Skip to main content
Top
Published in: Journal of Gastroenterology 11/2023

Open Access 12-09-2023 | Aspiration | Review

Current status and issues in genomic analysis using EUS-FNA/FNB specimens in hepatobiliary–pancreatic cancers

Authors: Yoshinori Ozono, Hiroshi Kawakami, Naomi Uchiyama, Hiroshi Hatada, Souichiro Ogawa

Published in: Journal of Gastroenterology | Issue 11/2023

Login to get access

Abstract

Comprehensive genomic profiling based on next-generation sequencing has recently been used to provide precision medicine for various advanced cancers. Endoscopic ultrasound (EUS)-guided fine-needle aspiration (EUS-FNA) and EUS-guided fine-needle biopsy (EUS-FNB) play essential roles in the diagnosis of abdominal masses, mainly pancreatic cancers. In recent years, CGP analysis using EUS-FNA/FNB specimens for hepatobiliary–pancreatic cancers has increased; however, the success rate of CGP analysis is not clinically satisfactory, and many issues need to be resolved to improve the success rate of CGP analysis. In this article, we review the transition from EUS-FNA to FNB, compare each test, and discuss the current status and issues in genomic analysis of hepatobiliary–pancreatic cancers using EUS-FNA/FNB specimens.
Literature
1.
go back to reference Wani S, Muthusamy VR, Komanduri S. EUS-guided tissue acquisition: an evidence-based approach (with videos). Gastrointest Endosc. 2014;80:939-59.e7.PubMedCrossRef Wani S, Muthusamy VR, Komanduri S. EUS-guided tissue acquisition: an evidence-based approach (with videos). Gastrointest Endosc. 2014;80:939-59.e7.PubMedCrossRef
2.
go back to reference Banafea O, Mghanga FP, Zhao J, et al. Endoscopic ultrasonography with fine-needle aspiration for histological diagnosis of solid pancreatic masses: a meta-analysis of diagnostic accuracy studies. BMC Gastroenterol. 2016;16:108.PubMedCrossRefPubMedCentral Banafea O, Mghanga FP, Zhao J, et al. Endoscopic ultrasonography with fine-needle aspiration for histological diagnosis of solid pancreatic masses: a meta-analysis of diagnostic accuracy studies. BMC Gastroenterol. 2016;16:108.PubMedCrossRefPubMedCentral
3.
go back to reference Matsumoto K, Takeda Y, Onoyama T, et al. Endoscopic ultrasound-guided fine-needle aspiration biopsy—recent topics and technical tips. World J Clin Cases. 2019;7:1775–83.PubMedCrossRefPubMedCentral Matsumoto K, Takeda Y, Onoyama T, et al. Endoscopic ultrasound-guided fine-needle aspiration biopsy—recent topics and technical tips. World J Clin Cases. 2019;7:1775–83.PubMedCrossRefPubMedCentral
4.
go back to reference Khan MA, Grimm IS, Ali B, et al. A meta-analysis of endoscopic ultrasound-fine-needle aspiration compared to endoscopic ultrasound-fine-needle biopsy: diagnostic yield and the value of onsite cytopathological assessment. Endosc Int Open. 2017;5:E363–75.PubMedCrossRefPubMedCentral Khan MA, Grimm IS, Ali B, et al. A meta-analysis of endoscopic ultrasound-fine-needle aspiration compared to endoscopic ultrasound-fine-needle biopsy: diagnostic yield and the value of onsite cytopathological assessment. Endosc Int Open. 2017;5:E363–75.PubMedCrossRefPubMedCentral
5.
go back to reference Tiriac H, Bucobo JC, Tzimas D, et al. Successful creation of pancreatic cancer organoids by means of EUS-guided fine-needle biopsy sampling for personalized cancer treatment. Gastrointest Endosc. 2018;87:1474–80.PubMedCrossRefPubMedCentral Tiriac H, Bucobo JC, Tzimas D, et al. Successful creation of pancreatic cancer organoids by means of EUS-guided fine-needle biopsy sampling for personalized cancer treatment. Gastrointest Endosc. 2018;87:1474–80.PubMedCrossRefPubMedCentral
6.
go back to reference Puli SR, Bechtold ML, Buxbaum JL, et al. How good is endoscopic ultrasound-guided fine-needle aspiration in diagnosing the correct etiology for a solid pancreatic mass?: a meta-analysis and systematic review. Pancreas. 2013;42:20–6.PubMedCrossRef Puli SR, Bechtold ML, Buxbaum JL, et al. How good is endoscopic ultrasound-guided fine-needle aspiration in diagnosing the correct etiology for a solid pancreatic mass?: a meta-analysis and systematic review. Pancreas. 2013;42:20–6.PubMedCrossRef
7.
