensitivities[13,14]. Upon incorporation of stabilizing buffers and introduction of more sensitive detection techniques such as the digital melt curve method and beads, emulsion, amplification, and magnetics (BEAMing), the initial detection threshold of 1% of mutated copies was decreased to less than 0.1%[10,12].
Furthermore, implementation of the allele-specific quantitative real-time target and signal amplification (QuARTS) technique led to detection of less frequent mutations, thus improving the sensitivity for AA[12]. Another technique termed fluorescent long DNA (FL-DNA), allows for identification of tumor DNA fragments longer than 150-200 base pairs, given that cancer cells evade apoptosis and subsequent DNA degradation. FL-DNA detects CRC with a sensitivity of 80%[29]. Other advances that have been introduced in different studies include neutralization of bacterial enzymes with EDTA[30], enrichment of the panel of DNA markers (e.g., vimentin gene), and inclusion of hemoglobin detection in the same panel[16,31].
STRENGTHS AND LIMITATIONS OF FECAL DNA TESTS
A major advantage of fecal DNA tests as compared to either FOBT or colonoscopy is the fact that they are not affected by proximal location of tumors[32,33]. Another advantage is the lack of need for purging or dietary changes.
However, the sensitivity of fecal DNA tests appears to be lower for adenomas when compared to CRC detection (Table 1). In addition, although there is evidence of reductions in CRC incidence and mortality from randomized controlled trials of fecal occult blood test (FOBT) screening[34], similar data are lacking for fecal DNA tests.
Other technical difficulties may involve the burden of large volume stool collection and shipping for the patients undergoing screening[31]. In addition, the fact that in the latest study of Imperiale et al[17] the DNA tests had over twice as many abnormal results as FIT, with a higher rate of false-positive results implies that more colonoscopies would be needed to further evaluate for CRC in the former arm. Thus, the inevitably higher number of diagnostic testing would increase the costs and risks of screening. Only with the current screening method of gFOBT, 690011 colonoscopies for false positive screening tests result in an additional estimated annual cost of £800000000[19].
Cost-effectiveness per se seems to be a major disadvantage of fecal DNA tests as both older and newer studies, particularly based on a Markov model, have concluded that fecal DNA is cost-effective only when compared with no screening, but is essentially dominated by most of the other available screening options, including FOBT and colonoscopy[36,37]. This may necessitate the limitation of number of DNA markers to render their clinical use more reasonable[38].
CURRENT STATUS OF FECAL DNA TESTING (COLOGUARD®)
The United States Food and Drug Administration has recently approved Cologuard® (Exact Sciences Corporation, Madison, WI, United States), a multitarget stool DNA test in CRC screening[39]. The frequency of interval testing was determined to be every 3 years with adequate Medicare coverage[40]. Cologuard® incorporates molecular assays for aberrantly methylated BMP3 and NDRG4 gene promoter regions, mutant KRAS and β-actin as well as an immunochemical assay for human hemoglobin. It is based on the recent study of Imperiale et al[17] which showed a significantly better sensitivity for cancer detection compared to FIT. Further laboratory-based processing of the samples is necessary, entailing amplification and detection with the use of Quantitative Allele-specific Real-time Target and Signal Amplification (QuARTSTM) technology[41].
FUTURE PERSPECTIVES FOR FECAL DNA SCREENING TESTS
The combined use of screening tests would likely maximize the benefits of different biomarkers for early detection of CRC and adenomas. However, synchronous implementation of these tests in a mass screening program would not fulfill the cost-effectiveness requirement for clinical use.
Thus, there is a need for prospectively designed, systematic evaluations of the most promising fecal tests in a well-defined, large-scale screening population, with standardized sample collection, processing, and storage. This assessment should be combined with sigmoidoscopy or colonoscopy screening and ideally involve repeated testing and longitudinal monitoring of the screened population[19]. Another parameter that merits prospective evaluation is the clinical significance of fecal DNA-positive results in patients with negative colonoscopy results[40].
In the future, Imperiale and colleagues plan to “take this work forward by conducting a post-approval study, which will inform the important issue of test interval, that is, how often does the test need to be repeated”. They will also conduct computer simulation studies that will inform comparative effectiveness and cost-effectiveness relative to other screening tests and strategies[42].
