Acquisition of Portal Venous Circulating Tumor Cells From Patients With Pancreaticobiliary Cancers by Endoscopic Ultrasound

Daniel V T Catenacci, Christopher G Chapman, Peng Xu, Ann Koons, Vani J Konda, Uzma D Siddiqui, Irving Waxman, Daniel V T Catenacci, Christopher G Chapman, Peng Xu, Ann Koons, Vani J Konda, Uzma D Siddiqui, Irving Waxman

Abstract

Background & aims: Tumor cells circulate in low numbers in peripheral blood; their detection is used predominantly in metastatic disease. We evaluated the feasibility and safety of sampling portal venous blood via endoscopic ultrasound (EUS) to count portal venous circulating tumor cells (CTCs), compared with paired peripheral CTCs, in patients with pancreaticobiliary cancers (PBCs).

Methods: In a single-center cohort study, we evaluated 18 patients with suspected PBCs. Under EUS guidance, a 19-gauge EUS fine needle was advanced transhepatically into the portal vein and as many as four 7.5-mL aliquots of blood were aspirated. Paired peripheral blood samples were obtained. Epithelial-derived CTCs were sorted magnetically based on expression of epithelial cell adhesion molecules; only those with a proper morphology and found to be CD45 negative and positive for cytokeratins 8, 18, and/or 19 and 4',6-diamidino-2-phenylindole were considered to be CTCs. For 5 samples, CTCs also were isolated by flow cytometry and based on CD45 depletion. ImageStream was used to determine the relative protein levels of P16, SMAD4, and P53. DNA was extracted from CTCs for sequencing of select KRAS codons.

Results: There were no complications from portal vein blood acquisition. We detected CTCs in portal vein samples from all 18 patients (100%) vs peripheral blood samples from only 4 patients (22.2%). Patients with confirmed PBCs had a mean of 118.4 ± 36.8 CTCs/7.5 mL portal vein blood, compared with a mean of 0.8 ± 0.4 CTCs/7.5 mL peripheral blood (P < .01). The 9 patients with nonmetastatic, resectable, or borderline-resectable PBCs had a mean of 83.2 CTCs/7.5 mL portal vein blood (median, 62.0 CTCs/7.5 mL portal vein blood). In a selected patient, portal vein CTCs were found to carry the same mutations as those detected in a metastatic lymph node and expressed similar levels of P16, SMAD4, and P53 proteins.

Conclusions: It is feasible and safe to collect portal venous blood from patients undergoing EUS. We identified CTCs in all portal vein blood samples from patients with PBCs, but less than 25% of peripheral blood samples. Portal vein CTCs can be used for molecular characterization of PBCs and share features of metastatic tissue. This technique might be used to study the pathogenesis and progression of PBCs, as well as a diagnostic or prognostic tool to stratify risk of cancer recurrence or developing metastases.

Keywords: Endoscopic Ultrasound; EpCAM; Pancreatic Cancer; Portal Venous Blood.

Conflict of interest statement

Conflicts of interest

The authors disclose no conflicts.

