DOI QR코드

DOI QR Code

Clinical Utility of Portal Venous Circulating Tumor Cells in Pancreatic Cancer

췌장암에서 간 문맥 순환 종양 세포의 임상적인 유용성

  • Seung Bae Yoon (Department of Internal Medicine, Eunpyeong St. Mary's Hospital, The Catholic University of Korea College of Medicine) ;
  • Sung Woo Ko (Department of Internal Medicine, Eunpyeong St. Mary's Hospital, The Catholic University of Korea College of Medicine)
  • 윤승배 (가톨릭대학교 의과대학 은평성모병원 내과학교실) ;
  • 고성우 (가톨릭대학교 의과대학 은평성모병원 내과학교실)
  • Received : 2023.02.20
  • Accepted : 2023.02.24
  • Published : 2023.04.20

Abstract

Despite recent advancements in the diagnosis and treatment of pancreatic cancer, clinical results remain dismal. Furthermore, there are no reliable biomarkers or alternatives beyond carbohydrate antigen 19-9. Circulating tumor cells (CTCs) may be a potential biomarker, but their therapeutic application is constrained by their rarity in peripheral venous blood. Theoretically, the portal vein can be a more appropriate location for the detection of CTCs, because the first venous drainage of pancreatic cancer is portal circulation. According to several studies, the number and detection rate of CTCs may be higher in the portal blood than in the peripheral blood. CTC counts in the portal blood are strongly correlated with several prognostic parameters such as hepatic metastasis, recurrence after surgery, and survival. The phenotypic and genotypic properties analyzed in the captured portal CTCs can assist us to comprehend tumor heterogeneity and predicting the prognosis of pancreatic cancer. The investigations to date are limited by small sample sizes and varied CTC detection techniques. Therefore, a large number of prospective studies are required to confirm portal CTCs as a valid biomarker in pancreatic cancer.