Given the high sensitivity for CRC that is unaffected by tumor location and its superior sensitivity over FIT for detection of SSA and AA with greatest risk of progression, Cologuard® may be a good candidate for interval testing after initial colonoscopy. For the same reason, in cases of poor preparation or incomplete colonoscopy, it might represent a convenient follow-up screening test alternative to repeat colonoscopy or other CT colonography, particularly for those patients who are either unable or unwilling to undergo repeat bowel preparation and invasive endoscopy[40].
In an expanding view, fecal DNA testing could be implemented as a screening in CRC predisposing conditions, such as inflammatory bowel disease, playing a role complementary to colonoscopy for early dysplasia detection and surveillance[40,43]. A relevant multicenter validation study has recently been initiated (Government-registered Trial: NCT01819766) and its results are eagerly awaited.
Finally, technological advancements in detection assays of small fragment DNA from stool may render the identification of altered DNA shed from upper GI pre-cancerous and malignant lesions feasible[44-46].
Discussion of screening tests involving non-DNA (e.g., mRNA, miRNA) or non-fecal origin (e.g., blood, urine) biomarkers was beyond the scope of this review. However, it is reasonable to assume that fecal shedding of tumor DNA is an earlier event compared to inner tissue and bloodstream invasion, and is also directly related to the natural, constant process of luminal colonic mucosa exfoliation; thus rendering fecal testing more timely sensitive for the purpose of screening.
Collectively, the accumulation of experience from clinical use of Cologuard® and the numerous ongoing studies on a plethora of biomarkers, as well as further technological advancement of colonoscopy with the full-spectrum endoscopy[47] are expected to further elucidate and expand the landscape of CRC screening research in the coming years, with the hope of further reducing CRC-specific mortality through earlier and accurate detection of pre-cancerous lesions.
Footnotes
Conflict-of-interest statement: There is no conflict of interest related to this paper.
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/
Peer-review started: May 7, 2015
First decision: June 2, 2015
Article in press: August 28, 2015
P- Reviewer: Cao H S- Editor: Ma YJ L- Editor: A E- Editor: Wu HL
References
- 1.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65:5–29. doi: 10.3322/caac.21254. [DOI] [PubMed] [Google Scholar]
- 2.Murphy G, Devesa SS, Cross AJ, Inskip PD, McGlynn KA, Cook MB. Sex disparities in colorectal cancer incidence by anatomic subsite, race and age. Int J Cancer. 2011;128:1668–1675. doi: 10.1002/ijc.25481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Stracci F, Zorzi M, Grazzini G. Colorectal cancer screening: tests, strategies, and perspectives. Front Public Health. 2014;2:210. doi: 10.3389/fpubh.2014.00210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Available from: http://www.cancer.org/acs/groups/cid/documents/webcontent/003170-pdf.pdf.
- 5.Vatandoost N, Ghanbari J, Mojaver M, Avan A, Ghayour-Mobarhan M, Nedaeinia R, Salehi R. Early detection of colorectal cancer: from conventional methods to novel biomarkers. J Cancer Res Clin Oncol. 2015:Feb 17; Epub ahead of print. doi: 10.1007/s00432-015-1928-z. [DOI] [PubMed] [Google Scholar]