Copyright © 2015 AGA Institute. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1
Figure 1
EUS-guided access of the portal vein. (A) The portal vein is identified under EUS guidance with Doppler wave verification. (B) EUS-guided, transhepatic, FNA puncture of the portal vein with a 19G EUS FNA needle for portal venous blood acquisition. PV, portal vein.
Figure 2
Figure 2
CTC enumeration from peripheral blood and portal venous blood. (A) Summary table of CTC enumeration per 7.5 mL of peripheral and portal venous blood by malignancy type. (B) Representative immunofluorescence staining of a single CTC and a CTC cluster captured by CellSearch enumeration (DAPI, magenta; CK, green) (C) Vertical scatter plot with the means ± SEM for CTCs enumerated from the peripheral blood and portal venous blood in patients with confirmed PBCs. Bar indicates a statistically significant difference using paired t test. (D) Box and whisker plot with individual points for CTC enumeration per 7.5 mL of portal venous blood (PVB) in all patients with confirmed PBCs and the subset of patients with borderline-resectable or resected PBCs vs unresectable PBCs. IPMN, intraductal papillary mucinous neoplasm.
Figure 3
Figure 3
Tumor-suppressor genes, p16/CDKN2A, TP53, and SMAD4 protein expression in AGS control cells and portal venous CTCs. (A) Flow cytometry of CD45-depleted portal venous blood gated by ImageStream for EpCAM+, DAPI+ CTCs. Representative immunofluorescent staining of the proteins in (B) individual AGS control cells and (C) portal venous CTCs during EpCAM+ flow cytometry via ImageStream technology. (D) Representative immunofluorescent staining of the proteins in CTC clusters identified in portal venous blood. (E) Median intensity for immunofluorescence of CTC p53, smad4, p16 proteins relative to AGS control cell line normalized to mean cell size (mean aspect ratio of the bright field). (F) Immunohistochemistry confirmation of positive smad4, loss of p16, and heterogeneous p53 expression in a metastatic lymph node relative to AGS control cells and positive control tissues (40× magnification; scale bars: 50 µm).
Figure 4
Figure 4
Confirmation of CTNNB1 mutation detected in CTC DNA. Top 2 sequences show reference protein (top row) and complementary DNA (cDNA) (second row) sequences as G (glycine) and GGA, respectively. Bottom 2 sequencing traces show forward GAA (third row) and reverse TTC (bottom row) sequencing of portal venous CTCs isolated from the patient consistent with G34E missense mutation as found in a metastatic lymph node. Red boxes highlight the sequencing location of the CTNNB1 gene with G>A nucleotide change resulting in a G34E mutation.