Keywords

References

  1. Henley SJ, Ward EM, Scott S, et al. Annual report to the nation on the status of cancer, part I: national cancer statistics. Cancer 2020;126:2225-2249. https://doi.org/10.1002/cncr.32802 
  2. Yeo D, Bastian A, Strauss H, Saxena P, Grimison P, Rasko JEJ. Exploring the clinical utility of pancreatic cancer circulating tumor cells. Int J Mol Sci 2022;23:1671. https://doi.org/10.3390/ijms23031671 
  3. Neesse A, Bauer CA, Ohlund D, et al. Stromal biology and therapy in pancreatic cancer: ready for clinical translation? Gut 2019;68:159-171. https://doi.org/10.1136/gutjnl-2018-316451 
  4. Zhang HQ, Li J, Tan CL, Chen YH, Zheng ZJ, Liu XB. Neoadjuvant therapy in resectable pancreatic cancer: a promising curative method to improve prognosis. World J Gastrointest Oncol 2022;14:1903-1917. https://doi.org/10.4251/wjgo.v14.i10.1903 
  5. Groot VP, Gemenetzis G, Blair AB, et al. Defining and predicting early recurrence in 957 patients with resected pancreatic ductal adenocarcinoma. Ann Surg 2019;269:1154-1162. https://doi.org/10.1097/SLA.0000000000002734 
  6. Yang J, Xu R, Wang C, Qiu J, Ren B, You L. Early screening and diagnosis strategies of pancreatic cancer: a comprehensive review. Cancer Commun (Lond) 2021;41:1257-1274. https://doi.org/10.1002/cac2.12204 
  7. Massague J, Obenauf AC. Metastatic colonization by circulating tumour cells. Nature 2016;529:298-306. https://doi.org/10.1038/nature17038 
  8. 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. https://doi.org/10.1056/NEJMoa040766 
  9. Groot Koerkamp B, Rahbari NN, Buchler MW, Koch M, Weitz J. Circulating tumor cells and prognosis of patients with resectable colorectal liver metastases or widespread metastatic colorectal cancer: a meta-analysis. Ann Surg Oncol 2013;20:2156-2165. https://doi.org/10.1245/s10434-013-2907-8 
  10. Doyen J, Alix-Panabieres C, Hofman P, et al. Circulating tumor cells in prostate cancer: a potential surrogate marker of survival. Crit Rev Oncol Hematol 2012;81:241-256. https://doi.org/10.1016/j.critrevonc.2011.05.004 
  11. 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. https://doi.org/10.1200/JCO.2010.28.7045 
  12. 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. https://doi.org/10.1016/j.celrep.2014.08.029 
  13. Martini V, Timme-Bronsert S, Fichtner-Feigl S, Hoeppner J, Kulemann B. Circulating tumor cells in pancreatic cancer: current perspectives. Cancers (Basel) 2019;11:1659. https://doi.org/10.3390/cancers11111659 
  14. Luo K, Wang X, Zhang X, Liu Z, Huang S, Li R. The value of circulating tumor cells in the prognosis and treatment of pancreatic cancer. Front Oncol 2022;12:933645. https://doi.org/10.3389/fonc.2022.933645 
  15. Dong Y, Skelley AM, Merdek KD, et al. Microfluidics and circulating tumor cells. J Mol Diagn 2013;15:149-157. https://doi.org/10.1016/j.jmoldx.2012.09.004 
  16. Wang Y, Yu X, Hartmann D, Zhou J. Circulating tumor cells in peripheral blood of pancreatic cancer patients and their prognostic role: a systematic review and meta-analysis. HPB (Oxford) 2020;22:660-669. https://doi.org/10.1016/j.hpb.2019.11.003 
  17. 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. https://doi.org/10.1158/1078-0432.CCR-04-0378 
  18. 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;149:1794-1803.e4. https://doi.org/10.1053/j.gastro.2015.08.050 
  19. 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;9:2038-2045. https://doi.org/10.7150/jca.23989 
  20. Gress FG. The early history of interventional endoscopic ultrasound. Gastrointest Endosc Clin N Am 2017;27:547-550. https://doi.org/10.1016/j.giec.2017.06.015 
  21. Dietrich CF. Controversies in EUS. Endosc Ultrasound 2021;10:1-2. https://doi.org/10.4103/EUS-D-21-00024 
  22. Trikudanathan G, Pannala R, Bhutani MS, et al. EUS-guided portal vein interventions. Gastrointest Endosc 2017;85:883-888. https://doi.org/10.1016/j.gie.2017.02.019 Erratum in: Gastrointest Endosc 2017;85:1312. https://doi.org/10.1016/j.gie.2017.04.022 
  23. 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 (Baltimore) 2016;95:e3407. https://doi.org/10.1097/MD.0000000000003407 
  24. 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;20:1747-1754. https://doi.org/10.1016/j.pan.2020.10.039 
  25. Zhang Y, Su H, Wang H, et al. Endoscopic ultrasound-guided acquisition of portal venous circulating tumor cells as a potential diagnostic and prognostic tool for pancreatic cancer. Cancer Manag Res 2021;13:7649-7661. https://doi.org/10.2147/CMAR.S330473 
  26. Choi YH, Hong TH, Yoon SB, et al. Prognostic implications of portal venous circulating tumor cells in resectable pancreatic cancer. Biomedicines 2022;10:1289. https://doi.org/10.3390/biomedicines10061289 
  27. Vilhav C, Engstrom C, Naredi P, et al. Fractional uptake of circulating tumor cells into liver-lung compartments during curative resection of periampullary cancer. Oncol Lett 2018;16:6331-6338. https://doi.org/10.3892/ol.2018.9435 
