A prospective study on the changes and clinical significance of pre-operative and post-operative circulating tumor cells in resectable gastric cancer

Qiyue Zhang, Fei Shan, Ziyu Li, Jing Gao, Yilin Li, Lin Shen, Jiafu Ji, Ming Lu, Qiyue Zhang, Fei Shan, Ziyu Li, Jing Gao, Yilin Li, Lin Shen, Jiafu Ji, Ming Lu

Abstract

Background: Circulating tumor cells (CTCs) have been suggested as potential prognostic indicators for multiple tumors, including gastric cancer; however, pre- and post-operative CTC changes in resectable gastric cancer and possible correlations to post-operative recurrence have not been evaluated.

Methods: Subjects (n = 93) with resectable gastric cancer were prospectively reviewed from July 2013 to December 2014 at Peking University Cancer Hospital. The proportion of CTCs were evaluated before (n = 93) and after (n = 63) radical operation using a standardized CellSearch system.

Results: CTCs ≥ 1 were measured in the pre-operative blood of 31 (33.3%) patients and in the post-operative blood of 21 patients (33.3%). Patients with relatively poor clinicopathological features had more pre- and post-operative CTCs. The 3-year disease-free survival (DFS) rate for patients with CTCs ≥ 5/7.5 ml was significantly lower than for patients with CTCs < 5/7.5 ml (40.0% vs 66.4%, p < 0.001 for pre-surgery; 25.0% vs 62.2%, p < 0.001 for post-surgery). Patients with CTCs ≥ 5/7.5 ml in post-operative blood had significantly shorter mean DFS (1.28 vs 31.6 months; p = 0.002) and overall survival (OS; 10.0 vs 34.9 months; p = 0.001) than other patients. Among the 10 patients with hematogenous recurrence, 3 had post-operative CTCs ≥ 2/7.5 ml and had early recurrence (DFS 1.1, 1.1, 1.4 months). Moreover, DFS for the seven patients was 20.2, 11.9, 20.0, 6.0, 15.5, 25.9, 30.0 months, respectively. DFS for the three patients with increased CTCs after surgery was shorter than for patients with mildly increased, stable, or decreased CTCs.

Conclusions: Pre- and post-operative CTCs are promising prognostic markers for resectable gastric cancer. Our study further suggests that increased post-operative CTCs may be correlated with hematogenous recurrence. Trial registration (ClinicalTrials.gov Identifier: NCT01848015). Registered 7 May 2013. https://ichgcp.net/clinical-trials-registry/NCT01848015.

Keywords: CellSearch; Circulating tumor cells; Hematogenous metastasis; Recurrence; Resectable gastric cancer.

Figures

Fig. 1
Fig. 1
CTCs were significantly (p a), TNM classification (b), depth of penetration (c) and lymph-vascular invasion (d), and were obviously, though not significantly, correlated with age (e), differentiation (f), lymph node metastasis (g), and CEA level (h) in preoperative blood, *p < 0.05
Fig. 2
Fig. 2
CTCs were significantly (p a) and were obviously, though not significantly, correlated with TNM classification (b) in postoperative blood, *p < 0.05
Fig. 3
Fig. 3
3-year DFS was lower in patients who had more CTCs (a), the filled black columns represent the 3-year DFS of patients who had CTCs no more than the cut-off value, and the empty white columns represent 3-year DFS of patients who had CTC more than the cutoff value. Patients who have ≥ 5 CTCs/7.5 ml blood pre-operatively had shorter DFS (b, d) and OS (c, e)
Fig. 4
Fig. 4
Among hematogenous metastases subjects, patients who had post-operative CTCs ≥ 2/7.5 ml (a) and patients who had markedly increased CTCs after surgery (b) had markedly shorter DFS

