Methylated SEPTIN9 plasma test for colorectal cancer detection may be applicable to Lynch syndrome

Megan P Hitchins, Ingrid P Vogelaar, Kevin Brennan, Sigurdis Haraldsdottir, Nianmin Zhou, Brock Martin, Rocio Alvarez, Xiaopu Yuan, Sungjin Kim, Maha Guindi, Andrew E Hendifar, Matthew F Kalady, Jennifer DeVecchio, James M Church, Albert de la Chapelle, Heather Hampel, Rachel Pearlman, Maria Christensen, Carrie Snyder, Stephen J Lanspa, Robert W Haile, Henry T Lynch, Megan P Hitchins, Ingrid P Vogelaar, Kevin Brennan, Sigurdis Haraldsdottir, Nianmin Zhou, Brock Martin, Rocio Alvarez, Xiaopu Yuan, Sungjin Kim, Maha Guindi, Andrew E Hendifar, Matthew F Kalady, Jennifer DeVecchio, James M Church, Albert de la Chapelle, Heather Hampel, Rachel Pearlman, Maria Christensen, Carrie Snyder, Stephen J Lanspa, Robert W Haile, Henry T Lynch

Abstract

Objective: The plasma-based methylated SEPTIN9 (mSEPT9) is a colorectal cancer (CRC) screening test for adults aged 50-75 years who are at average risk for CRC and have refused colonoscopy or faecal-based screening tests. The applicability of mSEPT9 for high-risk persons with Lynch syndrome (LS), the most common hereditary CRC condition, has not been assessed. This study sought preliminary evidence for the utility of mSEPT9 for CRC detection in LS.

Design: Firstly, SEPT9 methylation was measured in LS-associated CRC, advanced adenoma, and subject-matched normal colorectal mucosa tissues by pyrosequencing. Secondly, to detect mSEPT9 as circulating tumor DNA, the plasma-based mSEPT9 test was retrospectively evaluated in LS subjects using the Epi proColon 2.0 CE assay adapted for 1mL plasma using the "1/1 algorithm". LS case groups included 20 peri-surgical cases with acolonoscopy-based diagnosis of CRC (stages I-IV), 13 post-surgical metastatic CRC, and 17 pre-diagnosis cases. The control group comprised 31 cancer-free LS subjects.

Results: Differential hypermethylation was found in 97.3% (36/37) of primary CRC and 90.0% (18/20) of advanced adenomas, showing LS-associated neoplasia frequently produce the mSEPT9 biomarker. Sensitivity of plasma mSEPT9 to detect CRC was 70.0% (95% CI, 48%-88%)in cases with a colonoscopy-based CRC diagnosis and 92.3% (95% CI, 64%-100%) inpost-surgical metastatic cases. In pre-diagnosis cases, plasma mSEPT9 was detected within two months prior to colonoscopy-based CRC diagnosis in 3/5 cases. Specificity in controls was 100% (95% CI 89%-100%).

Conclusion: These preliminary findings suggest mSEPT9 may demonstrate similar diagnostic performance characteristics in LS as in the average-risk population, warranting a well-powered prospective case-control study.

Keywords: SEPTIN9; circulating tumor dna; colorectal cancer; lynch syndrome; plasma.

Conflict of interest statement

Competing interests: None declared.