  28. Poruk KE, Valero V 3rd, Saunders T, et al. Circulating tumor cell phenotype predicts recurrence and survival in pancreatic adenocarcinoma. Ann Surg 2016;264:1073-1081. https://doi.org/10.1097/SLA.0000000000001600 
  29. Bissolati M, Sandri MT, Burtulo G, Zorzino L, Balzano G, Braga M. Portal vein-circulating tumor cells predict liver metastases in patients with resectable pancreatic cancer. Tumour Biol 2015;36:991-996. https://doi.org/10.1007/s13277-014-2716-0 
  30. Pang TCY, Po JW, Becker TM, et al. Circulating tumour cells in pancreatic cancer: a systematic review and meta-analysis of clinicopathological implications. Pancreatology 2021;21:103-114. https://doi.org/10.1016/j.pan.2020.11.022 
  31. Song BG, Kwon W, Kim H, et al. Detection of circulating tumor cells in resectable pancreatic ductal adenocarcinoma: a prospective evaluation as a prognostic marker. Front Oncol 2021;10:616440. https://doi.org/10.3389/fonc.2020.616440 
  32. Padillo-Ruiz J, Suarez G, Pereira S, et al. Circulating tumor cells enumeration from the portal vein for risk stratification in early pancreatic cancer patients. Cancers (Basel) 2021;13:6153. https://doi.org/10.3390/cancers13246153 
  33. Parkinson DR, Dracopoli N, Petty BG, et al. Considerations in the development of circulating tumor cell technology for clinical use. J Transl Med 2012;10:138. https://doi.org/10.1186/1479-5876-10-138 
  34. 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;28:4615-4622. https://doi.org/10.1245/s10434-020-09518-y 
  35. Alix-Panabieres C, Pantel K. Liquid biopsy: from discovery to clinical application. Cancer Discov 2021;11:858-873. https://doi.org/10.1158/2159-8290.CD-20-1311 
  36. Kalluri R, Neilson EG. Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest 2003; 112:1776-1784. https://doi.org/10.1172/JCI20530 
  37. Pastushenko I, Brisebarre A, Sifrim A, et al. Identification of the tumour transition states occurring during EMT. Nature 2018;556:463-468. https://doi.org/10.1038/s41586-018-0040-3 
  38. 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;25:138-150. https://doi.org/10.3748/wjg.v25.i1.138 
  39. Semaan A, Bernard V, Kim DU, et al. Characterisation of circulating tumour cell phenotypes identifies a partial-EMT sub-population for clinical stratification of pancreatic cancer. Br J Cancer 2021;124:1970-1977. https://doi.org/10.1038/s41416-021-01350-9 
  40. Pan Y, Li D, Yang J, et al. Portal venous circulating tumor cells undergoing epithelial-mesenchymal transition exhibit distinct clinical significance in pancreatic ductal adenocarcinoma. Front Oncol 2021;11:757307. https://doi.org/10.3389/fonc.2021.757307 
  41. Lamy A, Blanchard F, Le Pessot F, et al. Metastatic colorectal cancer KRAS genotyping in routine practice: results and pitfalls. Mod Pathol 2011;24:1090-100. https://doi.org/10.1038/modpathol.2011.60 
  42. Vanderlaan PA, Yamaguchi N, Folch E, et al. Success and failure rates of tumor genotyping techniques in routine pathological samples with non-small-cell lung cancer. Lung Cancer 2014;84:39-44. https://doi.org/10.1016/j.lungcan.2014.01.013 
  43. Diaz LA Jr, Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol 2014;32:579-586. https://doi.org/10.1200/JCO.2012.45.2011 
  44. Ankeny JS, Court CM, Hou S, et al. Circulating tumour cells as a biomarker for diagnosis and staging in pancreatic cancer. Br J Cancer 2016;114:1367-1375. https://doi.org/10.1038/bjc.2016.121 
  45. Kulemann B, Rosch S, Seifert S, et al. Pancreatic cancer: circulating tumor cells and primary tumors show heterogeneous KRAS mutations. Sci Rep 2017;7:4510. https://doi.org/10.1038/s41598-017-04601-z Erratum in: Sci Rep 2017;7:14631. https://doi.org/10.1038/s41598-017-14870-3 
  46. Campbell PJ, Yachida S, Mudie LJ, et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature 2010;467:1109-1113. https://doi.org/10.1038/nature09460 
  47. Bailey P, Chang DK, Nones K, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 2016;531:47-52. https://doi.org/10.1038/nature16965 
  48. Gorgannezhad L, Umer M, Islam MN, Nguyen NT, Shiddiky MJA. Circulating tumor DNA and liquid biopsy: opportunities, challenges, and recent advances in detection technologies. Lab Chip 2018;18:1174-1196. https://doi.org/10.1039/C8LC00100F 
  49. Yu KH, Ricigliano M, McCarthy B, et al. Circulating tumor and invasive cell gene expression profile predicts treatment response and survival in pancreatic adenocarcinoma. Cancers (Basel) 2018;10:467. https://doi.org/10.3390/cancers10120467 
  50. Aslanian HR, Lee JH, Canto MI. AGA clinical practice update on pancreas cancer screening in high-risk individuals: expert review. Gastroenterology 2020;159:358-362. https://doi.org/10.1053/j.gastro.2020.03.088 
  51. Cohen JD, Javed AA, Thoburn C, et al. Combined circulating tumor DNA and protein biomarker-based liquid biopsy for the earlier detection of pancreatic cancers. Proc Natl Acad Sci U S A 2017;114:10202-10207. https://doi.org/10.1073/pnas.1704961114