Endoscopic Ultrasound-Guided Acquisition of Portal Venous Circulating Tumor Cells as a Potential Diagnostic and Prognostic Tool for Pancreatic Cancer

Yixuan Zhang, Haochen Su, Haibo Wang, Chenghu Xu, Siqi Zhou, Jing Zhao, Shanshan Shen, Guifang Xu, Lei Wang, Xiaoping Zou, Shu Zhang, Ying Lv, Yixuan Zhang, Haochen Su, Haibo Wang, Chenghu Xu, Siqi Zhou, Jing Zhao, Shanshan Shen, Guifang Xu, Lei Wang, Xiaoping Zou, Shu Zhang, Ying Lv

Abstract

Background: Circulating tumor cells (CTCs) were a promising liquid biopsy for pancreatic cancer (PC) but circulate in low counts in peripheral blood. We evaluated the diagnostic and prognostic values of portal vein (PoV) CTCs in PC patients.

Methods: PoV was aspirated under EUS guidance from 40 patients with suspected pancreaticobiliary cancers. Epithelial-mesenchymal-transition-related subtypes of CTCs were identified via immunofluorescence using EpCAM and Twist antibodies. The diagnostic and prognostic performance of PoV CTCs was investigated by receiver-operating characteristic (AUC) curve and Kaplan-Meier survival analysis.

Results: In total, 40 patients including 31 with PC, 4 with non-pancreatic periampullary cancer and 5 with benign pancreatic diseases (BPD) were enrolled. CTCs were detected more in PoV compared with peripheral blood. PoV CTC numbers in BPD patients were lower than in PC patients. The number of PoV CTCs, especially mesenchymal-CTCs (M-CTCs), was positively correlated with the tumor burden, instead of epithelial-CTCs (E-CTCs). The combination of PoV CTC numbers and CA19-9 demonstrated better diagnostic efficiency (AUC value 0.987) than either alone in differentiating PC with BPD. Moreover, the diagnostic efficacy of PoV CTCs and M-CTCs were obviously better than that of E-CTCs and CA19-9 in distinguishing early and late stage PC. Lastly, high PoV CTC and M-CTC numbers were both associated with shorter overall survival.

Conclusion: Acquisition of the PoV samples in PC patients via EUS-guided procedures has been proved safe and feasible. PoV CTCs, especially M-CTCs, have great potentials in diagnosing and predicting the prognosis of PC, especially in combination with CA19-9.

Keywords: circulating tumor cell; diagnosis; endoscopic ultrasound; portal vein blood; prognosis.

Conflict of interest statement

The authors report no conflicts of interest in this work.

© 2021 Zhang et al.

Figures

Figure 1
Figure 1
EUS-guided access of the portal vein. (A) Pancreatic mass was hypoechoic with poorly defined boundaries. (B) EUS-FNA to obtain histological cytology and histological examination. (C) The main portal vein was identified under EUS guidance with Doppler wave verification. (D) Puncture the primary vein with a needle for portal venous blood acquisition.
Figure 2
Figure 2
Flowchart of the protocol for CTC detection in our study. The peripheral and portal vein blood were collected from each patient. Red blood Cells (RBCs) in the samples were lysed, and residual cell pellets were resuspended. Samples were labeled with anti-leukocyte-specific antibodies (CD45) monoclonal antibody coated magnetic beads for 20 min, followed by separation of the beads using a gradient centrifugation liquid and magnetic stand. The resulting solution with any CTCs was smeared on a single slide, fixed, and dried for CTC EMT phenotype detection by immunofluorescence.
Figure 3
Figure 3
Flowchart of the enrolled patients.
Figure 4
Figure 4
Verification and detection of EMT phenotype in PC cell lines and portal vein CTCs from PC patients. (A) EMT phenotype of this platform was verified in representative “epithelial” (Bxpc3, sw1990) and “mesenchymal” (Miapaca-2, Panc-1) PC cell lines. The cells were analyzed using a 200X× objective. (B) Representative images of CTCs in portal vein blood from a patient with PC. Leukocytes were stained for CD45 (red fluorescence). CTCs were stained for epithelial markers (EpCAM, Orange fluorescence) and mesenchymal markers (Twist, green fluorescence). The cells were analyzed using a 400× objective.
Figure 5
Figure 5
Circulating tumor cells (CTCs) in portal vein blood (PoV) constitute a potential biomarker for PC. (A) CTC numbers in PoV were significantly higher than in peripheral blood (median, 10 vs 6 cell/5mL of blood, p<0.0001); (B) Comparison of PoV CTC numbers in patients with PC, non-PC and BPD. PC, pancreatic cancer; non-PC, non-pancreatic periampullary cancer; BPD, benign pancreatic diseases. (C) CTC numbers among patients with BPD, and PC patients at different stages. (D) The diagnostic performance of PoV CTC numbers, PV CTC numbers, CA19-9, and combined PoV CTC numbers with CA19-9 in differentiating PC and BPD was shown by ROC curves.
Figure 6
Figure 6
Correlation of the number of CTCs, M-CTCs and E-CTCs in PoV and the number of CTCs in PV with the clinical features of PC patients. (A) The comparison of PoV CTC numbers detected using Twist and EpCAM antibodies. (BE) Correlation of the numbers of CTCs (B) M-CTCs (C) E-CTCs (D) in portal vein blood and the numbers of CTCs in peripheral vein blood (E) with lymph node invasion, distant metastasis and PC stages. (F and G) ROC curves of CTCs, M-CTCs, E-CTCs in portal vein blood, CTCs in peripheral blood and serum CA19-9 to distinguish PC patients between stage I–II and III–IV.
Figure 7
Figure 7
Overall survival (OS) in patients with different levels of CTCs and M-CTCs in PoV. (A) Kaplan–Meier survival analysis showed that patients with lower CTC (CTCs<10/5 mL) had better OS compared with higher CTC (CTCs ≥10/5mL) (p=0.004). (B) OS in patients with lower M-CTC (M-CTCs <8/5mL) was superior to those with higher M-CTC (M-CTCs ≥8/5mL) (p=0.007). (C) The hazard ratio (HR) with 95% CI of the CTCs and M-CTCs parameters and clinicopathological characteristics for OS in PC patients based on COX regression analysis.