go back to reference Yang Y, Li L, Qu C, et al. Endoscopic ultrasound-guided fine needle core biopsy for the diagnosis of pancreatic malignant lesions: a systematic review and Meta-Analysis. Sci Rep. 2016;6:22978.PubMedCrossRefPubMedCentral Yang Y, Li L, Qu C, et al. Endoscopic ultrasound-guided fine needle core biopsy for the diagnosis of pancreatic malignant lesions: a systematic review and Meta-Analysis. Sci Rep. 2016;6:22978.PubMedCrossRefPubMedCentral
8.
go back to reference Madhoun MF, Wani SB, Rastogi A, et al. The diagnostic accuracy of 22-gauge and 25-gauge needles in endoscopic ultrasound-guided fine needle aspiration of solid pancreatic lesions: a meta-analysis. Endoscopy. 2013;45:86–92.PubMedCrossRef Madhoun MF, Wani SB, Rastogi A, et al. The diagnostic accuracy of 22-gauge and 25-gauge needles in endoscopic ultrasound-guided fine needle aspiration of solid pancreatic lesions: a meta-analysis. Endoscopy. 2013;45:86–92.PubMedCrossRef
9.
go back to reference Capurso G, Archibugi L, Petrone MC, et al. Slow-pull compared to suction technique for EUS-guided sampling of pancreatic solid lesions: a meta-analysis of randomized controlled trials. Endosc Int Open. 2020;8:E636–43.PubMedCrossRefPubMedCentral Capurso G, Archibugi L, Petrone MC, et al. Slow-pull compared to suction technique for EUS-guided sampling of pancreatic solid lesions: a meta-analysis of randomized controlled trials. Endosc Int Open. 2020;8:E636–43.PubMedCrossRefPubMedCentral
10.
go back to reference Wang J, Xu H, Xu C, et al. Stylet slow-pull versus standard suction for endoscopic ultrasound-guided fine-needle aspiration/biopsy of pancreatic solid masses: a meta-analysis. J Clin Gastroenterol. 2021;55:103–9.PubMedCrossRef Wang J, Xu H, Xu C, et al. Stylet slow-pull versus standard suction for endoscopic ultrasound-guided fine-needle aspiration/biopsy of pancreatic solid masses: a meta-analysis. J Clin Gastroenterol. 2021;55:103–9.PubMedCrossRef
11.
go back to reference Nakai Y, Hamada T, Hakuta R, et al. A meta-analysis of slow pull versus suction for endoscopic ultrasound-guided tissue acquisition. Gut Liver. 2021;15:625–33.PubMedCrossRefPubMedCentral Nakai Y, Hamada T, Hakuta R, et al. A meta-analysis of slow pull versus suction for endoscopic ultrasound-guided tissue acquisition. Gut Liver. 2021;15:625–33.PubMedCrossRefPubMedCentral
12.
go back to reference Storm AC, Lee LS. Endoscopic ultrasound-guided techniques for diagnosing pancreatic mass lesions: can we do better? World J Gastroenterol. 2016;22:8658–69.PubMedCrossRefPubMedCentral Storm AC, Lee LS. Endoscopic ultrasound-guided techniques for diagnosing pancreatic mass lesions: can we do better? World J Gastroenterol. 2016;22:8658–69.PubMedCrossRefPubMedCentral
13.
go back to reference Fabbri C, Luigiano C, Maimone A, et al. Endoscopic ultrasound-guided fine-needle biopsy of small solid pancreatic lesions using a 22-gauge needle with side fenestration. Surg Endosc. 2015;29:1586–90.PubMedCrossRef Fabbri C, Luigiano C, Maimone A, et al. Endoscopic ultrasound-guided fine-needle biopsy of small solid pancreatic lesions using a 22-gauge needle with side fenestration. Surg Endosc. 2015;29:1586–90.PubMedCrossRef
14.
go back to reference Kanno A, Masamune A, Hanada K, et al. Multicenter study of early pancreatic cancer in Japan. Pancreatology. 2018;18:61–7.PubMedCrossRef Kanno A, Masamune A, Hanada K, et al. Multicenter study of early pancreatic cancer in Japan. Pancreatology. 2018;18:61–7.PubMedCrossRef
15.
go back to reference Li Z, Liu W, Xu X, et al. A meta-analysis comparing endoscopic ultrasound-guided fine-needle aspiration with endoscopic ultrasound-guided fine-needle biopsy. J Clin Gsatroenterol. 2022;56:668–78.CrossRef Li Z, Liu W, Xu X, et al. A meta-analysis comparing endoscopic ultrasound-guided fine-needle aspiration with endoscopic ultrasound-guided fine-needle biopsy. J Clin Gsatroenterol. 2022;56:668–78.CrossRef
16.