Figures

Figure 1
Figure 1
Levels and prevalence of SEPT9 methylation in colon and rectal adenocarcinoma (COADREAD) compared with adjacent normal tissue in Infinium Human Methylation 450K Array data from The Cancer Genome Atlas (TCGA). (A) Dot plot of differential methylation levels (tumour-normal; y-axis) by genomic location (x-axis) according to the GRCh37/hg19 human genome assembly at each of the SEPT9 probes contained within the 450K array data from the TCGA COADREAD data set. The closest probe to the plasma-based Epi proColon 2.0 CE assay, cg2027558, located at Chr17: 75369484 is indicated. (B) Manhattan plot of the −log10 p values (Wilcoxon rank-sum test; y-axis) for a difference in methylation between tumour and normal tissues by genomic location (x-axis). Probe cg2027558, indicated, showed the most significant difference in tumour versus normal tissue. (C) Spaghetti plot of normalised levels of methylation (β values) at probe cg2027558 in normal tissue and COADREAD tumour tissue. Subject-matched sample pairs are linked by lines. Median, IQRs and 95% CI are indicated. Green=tumours showing hypermethylation, red=tumours with methylation levels in the normal range. (D) Histogram of the frequency of SEPT9 hypermethylation at cg2027558 in colorectal cancer (CRC) from COADREAD stratified by tumour stage.
Figure 2
Figure 2
Measurement of SEPTIN9 methylation levels by quantitative CpG pyrosequencing in archival Lynch syndrome (LS)-associated colorectal cancer (CRC), advanced adenoma, and paired normal colorectal mucosa (NCM) tissues. (A) Assay design. Sequence coordinates GRCh37/hg19 assembly Chr17: 75369414–75369587 are shown as bisulfite converted. YG indicates CpG sites where the cytosine may be methylated (CG) or unmethylated (TG). PCR primer-binding sites are italicised and indicated by black arrows. Pyrosequencing primer-binding site is italicised and indicated by a red arrow. The sequence analysed (red text) contains five CpG sites, of which the first corresponds to Infinium probe cg20275528, located 92 bp upstream of the fluorescent probe-binding sequence within the plasma-based Epi proColon 2.0 CE assay (blue text). (B) Simulated pyrosequencing assay. Grey shading shows the five CpG sites interrogated. Yellow shading shows the single cytosine control for bisulfite conversion efficiency. (C) Analytical sensitivity and linearity of the quantitative CpG pyrosequencing assay to measure methylation levels at SEPTIN9. The titration curve shows the observed (±1 SD) versus expected methylation values for the hypermethylated RKO CRC cell line DNA (expected value 100% methylated) diluted into the peripheral blood DNA from a healthy control (0% expected methylation value) in specific proportions. (D) Illustrative pyrograms obtained from formalin-fixed paraffin-embedded (FFPE) samples of primary CRC, a liver metastasis, and paired NCM from a Lynch syndrome patient with an MSH2 mutation.
Figure 3
Figure 3
Frequent hypermethylation of SEPT9 in Lynch syndrome (LS)-associated colorectal cancer (CRC) and advanced adenomas. (A) Spaghetti plot showing actual values of SEPT9 methylation measured by pyrosequencing at five CpG sites, including cg20275528, in CRC, advanced adenomas, and normal colorectal mucosa (NCM) from patients with LS (each black dot represents a single tissue sample and subject-matched samples are linked by lines), overlaid with box and whisker plot indicating the median value (horizontal black line) and IQRs (boxes). Dotted horizontal lines indicate the threshold values of 23% and 18% for methylation-positive test results to discriminate CRC and adenoma samples from NCM samples, respectively. Accompanying clinicopathological and SEPTIN9 methylation data are provided in online supplementary tables 1 and 2. (B) Receiver operating characteristic (ROC) curve for SEPT9 methylation values in CRC versus paired NCM (left), and advanced adenomas versus paired NCM (right). The diagonal lines represent the non-informative prediction model with an area under the ROC curve (AUC) of 0.5. ROC analyses were performed for the subset of neoplasms with a subject-matched NCM sample, including 29 CRC samples with 28 unique paired NCM samples, and 13 adenoma samples with 10 unique paired NCM samples.