- 6.Tanaka T, Tanaka M, Tanaka T, Ishigamori R. Biomarkers for colorectal cancer. Int J Mol Sci. 2010;11:3209–3225. doi: 10.3390/ijms11093209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Coppedè F, Lopomo A, Spisni R, Migliore L. Genetic and epigenetic biomarkers for diagnosis, prognosis and treatment of colorectal cancer. World J Gastroenterol. 2014;20:943–956. doi: 10.3748/wjg.v20.i4.943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Kim HJ, Yu MH, Kim H, Byun J, Lee C. Noninvasive molecular biomarkers for the detection of colorectal cancer. BMB Rep. 2008;41:685–692. doi: 10.5483/bmbrep.2008.41.10.685. [DOI] [PubMed] [Google Scholar]
- 9.Wang X, Kuang YY, Hu XT. Advances in epigenetic biomarker research in colorectal cancer. World J Gastroenterol. 2014;20:4276–4287. doi: 10.3748/wjg.v20.i15.4276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Diehl F, Schmidt K, Durkee KH, Moore KJ, Goodman SN, Shuber AP, Kinzler KW, Vogelstein B. Analysis of mutations in DNA isolated from plasma and stool of colorectal cancer patients. Gastroenterology. 2008;135:489–498. doi: 10.1053/j.gastro.2008.05.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Sidransky D, Tokino T, Hamilton SR, Kinzler KW, Levin B, Frost P, Vogelstein B. Identification of ras oncogene mutations in the stool of patients with curable colorectal tumors. Science. 1992;256:102–105. doi: 10.1126/science.1566048. [DOI] [PubMed] [Google Scholar]
- 12.Zou H, Taylor WR, Harrington JJ, Hussain FT, Cao X, Loprinzi CL, Levine TR, Rex DK, Ahnen D, Knigge KL, et al. High detection rates of colorectal neoplasia by stool DNA testing with a novel digital melt curve assay. Gastroenterology. 2009;136:459–470. doi: 10.1053/j.gastro.2008.10.023. [DOI] [PubMed] [Google Scholar]
- 13.Ahlquist DA, Sargent DJ, Loprinzi CL, Levin TR, Rex DK, Ahnen DJ, Knigge K, Lance MP, Burgart LJ, Hamilton SR, et al. Stool DNA and occult blood testing for screen detection of colorectal neoplasia. Ann Intern Med. 2008;149:441–450, W81. doi: 10.7326/0003-4819-149-7-200810070-00004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Imperiale TF, Ransohoff DF, Itzkowitz SH, Turnbull BA, Ross ME. Fecal DNA versus fecal occult blood for colorectal-cancer screening in an average-risk population. N Engl J Med. 2004;351:2704–2714. doi: 10.1056/NEJMoa033403. [DOI] [PubMed] [Google Scholar]
- 15.Lidgard GP, Domanico MJ, Bruinsma JJ, Light J, Gagrat ZD, Oldham-Haltom RL, Fourrier KD, Allawi H, Yab TC, Taylor WR, et al. Clinical performance of an automated stool DNA assay for detection of colorectal neoplasia. Clin Gastroenterol Hepatol. 2013;11:1313–1318. doi: 10.1016/j.cgh.2013.04.023. [DOI] [PubMed] [Google Scholar]
- 16.Ahlquist DA, Zou H, Domanico M, Mahoney DW, Yab TC, Taylor WR, Butz ML, Thibodeau SN, Rabeneck L, Paszat LF, et al. Next-generation stool DNA test accurately detects colorectal cancer and large adenomas. Gastroenterology. 2012;142:248–256; quiz e25-26. doi: 10.1053/j.gastro.2011.10.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.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. doi: 10.1056/NEJMoa1311194. [DOI] [PubMed] [Google Scholar]
- 18.Robertson DJ, Dominitz JA. Stool DNA and colorectal-cancer screening. N Engl J Med. 2014;370:1350–1351. doi: 10.1056/NEJMe1400092. [DOI] [PubMed] [Google Scholar]
- 19.Shah R, Jones E, Vidart V, Kuppen PJ, Conti JA, Francis NK. Biomarkers for early detection of colorectal cancer and polyps: systematic review. Cancer Epidemiol Biomarkers Prev. 2014;23:1712–1728. doi: 10.1158/1055-9965.EPI-14-0412. [DOI] [PubMed] [Google Scholar]