go back to reference Riet PA, Erler NS, Bruno MJ, et al. Comparison of fine-needle aspiration and fine-needle biopsy devices for endoscopic ultrasound-guided sampling of solid lesions: a systemic review and meta-analysis. Endoscopy. 2021;53:411–23.PubMedCrossRef Riet PA, Erler NS, Bruno MJ, et al. Comparison of fine-needle aspiration and fine-needle biopsy devices for endoscopic ultrasound-guided sampling of solid lesions: a systemic review and meta-analysis. Endoscopy. 2021;53:411–23.PubMedCrossRef
17.
go back to reference Facciorusso A, Bajwa HS, Menon K, et al. Comparison between 22G aspiration and 22G biopsy needles for EUS-guided sampling of pancreatic lesions: a meta-analysis. Endosc Ultrasound. 2020;9:167–74.PubMedCrossRef Facciorusso A, Bajwa HS, Menon K, et al. Comparison between 22G aspiration and 22G biopsy needles for EUS-guided sampling of pancreatic lesions: a meta-analysis. Endosc Ultrasound. 2020;9:167–74.PubMedCrossRef
18.
go back to reference Facciorusso A, Wani S, Triantafyllou K, et al. Comparative accuracy of needle sizes and designs for EUS tissue sampling of solid pancreatic masses: a network meta-analysis. Gastrointest Endosc. 2019;90:893-903.e7.PubMedCrossRef Facciorusso A, Wani S, Triantafyllou K, et al. Comparative accuracy of needle sizes and designs for EUS tissue sampling of solid pancreatic masses: a network meta-analysis. Gastrointest Endosc. 2019;90:893-903.e7.PubMedCrossRef
19.
go back to reference Li H, Li W, Zhou QY, et al. Fine needle biopsy is superior to fine needle aspiration in endoscopic ultrasound guided sampling of pancreatic masses: a meta-analysis of randomized controlled trials. Medicine (Baltimore). 2018;97: e0207.PubMedCrossRef Li H, Li W, Zhou QY, et al. Fine needle biopsy is superior to fine needle aspiration in endoscopic ultrasound guided sampling of pancreatic masses: a meta-analysis of randomized controlled trials. Medicine (Baltimore). 2018;97: e0207.PubMedCrossRef
20.
go back to reference Wang J, Zhao S, Chen Y, et al. Endoscopic ultrasound guided fine needle aspiration versus endoscopic ultrasound guided fine needle biopsy in sampling pancreatic masses: a meta-analysis. Medicine (Baltimore). 2017;96: e7452.PubMedCrossRef Wang J, Zhao S, Chen Y, et al. Endoscopic ultrasound guided fine needle aspiration versus endoscopic ultrasound guided fine needle biopsy in sampling pancreatic masses: a meta-analysis. Medicine (Baltimore). 2017;96: e7452.PubMedCrossRef
21.
go back to reference Renelus BD, Jamorabo DS, Boston I, et al. Endoscopic ultrasound-guided fine needle biopsy needles provide higher diagnostic yield compared to endoscopic ultrasound-guided fine needle aspiration needles when sampling solid pancreatic lesions: a meta-analysis. Clin Endosc. 2021;54:261–8.PubMedCrossRef Renelus BD, Jamorabo DS, Boston I, et al. Endoscopic ultrasound-guided fine needle biopsy needles provide higher diagnostic yield compared to endoscopic ultrasound-guided fine needle aspiration needles when sampling solid pancreatic lesions: a meta-analysis. Clin Endosc. 2021;54:261–8.PubMedCrossRef
22.
go back to reference Pouw RE, Barret M, Biermann K, et al. Endoscopic tissue sampling—part 1: upper gastrointestinal and hepatopancreatobiliary tracts. European Society of Gastrointestinal Endoscopy (ESGE) Guideline. Endoscopy. 2021;53:1174–88.PubMedCrossRef Pouw RE, Barret M, Biermann K, et al. Endoscopic tissue sampling—part 1: upper gastrointestinal and hepatopancreatobiliary tracts. European Society of Gastrointestinal Endoscopy (ESGE) Guideline. Endoscopy. 2021;53:1174–88.PubMedCrossRef
23.