References

    1. Moore A, Donahue T. Pancreatic cancer. JAMA. 2019;322(14):1426. doi:10.1001/jama.2019.14699
    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69:7–34.
    1. Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–2921. doi:10.1158/0008-5472.CAN-14-0155
    1. Mobarki M, Dumollard JM, Dal Col P, Camy F, Peoc’h M, Karpathiou G. Granular cell tumor a study of 42 cases and systemic review of the literature. Pathol Res Pract. 2020;216(4):152865. doi:10.1016/j.prp.2020.152865
    1. Luo G, Liu C, Guo M, et al. Potential biomarkers in Lewis negative patients with pancreatic cancer. Ann Surg. 2017;265(4):800–805. doi:10.1097/SLA.0000000000001741
    1. Zhou B, Xu JW, Cheng YG, et al. Early detection of pancreatic cancer: Where are we now and where are we going? Int J Cancer. 2017;141(2):231–241. doi:10.1002/ijc.30670
    1. Pantel K, Speicher MR. The biology of circulating tumor cells. Oncogene. 2016;35(10):1216–1224. doi:10.1038/onc.2015.192
    1. Miyamoto DT, Lee RJ, Kalinich M, et al. An RNA-based digital circulating tumor cell Signature is predictive of drug response and early dissemination in prostate cancer. Cancer Discov. 2018;8(3):288–303. doi:10.1158/-16-1406
    1. Guo W, Sun YF, Shen MN, et al. Circulating tumor cells with stem-like phenotypes for diagnosis, prognosis, and therapeutic response evaluation in hepatocellular carcinoma. Clin Cancer Res. 2018;24(9):2203–2213. doi:10.1158/1078-0432.CCR-17-1753
    1. Deneve E, Riethdorf S, Ramos J, et al. Capture of viable circulating tumor cells in the liver of colorectal cancer patients. Clin Chem. 2013;59(9):1384–1392. doi:10.1373/clinchem.2013.202846
    1. Nieto J, Grossbard ML, Kozuch P. Metastatic pancreatic cancer 2008: is the glass less empty? Oncologist. 2008;13(5):562–576. doi:10.1634/theoncologist.2007-0181
    1. Yu M, Stott S, Toner M, Maheswaran S, Haber DA. Circulating tumor cells: approaches to isolation and characterization. J Cell Biol. 2011;192(3):373–382. doi:10.1083/jcb.201010021
    1. Buscail E, Chiche L, Laurent C, et al. Tumor-proximal liquid biopsy to improve diagnostic and prognostic performances of circulating tumor cells. Mol Oncol. 2019;13(9):1811–1826. doi:10.1002/1878-0261.12534
    1. Rahbari NN, Bork U, Kircher A, et al. Compartmental differences of circulating tumor cells in colorectal cancer. Ann Surg Oncol. 2012;19(7):2195–2202. doi:10.1245/s10434-011-2178-1
    1. Fang ZT, Zhang W, Wang GZ, et al. Circulating tumor cells in the central and peripheral venous compartment - assessing hematogenous dissemination after transarterial chemoembolization of hepatocellular carcinoma. Onco Targets Ther. 2014;7:1311–1318. doi:10.2147/OTT.S62605
    1. Reddy RM, Murlidhar V, Zhao L, et al. Pulmonary venous blood sampling significantly increases the yield of circulating tumor cells in early-stage lung cancer. J Thorac Cardiovasc Surg. 2016;151(3):852–858. doi:10.1016/j.jtcvs.2015.09.126
    1. Tien YW, Kuo HC, Ho BI, et al. A high circulating tumor cell count in portal vein predicts liver metastasis from periampullary or pancreatic cancer: a high portal venous CTC count predicts liver metastases. Medicine. 2016;95(16):e3407. doi:10.1097/MD.0000000000003407
    1. Chapman CG, Waxman I. EUS-guided portal venous sampling of circulating tumor cells. Curr Gastroenterol Rep. 2019;21(12):68. doi:10.1007/s11894-019-0733-2
    1. Catenacci DV, Chapman CG, Xu P, et al. Acquisition of portal venous circulating tumor cells from patients with pancreaticobiliary cancers by endoscopic ultrasound. Gastroenterology. 2015;149(7):1794–1803 e1794. doi:10.1053/j.gastro.2015.08.050
    1. Lee JS, Park SS, Lee YK, Norton JA, Jeffrey SS. Liquid biopsy in pancreatic ductal adenocarcinoma: current status of circulating tumor cells and circulating tumor DNA. Mol Oncol. 2019;13(8):1623–1650. doi:10.1002/1878-0261.12537