References

    1. Administration USFaD. Premarket approval (PMA) for EPI proColon, 2016.
    1. deVos T, Tetzner R, Model F, et al. . Circulating methylated SEPT9 DNA in plasma is a biomarker for colorectal cancer. Clin Chem 2009;55:1337–46. 10.1373/clinchem.2008.115808
    1. Lofton-Day C, Model F, Devos T, et al. . DNA methylation biomarkers for blood-based colorectal cancer screening. Clin Chem 2008;54:414–23. 10.1373/clinchem.2007.095992
    1. Wasserkort R, Kalmar A, Valcz G, et al. . Aberrant septin 9 DNA methylation in colorectal cancer is restricted to a single CpG island. BMC Cancer 2013;13 10.1186/1471-2407-13-398
    1. Song L, Jia J, Peng X, et al. . The performance of the SEPT9 gene methylation assay and a comparison with other CRC screening tests: a meta-analysis. Sci Rep 2017;7 10.1038/s41598-017-03321-8
    1. Potter NT, Hurban P, White MN, et al. . Validation of a real-time PCR-based qualitative assay for the detection of methylated SEPT9 DNA in human plasma. Clin Chem 2014;60:1183–91. 10.1373/clinchem.2013.221044
    1. Johnson DA, Barclay RL, Mergener K, et al. . Plasma Septin9 versus fecal immunochemical testing for colorectal cancer screening: a prospective multicenter study. PLoS One 2014;9:e98238 10.1371/journal.pone.0098238
    1. Hampel H, Frankel WL, Martin E, et al. . Screening for the Lynch syndrome (hereditary nonpolyposis colorectal cancer). N Engl J Med 2005;352:1851–60. 10.1056/NEJMoa043146
    1. Win AK, Jenkins MA, Dowty JG, et al. . Prevalence and penetrance of major genes and polygenes for colorectal cancer. Cancer Epidemiol Biomarkers Prev 2017;26:404–12. 10.1158/1055-9965.EPI-16-0693
    1. Haraldsdottir S, Rafnar T, Frankel WL, et al. . Comprehensive population-wide analysis of Lynch syndrome in Iceland reveals founder mutations in MSH6 and pms2. Nat Commun 2017;8 10.1038/ncomms14755
    1. Lynch HT, Snyder CL, Shaw TG, et al. . Milestones of Lynch syndrome: 1895-2015. Nat Rev Cancer 2015;15:181–94. 10.1038/nrc3878
    1. Barrow E, Hill J, Evans DG. Cancer risk in Lynch syndrome. Fam Cancer 2013;12:229–40. 10.1007/s10689-013-9615-1
    1. Vasen HFA, Tomlinson I, Castells A. Clinical management of hereditary colorectal cancer syndromes. Nat Rev Gastroenterol Hepatol 2015;12:88–97. 10.1038/nrgastro.2014.229
    1. Baglietto L, Lindor NM, Dowty JG, et al. . Risks of Lynch syndrome cancers for Msh6 mutation carriers. J Natl Cancer Inst 2010;102:193–201. 10.1093/jnci/djp473
    1. Network NCC Genetic/Familial high-risk assessment: colorectal. NCCN Clinical Practice Guidelines in Oncology, 2018.
    1. Järvinen HJ, Aarnio M, Mustonen H, et al. . Controlled 15-year trial on screening for colorectal cancer in families with hereditary nonpolyposis colorectal cancer. Gastroenterology 2000;118:829–34. 10.1016/S0016-5085(00)70168-5
    1. de Jong AE, Hendriks YMC, Kleibeuker JH, et al. . Decrease in mortality in Lynch syndrome families because of surveillance. Gastroenterology 2006;130:665–71. 10.1053/j.gastro.2005.11.032
    1. Newton K, Green K, Lalloo F, et al. . Colonoscopy screening compliance and outcomes in patients with Lynch syndrome. Colorectal Dis 2015;17:38–46. 10.1111/codi.12778
    1. Blue Ribbon Panel NCI Cancer Moonshot blue ribbon panel report, 2016. Available:
    1. Stoffel EM, Mercado RC, Kohlmann W, et al. . Prevalence and predictors of appropriate colorectal cancer surveillance in Lynch syndrome. Am J Gastroenterol 2010;105:1851–60. 10.1038/ajg.2010.120
    1. Aktan-Collan K, Kääriäinen H, Järvinen H, et al. . Psychosocial consequences of predictive genetic testing for Lynch syndrome and associations to surveillance behaviour in a 7-year follow-up study. Fam Cancer 2013;12:639–46. 10.1007/s10689-013-9628-9
    1. de Vos tot Nederveen Cappel WH, Järvinen HJ, Lynch PM, Engel C, et al. . Colorectal surveillance in Lynch syndrome families. Fam Cancer 2013;12:261–5. 10.1007/s10689-013-9631-1
    1. Vasen HF, Nagengast FM, Khan PM. Interval cancers in hereditary non-polyposis colorectal cancer (Lynch syndrome). Lancet 1995;345:1183–4. 10.1016/S0140-6736(95)91016-6
