Prospective study to validate the clinical utility of DNA diagnosis of peritoneal fluid cytology test in gastric cancer

Hiroki Harada, Takafumi Soeno, Nobuyuki Nishizawa, Marie Washio, Mikiko Sakuraya, Hideki Ushiku, Masahiro Niihara, Kei Hosoda, Yusuke Kumamoto, Takeshi Naitoh, Takafumi Sangai, Naoki Hiki, Keishi Yamashita, Hiroki Harada, Takafumi Soeno, Nobuyuki Nishizawa, Marie Washio, Mikiko Sakuraya, Hideki Ushiku, Masahiro Niihara, Kei Hosoda, Yusuke Kumamoto, Takeshi Naitoh, Takafumi Sangai, Naoki Hiki, Keishi Yamashita

Abstract

The clinical efficacy of DNA cytology test (CY) in gastric cancer (GC) has been retrospectively proposed using cancer-specific methylation of cysteine dioxygenase type 1 (CDO1). We confirmed the clinical utility of DNA CY in a prospective cohort. Four hundred GC samples were prospectively collected for washing cytology (UMIN000026191), and detection of the DNA methylation of CDO1 was assessed by quantitative methylation-specific PCR in the sediments. Endpoint was defined as the match rate between conventional CY1 and DNA CY1 (diagnostic sensitivity), and the DNA CY0 rate (diagnostic specificity) in pStage IA. DNA CY1 was detected in 45 cases (12.5%), while CY1 was seen in 31 cases (8.6%) of 361 chemotherapy-naïve samples, where the sensitivity and specificity of the DNA CY in the peritoneal solutions were 74.2% and 96.5%, respectively. The DNA CY was positive for 3.5/0/4.9/11.4/58.8% in pStage IA/IB/II/III/IV, respectively (P < .01). In the multivariate analysis, DNA CY1 was independently correlated with pathological tumor depth (pT) (P = .0012), female gender (P = .0099), CY1 (P = .0135), P1 (P = .019), and carcinoembryonic antigen (CEA) (P = .036). The combination of DNA CY1 and P factor nearly all covered the potential peritoneal dissemination (P1 and/or CY1 and/or DNA CY1) (58/61:95.1%). DNA CY1 had a significantly poorer prognosis than DNA CY0 in GC patients (P < .0001). DNA CY1 detected by CDO1 promoter DNA methylation has a great value to detect minimal residual disease of the peritoneum in GC clinics, representing poor prognosis as a novel single DNA marker.

Keywords: Cysteine dioxygenase type 1; gastric cancer; methylation-specific PCR; peritoneal dissemination; washing cytology test.

Conflict of interest statement

The authors have no competing interest to declare.

© 2021 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association.

Figures

FIGURE 1
FIGURE 1
Bar graphs representing diagnostic sensitivity of the conventional CY1 (red bars) and the DNA CY1 (blue bars) according to pathological factor of gastric cancer (GC). A, Pathological T factor. B, Pathological N factor. C, Pathological stage
FIGURE 2
FIGURE 2
Quantitative methylation‐specific PCR (Q‐MSP) by using CDO1 DNA methylation. A, TaqMeth values of CDO1 in the peritoneal fluid samples of the 361 gastric cancers (GCs) (upper panel). The quantified outcomes of the DNA CY1 are magnified (lower panel). In the lower panel, red bar graphs represent conventional CY1, while blue graphs indicate conventional CY0. B, Q‐MSP curves of the representative cases. CDO1 TaqMeth values (Vs) were definitely represented as 39.8, 2.7, 1.2, and 0.3 in sample numbers 202, 156, 123, and 74, respectively (upper panel), while the minimum CDO1 TaqMeth V 0.003 among DNA CY1 cases was judged as positive in sample number 145
FIGURE 3
FIGURE 3
Prognosis analysis in DNA CY and CY. A, Overall survival of 346 gastric cancer (GC) patients associated with DNA CY. B, Overall survival of 165 advanced GC patients associated with DNA CY. C, Overall survival of 346 GC patients stratified according to the results of DNA CY and CY. D, Peritoneal dissemination–free survival of 346 GC patients stratified according to the results of DNA CY and CY. E, Overall survival of 51 GC patients with DNA CY and/or CY positive. F, Peritoneal dissemination–free survival of 51 GC patients with DNA CY and/or CY positive
FIGURE 4
FIGURE 4
Detailed assessment for the conventional CY1 cases (n = 27). In quantitative methylation‐specific PCR (Q‐MSP), CDO1 TaqMeth values (Vs) of the preoperative tumor biopsy samples (blue bars) were compared with those of the intraperitoneal fluid samples (red bars). Left scale represents CDO1 TaqMeth V in Q‐MSP. The red‐circled samples were judged to be negative in Q‐MSP because CDO1 TaqMeth V was 0