go back to reference Chen YI, Chatterjee A, Berger R, et al. Endoscopic ultrasound (EUS)-guided fine needle biopsy alone vs. EUS-guided fine needle aspiration with rapid onsite evaluation in pancreatic lesions: a multicenter randomized trial. Endoscopy. 2022;54:4–12.PubMedCrossRef Chen YI, Chatterjee A, Berger R, et al. Endoscopic ultrasound (EUS)-guided fine needle biopsy alone vs. EUS-guided fine needle aspiration with rapid onsite evaluation in pancreatic lesions: a multicenter randomized trial. Endoscopy. 2022;54:4–12.PubMedCrossRef
24.
go back to reference Kanno A, Yasuda I, Irisawa A, et al. Adverse events of endoscopic ultrasound-guided fine-needle aspiration for histologic diagnosis in Japanese tertiary centers: multicenter retrospective study. Dig Endosc. 2021;33:1146–57.PubMedCrossRef Kanno A, Yasuda I, Irisawa A, et al. Adverse events of endoscopic ultrasound-guided fine-needle aspiration for histologic diagnosis in Japanese tertiary centers: multicenter retrospective study. Dig Endosc. 2021;33:1146–57.PubMedCrossRef
25.
go back to reference Wang KX, Ben QW, Jin ZD, et al. Assessment of morbidity and mortality associated with EUS-guided FNA: a systematic review. Gastrointest Endosc. 2011;73:283–90.PubMedCrossRef Wang KX, Ben QW, Jin ZD, et al. Assessment of morbidity and mortality associated with EUS-guided FNA: a systematic review. Gastrointest Endosc. 2011;73:283–90.PubMedCrossRef
26.
go back to reference Facciorusso A, Crinò SF, Gkolfakis P, et al. Needle tract seeding after endoscopic ultrasound tissue acquisition of pancreatic lesions: a systematic review and meta-analysis. Diagnostics (Basel). 2022;12:2113.PubMedCrossRefPubMedCentral Facciorusso A, Crinò SF, Gkolfakis P, et al. Needle tract seeding after endoscopic ultrasound tissue acquisition of pancreatic lesions: a systematic review and meta-analysis. Diagnostics (Basel). 2022;12:2113.PubMedCrossRefPubMedCentral
27.
go back to reference Kawabata H, Miyazawa Y, Sato H, et al. Genetic analysis of postoperative recurrence of pancreatic cancer potentially owing to needle tract seeding during EUS-FNB. Endosc Int Open. 2019;7:E1768–72.PubMedCrossRefPubMedCentral Kawabata H, Miyazawa Y, Sato H, et al. Genetic analysis of postoperative recurrence of pancreatic cancer potentially owing to needle tract seeding during EUS-FNB. Endosc Int Open. 2019;7:E1768–72.PubMedCrossRefPubMedCentral
28.
go back to reference Nakatsubo R, Yamamoto K, Itoi T, et al. Histopathological evaluation of needle tract seeding caused by EUS-fine-needle biopsy based on resected specimens from patients with solid pancreatic masses: an analysis of 73 consecutive cases. Endosc Ultrasound. 2021;10:207–13.PubMedCrossRefPubMedCentral Nakatsubo R, Yamamoto K, Itoi T, et al. Histopathological evaluation of needle tract seeding caused by EUS-fine-needle biopsy based on resected specimens from patients with solid pancreatic masses: an analysis of 73 consecutive cases. Endosc Ultrasound. 2021;10:207–13.PubMedCrossRefPubMedCentral
29.
go back to reference Ishigaki K, Nakai Y, Oyama H, et al. Endoscopic ultrasound-guided tissue acquisition by 22-Gauge Franseen and standard needles for solid pancreatic lesions. Gut Liver. 2020;14:817–25.PubMedCrossRefPubMedCentral Ishigaki K, Nakai Y, Oyama H, et al. Endoscopic ultrasound-guided tissue acquisition by 22-Gauge Franseen and standard needles for solid pancreatic lesions. Gut Liver. 2020;14:817–25.PubMedCrossRefPubMedCentral
30.
go back to reference Dumonceau JM, Deprez PH, Jenssen C, et al. Indications, results, and clinical impact of endoscopic ultrasound (EUS)-guided sampling in gastroenterology: European Society of Gastrointestinal Endoscopy (ESGE) Clinical Guideline—Updated January 2017. Endoscopy. 2017;49:695–714.PubMedCrossRef Dumonceau JM, Deprez PH, Jenssen C, et al. Indications, results, and clinical impact of endoscopic ultrasound (EUS)-guided sampling in gastroenterology: European Society of Gastrointestinal Endoscopy (ESGE) Clinical Guideline—Updated January 2017. Endoscopy. 2017;49:695–714.PubMedCrossRef
31.