References

    1. Howlader NNA, Krapcho M, Garshell J, et al. SEER Cancer Statistics Review, 1975–2012, National Cancer Institute. Bethesda, MD: [Accessed: June 10, 2015]. Available from: , based on November 2014 SEER data submission.
    1. Neoptolemos JP, Stocken DD, Friess H, et al. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med. 2004;350:1200–1210.
    1. Oettle H, Post S, Neuhaus P, et al. Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing curative-intent resection of pancreatic cancer: a randomized controlled trial. JAMA. 2007;297:267–277.
    1. Barugola G, Falconi M, Bettini R, et al. The determinant factors of recurrence following resection for ductal pancreatic cancer. JOP. 2007;8:132–140.
    1. Kloek JJ, Ten Kate FJ, Busch OR, et al. Surgery for extrahepatic cholangiocarcinoma: predictors of survival. HPB (Oxford) 2008;10:190–195.
    1. Callery MP, Chang KJ, Fishman EK, et al. Pretreatment assessment of resectable and borderline resectable pancreatic cancer: expert consensus statement. Ann Surg Oncol. 2009;16:1727–1733.
    1. Donahue TR, Isacoff WH, Hines OJ, et al. Downstaging chemotherapy and alteration in the classic computed tomography/magnetic resonance imaging signs of vascular involvement in patients with pancreaticobiliary malignant tumors: influence on patient selection for surgery. Arch Surg. 2011;146:836–843.
    1. Freeny PC. Radiologic diagnosis and staging of pancreatic ductal adenocarcinoma. Radiol Clin North Am. 1989;27:121–128.
    1. Katz MH, Fleming JB, Bhosale P, et al. Response of borderline resectable pancreatic cancer to neoadjuvant therapy is not reflected by radiographic indicators. Cancer. 2012;118:5749–5756.
    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.
    1. Ting DT, Wittner BS, Ligorio M, et al. Single-cell RNA sequencing identifies extracellular matrix gene expression by pancreatic circulating tumor cells. Cell Rep. 2014;8:1905–1918.
    1. Plaks V, Koopman CD, Werb Z. Cancer. Circulating tumor cells. Science. 2013;341:1186–1188.
    1. Jiao LR, Apostolopoulos C, Jacob J, et al. Unique localization of circulating tumor cells in patients with hepatic metastases. J Clin Oncol. 2009;27:6160–6165.
    1. Waxman I, Koons A, Konda V, et al. Detection of portal vein (PV) circulating tumor cells (CTCs) in pancreatic cancer (PC) patients obtained by EUS guided PV sampling. A safety and feasibility trial. Gastrointest Endosc. 2014;79(suppl 5):AB173–AB174.
    1. Allard WJ, Matera J, Miller MC, et al. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin Cancer Res. 2004;10:6897–6904.
    1. Reyes EE, VanderWeele DJ, Isikbay M, et al. Quantitative characterization of androgen receptor protein expression and cellular localization in circulating tumor cells from patients with metastatic castration-resistant prostate cancer. J Transl Med. 2014;12:313.
    1. Oshima M, Okano K, Muraki S, et al. Immunohistochemically detected expression of 3 major genes (CDKN2A/p16, TP53, and SMAD4/DPC4) strongly predicts survival in patients with resectable pancreatic cancer. Ann Surg. 2013;258:336–346.
    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.
    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. Khoja L, Backen A, Sloane R, et al. A pilot study to explore circulating tumour cells in pancreatic cancer as a novel biomarker. Br J Cancer. 2012;106:508–516.
    1. Kurihara T, Itoi T, Sofuni A, et al. Detection of circulating tumor cells in patients with pancreatic cancer: a preliminary result. J Hepatobiliary Pancreat Surg. 2008;15:189–195.
    1. Negin BP, Meropol NJ, Alpaugh RK, et al. Characterization and prognostic significance of circulating tumor cells in the peripheral blood of patients with metastatic pancreatic cancer. J Clin Oncol. 2010;28(suppl):15s.
    1. Rhim AD, Thege FI, Santana SM, et al. Detection of circulating pancreas epithelial cells in patients with pancreatic cystic lesions. Gastroenterology. 2014;146:647–651.
    1. Yu M, Ting DT, Stott SL, et al. RNA sequencing of pancreatic circulating tumour cells implicates WNT signalling in metastasis. Nature. 2012;487:510–513.
    1. Li P, Mao Z, Peng Z, et al. Acoustic separation of circulating tumor cells. Proc Natl Acad Sci USA. 2015;112:4970–4975.
    1. Sarioglu AF, Aceto N, Kojic N, et al. A microfluidic device for label-free, physical capture of circulating tumor cell clusters. Nat Methods. 2015;12:685–691.
    1. Ozkumur E, Shah AM, Ciciliano JC, et al. Inertial focusing for tumor antigen-dependent and -independent sorting of rare circulating tumor cells. Sci Transl Med. 2013;5:179ra47.
    1. Jones S, Zhang X, Parsons DW, Cheng-Ho Lin J, Leary RJ, Angenendt P, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–1806.
    1. Hahn SA, Schutte M, Hoque AT, et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science. 1996;271:350–353.
    1. Blackford A, Serrano OK, Wolfgang CL, et al. SMAD4 gene mutations are associated with poor prognosis in pancreatic cancer. Clin Cancer Res. 2009;15:4674–4679.
    1. Caldas C, Hahn SA, da Costa LT, et al. Frequent somatic mutations and homozygous deletions of the p16 (MTS1) gene in pancreatic adenocarcinoma. Nat Genet. 1994;8:27–32.
    1. Schutte M, Hruban RH, Geradts J, et al. Abrogation of the Rb/p16 tumor-suppressive pathway in virtually all pancreatic carcinomas. Cancer Res. 1997;57:3126–3130.
    1. Rhim AD, Mirek ET, Aiello NM, et al. EMT and dissemination precede pancreatic tumor formation. Cell. 2012;148:349–361.
    1. Hishinuma S, Ogata Y, Tomikawa M, et al. Patterns of recurrence after curative resection of pancreatic cancer, based on autopsy findings. J Gastrointest Surg. 2006;10:511–518.
    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.
    1. Lohr JG, Adalsteinsson VA, Cibulskis K, et al. Whole-exome sequencing of circulating tumor cells provides a window into metastatic prostate cancer. Nat Biotechnol. 2014;32:479–484.

Source: PubMed

3
Suscribir