- 20.Li M, Chen WD, Papadopoulos N, Goodman SN, Bjerregaard NC, Laurberg S, Levin B, Juhl H, Arber N, Moinova H, et al. Sensitive digital quantification of DNA methylation in clinical samples. Nat Biotechnol. 2009;27:858–863. doi: 10.1038/nbt.1559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Wang DR, Tang D. Hypermethylated SFRP2 gene in fecal DNA is a high potential biomarker for colorectal cancer noninvasive screening. World J Gastroenterol. 2008;14:524–531. doi: 10.3748/wjg.14.524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Zhang J, Yang S, Xie Y, Chen X, Zhao Y, He D, Li J. Detection of methylated tissue factor pathway inhibitor 2 and human long DNA in fecal samples of patients with colorectal cancer in China. Cancer Epidemiol. 2012;36:73–77. doi: 10.1016/j.canep.2011.04.006. [DOI] [PubMed] [Google Scholar]
- 23.Huang Z, Li L, Wang J. Hypermethylation of SFRP2 as a potential marker for stool-based detection of colorectal cancer and precancerous lesions. Dig Dis Sci. 2007;52:2287–2291. doi: 10.1007/s10620-007-9755-y. [DOI] [PubMed] [Google Scholar]
- 24.Leung WK, To KF, Man EP, Chan MW, Hui AJ, Ng SS, Lau JY, Sung JJ. Detection of hypermethylated DNA or cyclooxygenase-2 messenger RNA in fecal samples of patients with colorectal cancer or polyps. Am J Gastroenterol. 2007;102:1070–1076. doi: 10.1111/j.1572-0241.2007.01108.x. [DOI] [PubMed] [Google Scholar]
- 25.Itzkowitz SH, Jandorf L, Brand R, Rabeneck L, Schroy PC, Sontag S, Johnson D, Skoletsky J, Durkee K, Markowitz S, et al. Improved fecal DNA test for colorectal cancer screening. Clin Gastroenterol Hepatol. 2007;5:111–117. doi: 10.1016/j.cgh.2006.10.006. [DOI] [PubMed] [Google Scholar]
- 26.Nagasaka T, Tanaka N, Cullings HM, Sun DS, Sasamoto H, Uchida T, Koi M, Nishida N, Naomoto Y, Boland CR, et al. Analysis of fecal DNA methylation to detect gastrointestinal neoplasia. J Natl Cancer Inst. 2009;101:1244–1258. doi: 10.1093/jnci/djp265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Baek YH, Chang E, Kim YJ, Kim BK, Sohn JH, Park DI. Stool methylation-specific polymerase chain reaction assay for the detection of colorectal neoplasia in Korean patients. Dis Colon Rectum. 2009;52:1452–1459; discussion 1459-1463. doi: 10.1007/DCR.0b013e3181a79533. [DOI] [PubMed] [Google Scholar]
- 28.Azuara D, Rodriguez-Moranta F, de Oca J, Soriano-Izquierdo A, Mora J, Guardiola J, Biondo S, Blanco I, Peinado MA, Moreno V, et al. Novel methylation panel for the early detection of colorectal tumors in stool DNA. Clin Colorectal Cancer. 2010;9:168–176. doi: 10.3816/CCC.2010.n.023. [DOI] [PubMed] [Google Scholar]
- 29.Calistri D, Rengucci C, Casadei Gardini A, Frassineti GL, Scarpi E, Zoli W, Falcini F, Silvestrini R, Amadori D. Fecal DNA for noninvasive diagnosis of colorectal cancer in immunochemical fecal occult blood test-positive individuals. Cancer Epidemiol Biomarkers Prev. 2010;19:2647–2654. doi: 10.1158/1055-9965.EPI-10-0291. [DOI] [PubMed] [Google Scholar]
- 30.Olson J, Whitney DH, Durkee K, Shuber AP. DNA stabilization is critical for maximizing performance of fecal DNA-based colorectal cancer tests. Diagn Mol Pathol. 2005;14:183–191. doi: 10.1097/01.pas.0000176768.18423.7e. [DOI] [PubMed] [Google Scholar]
- 31.Anderson JC, Shaw RD. Update on colon cancer screening: recent advances and observations in colorectal cancer screening. Curr Gastroenterol Rep. 2014;16:403. doi: 10.1007/s11894-014-0403-3. [DOI] [PubMed] [Google Scholar]
- 32.Vilkin A, Rozen P, Levi Z, Waked A, Maoz E, Birkenfeld S, Niv Y. Performance characteristics and evaluation of an automated-developed and quantitative, immunochemical, fecal occult blood screening test. Am J Gastroenterol. 2005;100:2519–2525. doi: 10.1111/j.1572-0241.2005.00231.x. [DOI] [PubMed] [Google Scholar]
- 33.Brenner H, Hoffmeister M, Arndt V, Stegmaier C, Altenhofen L, Haug U. Protection from right- and left-sided colorectal neoplasms after colonoscopy: population-based study. J Natl Cancer Inst. 2010;102:89–95. doi: 10.1093/jnci/djp436. [DOI] [PubMed] [Google Scholar]