References

    1. Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin. 2014;64:9–29. doi: 10.3322/caac.21208.
    1. Sasako M, Sakuramoto S, Katai H, et al. Five-year outcomes of a randomized phase III trial comparing adjuvant chemotherapy with S-1 versus surgery alone in stage II or III gastric cancer. J Clin Oncol. 2011;29:4387–4393. doi: 10.1200/JCO.2011.36.5908.
    1. Park SH, Sohn TS, Lee J, et al. Phase III trial to compare adjuvant chemotherapy with capecitabine and cisplatin versus concurrent chemoradiotherapy in gastric cancer: final report of the adjuvant chemoradiotherapy in stomach tumors trial, including survival and subset analyses. J Clin Oncol. 2015;33:3130–3136. doi: 10.1200/JCO.2014.58.3930.
    1. Sun Z, Wang ZN, Zhu Z, et al. Evaluation of the seventh edition of American Joint Committee on Cancer TNM staging system for gastric cancer: results from a Chinese monoinstitutional study. Ann Surg Oncol. 2012;19:1918–1927. doi: 10.1245/s10434-011-2206-1.
    1. Li Z, Zou X, Xie L, et al. Personalizing risk stratification by addition of PAK1 expression to TNM staging: improving the accuracy of clinical decision for gastroesophageal junction adenocarcinoma. Int J Cancer. 2015;136:1636–1645. doi: 10.1002/ijc.29167.
    1. Duffy MJ, Lamerz R, Haglund C, et al. Tumor markers in colorectal cancer, gastric cancer and gastrointestinal stromal cancers: european group on tumor markers 2014 guidelines update. Int J Cancer. 2014;134:2513–2522. doi: 10.1002/ijc.28384.
    1. Cristescu R, Lee J, Nebozhyn M, et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 2015;21:449–456. doi: 10.1038/nm.3850.
    1. Yoo CH, Noh SH, Shin DW, Choi SH, Min JS. Recurrence following curative resection for gastric carcinoma. Br J Surg. 2000;87:236–242. doi: 10.1046/j.1365-2168.2000.01360.x.
    1. Reymond N, d’Água BB, Ridley AJ. Crossing the endothelial barrier during metastasis. Nat Rev Cancer. 2013;13:858–870. doi: 10.1038/nrc3628.
    1. Aceto N, Bardia A, Miyamoto DT, et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell. 2014;158:1110–1122. doi: 10.1016/j.cell.2014.07.013.
    1. Cristofanilli M, Budd GT, Ellis MJ, et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med. 2004;351:781–791. doi: 10.1056/NEJMoa040766.
    1. Cohen SJ, Punt CJ, Iannotti N, et al. Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J Clin Oncol. 2008;26:3213–3221. doi: 10.1200/JCO.2007.15.8923.
    1. de Bono JS, Scher HI, Montgomery RB, et al. Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin Cancer Res. 2008;14:6302–6309. doi: 10.1158/1078-0432.CCR-08-0872.
    1. Krebs MG, Sloane R, Priest L, et al. Evaluation and prognostic significance of circulating tumor cells in patients with non-small-cell lung cancer. J Clin Oncol. 2011;29:1556–1563. doi: 10.1200/JCO.2010.28.7045.
    1. Yu M, Bardia A, Aceto N, et al. Cancer therapy. Ex vivo culture of circulating breast tumor cells for individualized testing of drug susceptibility. Science. 2014;345:216–220. doi: 10.1126/science.1253533.
    1. Liu MC, Shields PG, Warren RD, et al. Circulating tumor cells: a useful predictor of treatment efficacy in metastatic breast cancer. J Clin Oncol. 2009;27:5153–5159. doi: 10.1200/JCO.2008.20.6664.
    1. Li Y, Zhang X, Ge S, et al. Clinical significance of phenotyping and karyotyping of circulating tumor cells in patients with advanced gastric cancer. Oncotarget. 2014;5:6594–6602.
    1. Sotelo MJ, Sastre J, Maestro ML, et al. Role of circulating tumor cells as prognostic marker in resected stage III colorectal cancer. Ann Oncol. 2015;26:535–541. doi: 10.1093/annonc/mdu568.
    1. Iinuma H, Watanabe T, Mimori K, et al. Clinical significance of circulating tumor cells, including cancer stem-like cells, in peripheral blood for recurrence and prognosis in patients with Dukes’ stage B and C colorectal cancer. J Clin Oncol. 2011;29:1547–1555. doi: 10.1200/JCO.2010.30.5151.
    1. Lucci A, Hall CS, Lodhi AK, et al. Circulating tumour cells in non-metastatic breast cancer: a prospective study. Lancet Oncol. 2012;13:688–695. doi: 10.1016/S1470-2045(12)70209-7.
    1. Gazzaniga P, de Berardinis E, Raimondi C, et al. Circulating tumor cells detection has independent prognostic impact in high-risk non-muscle invasive bladder cancer. Int J Cancer. 2014;135:1978–1982. doi: 10.1002/ijc.28830.
    1. Pal SK, He M, Wilson T, et al. Detection and phenotyping of circulating tumor cells in high-risk localized prostate cancer. Clin Genitourin Cancer. 2015;13:130–136. doi: 10.1016/j.clgc.2014.08.014.
    1. Uenosono Y, Arigami T, Kozono T, et al. Clinical significance of circulating tumor cells in peripheral blood from patients with gastric cancer. Cancer. 2013;119:3984–3991. doi: 10.1002/cncr.28309.
    1. Kauffman EC, Lee MJ, Alarcon SV, et al. Lack of impact of robotic-assisted laparoscopic radical prostatectomy on intraoperative levels of prostate cancer circulating tumor cells. J Urol. 2015;195:1136–1142. doi: 10.1016/j.juro.2015.11.013.
    1. Krag DN, Ashikaga T, Moss TJ, et al. Breast cancer cells in the blood: a pilot study. Breast J. 1999;5:354–358. doi: 10.1046/j.1524-4741.1999.99016.x.
    1. Butler TP, Gullino PM. Quantitation of cell shedding into efferent blood of mammary adenocarcinoma. Cancer Res. 1975;35:512–516.

Source: PubMed

3
Abonneren