References

    1. Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394‐424.
    1. Yoshida K, Kodera Y, Kochi M, et al. Addition of docetaxel to oral fluoropyrimidine improves efficacy in patients with stage III gastric cancer: interim analysis of JACCRO GC‐07, a randomized controlled trial. J Clin Oncol. 2019;37:1296‐1304.
    1. Yamada Y, Higuchi K, Nishikawa K, et al. Phase III study comparing oxaliplatin plus S‐1 with cisplatin plus S‐1 in chemotherapy‐naive patients with advanced gastric cancer. Ann Oncol. 2015;26:141‐148.
    1. Wilke H, Muro K, Cutsem EV, et al. RAINBOW Study Group, Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro‐oesophageal junction adenocarcinoma (RAINBOW): a double‐blind, randomised phase 3 trial. Lancet Oncol. 2014;15:1224‐1235.
    1. Kang YK, Boku N, Satoh T, et al. Nivolumab in patients with advanced gastric or gastro‐oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO‐4538‐12, ATTRACTION‐2): a randomised, double‐blind, placebo‐controlled, phase 3 trial. Lancet. 2017;390:2461‐2471.
    1. Hosoda K, Watanabe M, Yamashita K. Re‐emerging role of macroscopic appearance in treatment strategy for gastric cancer. Ann Gastroenterol Surg. 2019;3:122‐129.
    1. Japanese Gastric Cancer Association . Japanese classification of gastric carcinoma ‐ 2nd English Edition. Gastric Cancer. 1998;1:10‐24.
    1. Japanese Gastric Cancer Association . Japanese classification of gastric carcinoma. 3rd English edition. Gastric Cancer. 2011;14:101‐112.
    1. Sobin LH, Gospodarowicz MK, Wittekind C. TNM Classification of Malignant Tumours, Seven Edition. Oxford, UK: Wiley‐Blackwell; 2009.
    1. Kodera Y, Nakanishi H, Ito S, et al. Quantitative detection of disseminated free cancer cells in peritoneal washes with real‐time reverse transcriptase‐polymerase chain reaction: a sensitive predictor of outcome for patients with gastric carcinoma. Ann Surg. 2002;235:499‐506.
    1. Yamashita K, Hosoda K, Nishizawa N, et al. Epigenetic biomarkers of promoter DNA methylation in the new era of cancer treatment. Cancer Sci. 2018;109:3695‐3706.
    1. Harada H, Hosoda K, Moriya H, et al. Cancer‐specific promoter DNA methylation of Cysteine dioxygenase type 1 (CDO1) gene as an important prognostic biomarker of gastric cancer. PLoS One. 2019;14:e0214872.
    1. Ushiku H, Yamashita K, Ema A, et al. DNA diagnosis of peritoneal fluid cytology test by CDO1 promoter DNA hypermethylation in gastric cancer. Gastric Cancer. 2017;20:784‐792.
    1. Andresen K, Boberg KM, Vedeld HM, et al. Four DNA methylation biomarkers in biliary brush samples accurately identify the presence of cholangiocarcinoma. Hepatology. 2015;61:1651‐1659.
    1. Nishizawa N, Harada H, Kumamoto Y, et al. Promoter DNA hypermethylation of the cysteine dioxygenase 1 (CDO1) gene and excellent diagnostic performance in pancreatic ductal adenocarcinoma. Cancer Sci. 2019;110:2846‐2855.
    1. Association JGC . Japanese Classification of Gastric Carcinoma [The, 15th Edition]. Oxford, UK: Kanehara‐Shuppan: 2017.
    1. Hiraki M, Kitajima Y, Sato S, et al. Aberrant gene methylation in the peritoneal fluid is a risk factor predicting peritoneal recurrence in gastric cancer. World J Gastroenterol. 2010;16:330‐338.
    1. Hiraki M, Kitajima Y, Koga Y, et al. Aberrant gene methylation is a biomarker for the detection of cancer cells in peritoneal wash samples from advanced gastric cancer patients. Ann Surg Oncol. 2011;18:3013‐3019.
    1. Kamiyama H, Noda H, Takata O, et al. Promoter hypermethylation of tumor‐related genes in peritoneal lavage and the prognosis of patients with colorectal cancer. J Surg Oncol. 2009;100:69‐74.
    1. Brait M, Ling S, Nagpal JK, et al. Cysteine dioxygenase 1 is a tumor suppressor gene silenced by promoter methylation in multiple human cancers. PLoS One. 2012;7:e44951.
    1. Nakanishi K, Kanda M, Umeda S, et al. The levels of SYT13 and CEA mRNAs in peritoneal lavages predict the peritoneal recurrence of gastric cancer. Gastric Cancer. 2019;22:1143‐1152.
    1. Yamashita K, Waraya M, Kim MS, et al. Detection of methylated CDO1 in plasma of colorectal cancer; a PCR study. PLoS One. 2014;9:e113546.
    1. Yamashita K, Hosoda K, Moriya H, et al. Long‐term prognostic outcome of cT1 gastric cancer patients who underwent laparoscopic gastrectomy after 5‐year follow‐up. Langenbecks Arch Surg. 2016;401:333‐339.
    1. Yamamoto M, Taguchi K, Baba H, et al. Peritoneal dissemination of early gastric cancer: report of a case. Surg Today. 2006;36:835‐858.
    1. Benevolo M, Mottolese M, Cosimelli M, et al. Diagnostic and prognostic value of peritoneal immunocytology in gastric cancer. J Clin Oncol. 1998;16:3406‐3411.
    1. Vogel P, Rüschoff J, Kümmel S, et al. Immunocytology improves prognostic impact of peritoneal tumour cell detection compared to conventional cytology in gastric cancer. Eur J Surg Oncol. 1999;25:515‐519.
    1. Kitayama J, Emoto S, Yamaguchi H, et al. Flow cytometric quantification of intraperitoneal free tumor cells is a useful biomarker in gastric cancer patients with peritoneal metastasis. Ann Surg Oncol. 2015;22:2336‐2342.
    1. Cancer Genome Atlas Research Network . Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202‐209.

Source: PubMed

3
구독하다