- 34.US Preventive Services Task Force. Screening for colorectal cancer: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med. 2008;149:627–637. doi: 10.7326/0003-4819-149-9-200811040-00243. [DOI] [PubMed] [Google Scholar]
- 35.Hoffman RM. In persons at average risk, stool DNA tests had higher sensitivity than FIT for detecting colorectal cancer. Ann Intern Med. 2014;161:JC10. doi: 10.7326/0003-4819-161-2-201407150-02010. [DOI] [PubMed] [Google Scholar]
- 36.Song K, Fendrick AM, Ladabaum U. Fecal DNA testing compared with conventional colorectal cancer screening methods: a decision analysis. Gastroenterology. 2004;126:1270–1279. doi: 10.1053/j.gastro.2004.02.016. [DOI] [PubMed] [Google Scholar]
- 37.Skally M, Hanly P, Sharp L. Cost effectiveness of fecal DNA screening for colorectal cancer: a systematic review and quality appraisal of the literature. Appl Health Econ Health Policy. 2013;11:181–192. doi: 10.1007/s40258-013-0010-8. [DOI] [PubMed] [Google Scholar]
- 38.Binefa G, Rodríguez-Moranta F, Teule A, Medina-Hayas M. Colorectal cancer: from prevention to personalized medicine. World J Gastroenterol. 2014;20:6786–6808. doi: 10.3748/wjg.v20.i22.6786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.A stool DNA test (Cologuard) for colorectal cancer screening. JAMA. 2014;312:2566. doi: 10.1001/jama.2014.15746. [DOI] [PubMed] [Google Scholar]
- 40.Ahlquist DA. Multi-target stool DNA test: a new high bar for noninvasive screening. Dig Dis Sci. 2015;60:623–633. doi: 10.1007/s10620-014-3451-5. [DOI] [PubMed] [Google Scholar]
- 41.Huddy JR, Ni MZ, Markar SR, Hanna GB. Point-of-care testing in the diagnosis of gastrointestinal cancers: current technology and future directions. World J Gastroenterol. 2015;21:4111–4120. doi: 10.3748/wjg.v21.i14.4111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Hutchinson L. Screening: Where does stool DNA testing FIT in the CRC screening menu? Nat Rev Clin Oncol. 2014;11:239. doi: 10.1038/nrclinonc.2014.60. [DOI] [PubMed] [Google Scholar]
- 43.Kisiel JB, Yab TC, Nazer Hussain FT, Taylor WR, Garrity-Park MM, Sandborn WJ, Loftus EV, Wolff BG, Smyrk TC, Itzkowitz SH, et al. Stool DNA testing for the detection of colorectal neoplasia in patients with inflammatory bowel disease. Aliment Pharmacol Ther. 2013;37:546–554. doi: 10.1111/apt.12218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kisiel JB, Yab TC, Taylor WR, Chari ST, Petersen GM, Mahoney DW, Ahlquist DA. Stool DNA testing for the detection of pancreatic cancer: assessment of methylation marker candidates. Cancer. 2012;118:2623–2631. doi: 10.1002/cncr.26558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Strauss BB, Yab TC, OConnor HM, Taylor WR, Mahoney DW, Simonson JA, Christensen JD, Chari ST, Ahlquist DA. Fecal recovery of ingested cellular DNA: implications for noninvasive detection of upper gastrointestinal neoplasms. Gastroenterology. 2014;146:S–323-S-324. doi: 10.1007/s10620-015-3845-z. [DOI] [PubMed] [Google Scholar]
- 46.Kisiel JB, Taylor WR, Yab TC, Mahoney DW, Sun Z, Middha S, Zou H, Smyrk TC, Romero Y, Boardman L, et al. Novel methylated DNA markers predict site of gastrointestinal cancer. Gastroenterology. 2013;144:S–84. [Google Scholar]
- 47.Gralnek IM, Siersema PD, Halpern Z, Segol O, Melhem A, Suissa A, Santo E, Sloyer A, Fenster J, Moons LM, et al. Standard forward-viewing colonoscopy versus full-spectrum endoscopy: an international, multicentre, randomised, tandem colonoscopy trial. Lancet Oncol. 2014;15:353–360. doi: 10.1016/S1470-2045(14)70020-8. [DOI] [PMC free article] [PubMed] [Google Scholar]