go back to reference Cheng B, Zhang Y, Chen Q, et al. Analysis of fine-needle biopsy vs fine-needle aspiration in diagnosis of pancreatic and abdominal masses: a prospective, multicenter randomized controlled trial. Clin Gastroenterol Hepatol. 2018;16:1314–21.PubMedCrossRef Cheng B, Zhang Y, Chen Q, et al. Analysis of fine-needle biopsy vs fine-needle aspiration in diagnosis of pancreatic and abdominal masses: a prospective, multicenter randomized controlled trial. Clin Gastroenterol Hepatol. 2018;16:1314–21.PubMedCrossRef
32.
go back to reference Tian L, Tang AL, Zhang L, et al. Evaluation of 22G fine-needle aspiration (FNA) versus fine-needle biopsy (FNB) for endoscopic ultrasound-guided sampling of pancreatic lesions: a prospective comparison study. Surg Endosc. 2018;32:3533–9.PubMedCrossRefPubMedCentral Tian L, Tang AL, Zhang L, et al. Evaluation of 22G fine-needle aspiration (FNA) versus fine-needle biopsy (FNB) for endoscopic ultrasound-guided sampling of pancreatic lesions: a prospective comparison study. Surg Endosc. 2018;32:3533–9.PubMedCrossRefPubMedCentral
33.
go back to reference Zhou W, Li SY, Jiang H, et al. Optimal number of needle passes during EUS-guided fine-needle biopsy of solid pancreatic lesions with 22G ProCore needles and different suction techniques: a randomized controlled trial. Endosc Ultrasound. 2021;10:62–70.PubMedCrossRefPubMedCentral Zhou W, Li SY, Jiang H, et al. Optimal number of needle passes during EUS-guided fine-needle biopsy of solid pancreatic lesions with 22G ProCore needles and different suction techniques: a randomized controlled trial. Endosc Ultrasound. 2021;10:62–70.PubMedCrossRefPubMedCentral
34.
go back to reference Jones S, Zhang X, Parsons DW, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–6.PubMedCrossRefPubMedCentral Jones S, Zhang X, Parsons DW, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–6.PubMedCrossRefPubMedCentral
35.
go back to reference Kameta E, Sugimori K, Kaneko T, et al. Diagnosis of pancreatic lesions collected by endoscopic ultrasound-guided fine-needle aspiration using next-generation sequencing. Oncol Lett. 2016;12:3875–81.PubMedCrossRefPubMedCentral Kameta E, Sugimori K, Kaneko T, et al. Diagnosis of pancreatic lesions collected by endoscopic ultrasound-guided fine-needle aspiration using next-generation sequencing. Oncol Lett. 2016;12:3875–81.PubMedCrossRefPubMedCentral
36.
go back to reference Dreyer SB, Jamieson NB, Evers L, et al. Feasibility and clinical utility of endoscopic ultrasound guided biopsy of pancreatic cancer for next-generation molecular profiling. Chin Clin Oncol. 2019;8:16.PubMedCrossRef Dreyer SB, Jamieson NB, Evers L, et al. Feasibility and clinical utility of endoscopic ultrasound guided biopsy of pancreatic cancer for next-generation molecular profiling. Chin Clin Oncol. 2019;8:16.PubMedCrossRef
37.
go back to reference Elhanafi S, Mahmud N, Vergara N, et al. Comparison of endoscopic ultrasound tissue acquisition methods for genomic analysis of pancreatic cancer. J Gastroenterol Hepatol. 2019;34:907–13.PubMedCrossRef Elhanafi S, Mahmud N, Vergara N, et al. Comparison of endoscopic ultrasound tissue acquisition methods for genomic analysis of pancreatic cancer. J Gastroenterol Hepatol. 2019;34:907–13.PubMedCrossRef
38.
39.
go back to reference Yang J, Li J, Zhu R, et al. K-ras mutational status in cytohistological tissue as a molecular marker for the diagnosis of pancreatic cancer: a systematic review and meta-analysis. Dis Markers. 2014;2014: 573783.PubMedCrossRefPubMedCentral Yang J, Li J, Zhu R, et al. K-ras mutational status in cytohistological tissue as a molecular marker for the diagnosis of pancreatic cancer: a systematic review and meta-analysis. Dis Markers. 2014;2014: 573783.PubMedCrossRefPubMedCentral
40.