    1. Lanspa SJ, Jenkins JX, Cavalieri RJ, et al. . Surveillance in Lynch syndrome: how aggressive? Am J Gastroenterol 1994;89:1978–80.
    1. Møller P, Seppälä T, Bernstein I, et al. . Cancer incidence and survival in Lynch syndrome patients receiving colonoscopic and gynaecological surveillance: first report from the prospective Lynch syndrome database. Gut 2017;66 10.1136/gutjnl-2015-309675
    1. Stuckless S, Green JS, Morgenstern M, et al. . Impact of colonoscopic screening in male and female Lynch syndrome carriers with an MSH2 mutation. Clin Genet 2012;82:439–45. 10.1111/j.1399-0004.2011.01802.x
    1. Jass JR, Smyrk TC, Stewart SM, et al. . Pathology of hereditary non-polyposis colorectal cancer. Anticancer Res 1994;14:1631–4.
    1. Lindgren G, Liljegren A, Jaramillo E, et al. . Adenoma prevalence and cancer risk in familial non-polyposis colorectal cancer. Gut 2002;50:228–34. 10.1136/gut.50.2.228
    1. Rijcken FEM, Hollema H, Kleibeuker JH. Proximal adenomas in hereditary non-polyposis colorectal cancer are prone to rapid malignant transformation. Gut 2002;50:382–6. 10.1136/gut.50.3.382
    1. Rondagh EJA, Gulikers S, Gómez-García EB, et al. . Nonpolypoid colorectal neoplasms: a challenge in endoscopic surveillance of patients with Lynch syndrome. Endoscopy 2013;45:257–64. 10.1055/s-0032-1326195
    1. van Rijn JC, Reitsma JB, Stoker J, et al. . Polyp miss rate determined by tandem colonoscopy: a systematic review. Am J Gastroenterol 2006;101:343–50. 10.1111/j.1572-0241.2006.00390.x
    1. Stoffel EM, Turgeon DK, Stockwell DH, et al. . Missed adenomas during colonoscopic surveillance in individuals with Lynch syndrome (hereditary nonpolyposis colorectal cancer). Cancer Prev Res 2008;1:470–5. 10.1158/1940-6207.CAPR-08-0098
    1. Niv Y, Moeslein G, Vasen HFA, et al. . Quality of colonoscopy in Lynch syndrome. Endosc Int Open 2014;2:E252–E255. 10.1055/s-0034-1377920
    1. Edelstein DL, Axilbund J, Baxter M, et al. . Rapid development of colorectal neoplasia in patients with Lynch syndrome. Clin Gastroenterol Hepatol 2011;9:340–3. 10.1016/j.cgh.2010.10.033
    1. Samur MK. RTCGAToolbox: a new tool for exporting TCGA Firehose data. PLoS One 2014;9:e106397 10.1371/journal.pone.0106397
    1. Troyanskaya O, Cantor M, Sherlock G, et al. . Missing value estimation methods for DNA microarrays. Bioinformatics 2001;17:520–5. 10.1093/bioinformatics/17.6.520
    1. Johnson WE, Li C, Rabinovic A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 2007;8:118–27. 10.1093/biostatistics/kxj037
    1. Gevaert O. MethylMix: an R package for identifying DNA methylation-driven genes. Bioinformatics 2015;31:1839–41. 10.1093/bioinformatics/btv020
    1. Youden WJ. Index for rating diagnostic tests. Cancer 1950;3:32–5. 10.1002/1097-0142(1950)3:1<32::AID-CNCR2820030106>;2-3
    1. Collett D, Data MB. Modelling binary data. Boca Raton Florida: Hall/CRC Ca, 1999.
    1. Cancer Genome Atlas Network Comprehensive molecular characterization of human colon and rectal cancer. Nature 2012;487:330–7. 10.1038/nature11252
    1. Song L, Li Y, Jia J, et al. . Algorithm optimization in methylation detection with multiple RT-qPCR. PLoS One 2016;11:e0163333 10.1371/journal.pone.0163333
    1. Song L, Jia J, Yu H, et al. . The performance of the mSEPT9 assay is influenced by algorithm, cancer stage and age, but not sex and cancer location. J Cancer Res Clin Oncol 2017;143:1093–101. 10.1007/s00432-017-2363-0
    1. Fu B, Yan P, Zhang S, et al. . Cell-free circulating methylated SEPT9 for noninvasive diagnosis and monitoring of colorectal cancer. Dis Markers 2018;2018:1–11. 10.1155/2018/6437104
    1. Lee HS, Hwang SM, Kim TS, et al. . Circulating methylated septin 9 nucleic acid in the plasma of patients with gastrointestinal cancer in the stomach and colon. Transl Oncol 2013;6:290–IN4. 10.1593/tlo.13118
    1. Møller P, Seppälä TT, Bernstein I, et al. . Cancer risk and survival in path_MMR carriers by gene and gender up to 75 years of age: a report from the Prospective Lynch Syndrome Database. Gut 2018;67:1306–16. 10.1136/gutjnl-2017-314057

Source: PubMed

3
Iratkozz fel