    1. Konigsberg R, Obermayr E, Bises G, et al. Detection of EpCAM positive and negative circulating tumor cells in metastatic breast cancer patients. Acta Oncol. 2011;50(5):700–710. doi:10.3109/0284186X.2010.549151
    1. Wei T, Zhang X, Zhang Q, et al. Vimentin-positive circulating tumor cells as a biomarker for diagnosis and treatment monitoring in patients with pancreatic cancer. Cancer Lett. 2019;452:237–243. doi:10.1016/j.canlet.2019.03.009
    1. Zhao XH, Wang ZR, Chen CL, et al. Molecular detection of epithelial-mesenchymal transition markers in circulating tumor cells from pancreatic cancer patients: potential role in clinical practice. World J Gastroenterol. 2019;25(1):138–150. doi:10.3748/wjg.v25.i1.138
    1. Chapman CG, Ayoub F, Swei E, et al. Endoscopic ultrasound acquired portal venous circulating tumor cells predict progression free survival and overall survival in patients with pancreaticobiliary cancers. Pancreatology. 2020;20(8):1747–1754. doi:10.1016/j.pan.2020.10.039
    1. Buscail L. Commentary: pancreatic cancer: is the worst to come? Int J Epidemiol. 2017;46(6):1774–1775. doi:10.1093/ije/dyx143
    1. Buscail E, Maulat C, Muscari F, Chiche L, Cordelier P, Dabernat S. Liquid biopsy approach for pancreatic ductal adenocarcinoma. Cancers. 2019;11(6):852. doi:10.3390/cancers11060852
    1. Chen J, Ye C, Dong J, et al. Metabolic classification of circulating tumor cells as a biomarker for metastasis and prognosis in breast cancer. J Transl Med. 2020;18(1):59. doi:10.1186/s12967-020-02237-8
    1. de Kruijff IE, Sieuwerts AM, Onstenk W, et al. Circulating tumor cell enumeration and characterization in metastatic castration-resistant prostate cancer patients treated with Cabazitaxel. Cancers. 2019;11(8):1212. doi:10.3390/cancers11081212
    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.
    1. Gao Y, Zhu Y, Zhang Z, Zhang C, Huang X, Yuan Z. Clinical significance of pancreatic circulating tumor cells using combined negative enrichment and immunostaining-fluorescence in situ hybridization. J Exp Clin Cancer Res. 2016;35(1):66. doi:10.1186/s13046-016-0340-0
    1. Chapman CG, Waxman I. Portal-vein blood samples as a new diagnostic entity for pancreatic cancer. Expert Rev Gastroenterol Hepatol. 2016;10(6):665–667. doi:10.1080/17474124.2016.1176911
    1. White MG, Lee A, Vicente D, et al. Measurement of portal vein blood circulating tumor cells is safe and may correlate with outcomes in resected pancreatic ductal adenocarcinoma. Ann Surg Oncol. 2021;28(8):4615–4622. doi:10.1245/s10434-020-09518-y
    1. Dong X, Ma Y, Zhao X, et al. Spatial heterogeneity in epithelial to mesenchymal transition properties of circulating tumor cells associated with distant recurrence in pancreatic cancer patients. Ann Transl Med. 2020;8(11):676. doi:10.21037/atm-20-782
    1. Gall TM, Jacob J, Frampton AE, et al. Reduced dissemination of circulating tumor cells with no-touch isolation surgical technique in patients with pancreatic cancer. JAMA Surg. 2014;149(5):482–485. doi:10.1001/jamasurg.2013.3643
    1. Liu X, Li C, Li J, et al. Detection of CTCs in portal vein was associated with intrahepatic metastases and prognosis in patients with advanced pancreatic cancer. J Cancer. 2018;9(11):2038–2045. doi:10.7150/jca.23989
    1. Bidard FC, Huguet F, Louvet C, et al. Circulating tumor cells in locally advanced pancreatic adenocarcinoma: the ancillary CirCe 07 study to the LAP 07 trial. Ann Oncol. 2013;24(8):2057–2061. doi:10.1093/annonc/mdt176
    1. Okubo K, Uenosono Y, Arigami T, et al. Clinical impact of circulating tumor cells and therapy response in pancreatic cancer. Eur J Surg Oncol. 2017;43:1050–1055.

Source: PubMed

3
订阅