go back to reference Gleeson FC, Kerr SE, Kipp BR, et al. Targeted next generation sequencing of endoscopic ultrasound acquired cytology from ampullary and pancreatic adenocarcinoma has the potential to aid patient stratification for optimal therapy selection. Oncotarget. 2016;7:54526–36.PubMedCrossRefPubMedCentral Gleeson FC, Kerr SE, Kipp BR, et al. Targeted next generation sequencing of endoscopic ultrasound acquired cytology from ampullary and pancreatic adenocarcinoma has the potential to aid patient stratification for optimal therapy selection. Oncotarget. 2016;7:54526–36.PubMedCrossRefPubMedCentral
41.
go back to reference Young G, Wang K, He J, et al. Clinical next-generation sequencing successfully applied to fine-needle aspirations of pulmonary and pancreatic neoplasms. Cancer Cytopathol. 2013;121:688–94.PubMedCrossRef Young G, Wang K, He J, et al. Clinical next-generation sequencing successfully applied to fine-needle aspirations of pulmonary and pancreatic neoplasms. Cancer Cytopathol. 2013;121:688–94.PubMedCrossRef
42.
go back to reference Mas L, Lupinacci RM, Cros J, et al. Intraductal papillary mucinous carcinoma versus conventional pancreatic ductal adenocarcinoma: a comprehensive review of clinical-pathological features, outcomes, and molecular insights. Int J Mol Sci. 2021;22:6756.PubMedCrossRefPubMedCentral Mas L, Lupinacci RM, Cros J, et al. Intraductal papillary mucinous carcinoma versus conventional pancreatic ductal adenocarcinoma: a comprehensive review of clinical-pathological features, outcomes, and molecular insights. Int J Mol Sci. 2021;22:6756.PubMedCrossRefPubMedCentral
43.
44.
go back to reference Fujikura K, Hosoda W, Felsenstein M, et al. Multiregion whole-exome sequencing of intraductal papillary mucinous neoplasms reveals frequent somatic KLF4 mutations predominantly in low-grade regions. Gut. 2021;70:928–39.PubMedCrossRef Fujikura K, Hosoda W, Felsenstein M, et al. Multiregion whole-exome sequencing of intraductal papillary mucinous neoplasms reveals frequent somatic KLF4 mutations predominantly in low-grade regions. Gut. 2021;70:928–39.PubMedCrossRef
45.
go back to reference Omori Y, Ono Y, Tanino M, et al. Pathways of progression from intraductal papillary mucinous neoplasm to pancreatic ductal adenocarcinoma based on molecular features. Gastroenterology. 2019;156:647–61.PubMedCrossRef Omori Y, Ono Y, Tanino M, et al. Pathways of progression from intraductal papillary mucinous neoplasm to pancreatic ductal adenocarcinoma based on molecular features. Gastroenterology. 2019;156:647–61.PubMedCrossRef
46.
go back to reference Yachida S, Totoki Y, Noë M, et al. Comprehensive genomic profiling of neuroendocrine carcinomas of the gastrointestinal system. Cancer Discov. 2022;12:692–711.PubMedCrossRefPubMedCentral Yachida S, Totoki Y, Noë M, et al. Comprehensive genomic profiling of neuroendocrine carcinomas of the gastrointestinal system. Cancer Discov. 2022;12:692–711.PubMedCrossRefPubMedCentral
47.
go back to reference Konukiewitz B, Jesinghaus M, Kasajima A, et al. Neuroendocrine neoplasms of the pancreas: diagnosis and pitfalls. Virchows Arch. 2022;480:247–57.PubMedCrossRef Konukiewitz B, Jesinghaus M, Kasajima A, et al. Neuroendocrine neoplasms of the pancreas: diagnosis and pitfalls. Virchows Arch. 2022;480:247–57.PubMedCrossRef
48.
go back to reference Larson BK, Tuli R, Jamil LH, et al. Utility of endoscopic ultrasound-guided biopsy for next-generation sequencing of pancreatic exocrine malignancies. Pancreas. 2018;47:990–5.PubMedCrossRef Larson BK, Tuli R, Jamil LH, et al. Utility of endoscopic ultrasound-guided biopsy for next-generation sequencing of pancreatic exocrine malignancies. Pancreas. 2018;47:990–5.PubMedCrossRef
49.
go back to reference Kandel P, Nassar A, Gomez V, et al. Comparison of endoscopic ultrasound-guided fine-needle biopsy versus fine-needle aspiration for genomic profiling and DNA yield in pancreatic cancer: a randomized crossover trial. Endoscopy. 2021;53:376–82.PubMedCrossRef Kandel P, Nassar A, Gomez V, et al. Comparison of endoscopic ultrasound-guided fine-needle biopsy versus fine-needle aspiration for genomic profiling and DNA yield in pancreatic cancer: a randomized crossover trial. Endoscopy. 2021;53:376–82.PubMedCrossRef
50.
go back to reference Gan Q, Roy-Chowdhuri S, Duose DY, et al. Adequacy evaluation and use of pancreatic adenocarcinoma specimens for next-generation sequencing acquired by endoscopic ultrasound-guided FNA and FNB. Cancer Cytopathol. 2022;130:275–83.PubMedCrossRef Gan Q, Roy-Chowdhuri S, Duose DY, et al. Adequacy evaluation and use of pancreatic adenocarcinoma specimens for next-generation sequencing acquired by endoscopic ultrasound-guided FNA and FNB. Cancer Cytopathol. 2022;130:275–83.PubMedCrossRef
51.
go back to reference Kondo T, Matsubara J, Quy PN, et al. Comprehensive genomic profiling for patients with chemotherapy-naïve advanced cancer. Cancer Sci. 2021;112:296–304.PubMedCrossRef Kondo T, Matsubara J, Quy PN, et al. Comprehensive genomic profiling for patients with chemotherapy-naïve advanced cancer. Cancer Sci. 2021;112:296–304.PubMedCrossRef
52.
go back to reference Ishizawa T, Makino N, Matsuda A, et al. Usefulness of rapid on-site evaluation specimens from endoscopic ultrasound-guided fine-needle aspiration for cancer gene panel testing: a retrospective study. PLoS ONE. 2020;15: e0228565.PubMedCrossRefPubMedCentral Ishizawa T, Makino N, Matsuda A, et al. Usefulness of rapid on-site evaluation specimens from endoscopic ultrasound-guided fine-needle aspiration for cancer gene panel testing: a retrospective study. PLoS ONE. 2020;15: e0228565.PubMedCrossRefPubMedCentral
53.
go back to reference Park JK, Lee JH, Noh DH, et al. Factors of endoscopic ultrasound-guided tissue acquisition for successful next-generation sequencing in pancreatic ductal adenocarcinoma. Gut Liver. 2020;14:387–94.PubMedCrossRef Park JK, Lee JH, Noh DH, et al. Factors of endoscopic ultrasound-guided tissue acquisition for successful next-generation sequencing in pancreatic ductal adenocarcinoma. Gut Liver. 2020;14:387–94.PubMedCrossRef
54.
go back to reference Lowery MA, Jordan EJ, Basturk O, et al. Real-time genomic profiling of pancreatic ductal adenocarcinoma: potential actionability and correlation with clinical phenotype. Clin Cancer Res. 2017;23:6094–100.PubMedCrossRef Lowery MA, Jordan EJ, Basturk O, et al. Real-time genomic profiling of pancreatic ductal adenocarcinoma: potential actionability and correlation with clinical phenotype. Clin Cancer Res. 2017;23:6094–100.PubMedCrossRef
55.
go back to reference Valero V 3rd, Saunders TJ, He J, et al. Reliable detection of somatic mutations in fine needle aspirates of pancreatic cancer with next-generation sequencing: implications for surgical management. Ann Surg. 2016;263:153–61.PubMedCrossRef Valero V 3rd, Saunders TJ, He J, et al. Reliable detection of somatic mutations in fine needle aspirates of pancreatic cancer with next-generation sequencing: implications for surgical management. Ann Surg. 2016;263:153–61.PubMedCrossRef
56.
go back to reference Rodriguez SA, Impey SD, Pelz C, et al. RNA sequencing distinguishes benign from malignant pancreatic lesions sampled by EUS-guided FNA. Gastrointest Endosc. 2016;84:252–8.PubMedCrossRef Rodriguez SA, Impey SD, Pelz C, et al. RNA sequencing distinguishes benign from malignant pancreatic lesions sampled by EUS-guided FNA. Gastrointest Endosc. 2016;84:252–8.PubMedCrossRef
57.
go back to reference Takano S, Fukasawa M, Shindo H, et al. Clinical significance of genetic alterations in endoscopically obtained pancreatic cancer specimens. Cancer Med. 2021;10:1264–74.PubMedCrossRefPubMedCentral Takano S, Fukasawa M, Shindo H, et al. Clinical significance of genetic alterations in endoscopically obtained pancreatic cancer specimens. Cancer Med. 2021;10:1264–74.PubMedCrossRefPubMedCentral
58.
go back to reference Habib JR, Zhu Y, Yin L, et al. Reliable detection of somatic mutations for pancreatic cancer in endoscopic ultrasonography-guided fine needle aspirates with next-generation sequencing: implications from a prospective cohort study. J Gastrointest Surg. 2021;25:3149–59.PubMedCrossRef Habib JR, Zhu Y, Yin L, et al. Reliable detection of somatic mutations for pancreatic cancer in endoscopic ultrasonography-guided fine needle aspirates with next-generation sequencing: implications from a prospective cohort study. J Gastrointest Surg. 2021;25:3149–59.PubMedCrossRef
59.
go back to reference Semaan A, Bernard V, Lee JJ, et al. Defining the comprehensive genomic landscapes of pancreatic ductal adenocarcinoma using real-world endoscopic aspiration samples. Clin Cancer Res. 2021;27:1082–93.PubMedCrossRef Semaan A, Bernard V, Lee JJ, et al. Defining the comprehensive genomic landscapes of pancreatic ductal adenocarcinoma using real-world endoscopic aspiration samples. Clin Cancer Res. 2021;27:1082–93.PubMedCrossRef
60.
go back to reference Carrara S, Soldà G, Di Leo M, et al. Side-by-side comparison of next-generation sequencing, cytology, and histology in diagnosing locally advanced pancreatic adenocarcinoma. Gastrointest Endosc. 2021;93:597–604.PubMedCrossRef Carrara S, Soldà G, Di Leo M, et al. Side-by-side comparison of next-generation sequencing, cytology, and histology in diagnosing locally advanced pancreatic adenocarcinoma. Gastrointest Endosc. 2021;93:597–604.PubMedCrossRef
61.
go back to reference Hisada Y, Hijioka S, Ikeda G, et al. Proportion of unresectable pancreatic cancer specimens obtained by endoscopic ultrasound-guided tissue acquisition meeting the OncoGuide™ NCC Oncopanel System analysis suitability criteria: a single-arm, phase II clinical trial. J Gastroenterol. 2022;57:990–8.PubMedCrossRef Hisada Y, Hijioka S, Ikeda G, et al. Proportion of unresectable pancreatic cancer specimens obtained by endoscopic ultrasound-guided tissue acquisition meeting the OncoGuide™ NCC Oncopanel System analysis suitability criteria: a single-arm, phase II clinical trial. J Gastroenterol. 2022;57:990–8.PubMedCrossRef
62.
go back to reference Ikeda G, Hijioka S, Nagashio Y, et al. Fine-needle biopsy with 19G needle is effective in combination with endoscopic ultrasound-guided tissue acquisition for genomic profiling of unresectable pancreatic cancer. Dig Endosc. 2023;35:124–33.PubMedCrossRef Ikeda G, Hijioka S, Nagashio Y, et al. Fine-needle biopsy with 19G needle is effective in combination with endoscopic ultrasound-guided tissue acquisition for genomic profiling of unresectable pancreatic cancer. Dig Endosc. 2023;35:124–33.PubMedCrossRef
63.
go back to reference Kubota Y, Kawakami H, Natsuizaka M, et al. CTNNB1 mutational analysis of solid-pseudopapillary neoplasms of the pancreas using endoscopic ultrasound-guided fine-needle aspiration and next-generation deep sequencing. J Gastroenterol. 2015;50:203–10.PubMedCrossRef Kubota Y, Kawakami H, Natsuizaka M, et al. CTNNB1 mutational analysis of solid-pseudopapillary neoplasms of the pancreas using endoscopic ultrasound-guided fine-needle aspiration and next-generation deep sequencing. J Gastroenterol. 2015;50:203–10.PubMedCrossRef
64.
go back to reference Kimura R, Ohtsuka T, Kubo M, et al. FoundationOne® CDx gene profiling in Japanese pancreatic ductal adenocarcinoma patients: a single-institution experience. Surg Today. 2021;51:619–26.PubMedCrossRef Kimura R, Ohtsuka T, Kubo M, et al. FoundationOne® CDx gene profiling in Japanese pancreatic ductal adenocarcinoma patients: a single-institution experience. Surg Today. 2021;51:619–26.PubMedCrossRef
65.
go back to reference Sone M, Arai Y, Sugawara S, et al. Feasibility of genomic profiling with next-generation sequencing using specimens obtained by image-guided percutaneous needle biopsy. Ups J Med Sci. 2019;124:119–24.PubMedCrossRefPubMedCentral Sone M, Arai Y, Sugawara S, et al. Feasibility of genomic profiling with next-generation sequencing using specimens obtained by image-guided percutaneous needle biopsy. Ups J Med Sci. 2019;124:119–24.PubMedCrossRefPubMedCentral
66.
go back to reference Facciorusso A, Prete VD, Buccino VR, et al. Diagnostic yield of Franseen and Fork-Tip biopsy needles for endoscopic ultrasound-guided tissue acquisition: a meta-analysis. Endosc Int Open. 2019;7:E1221–30.PubMedCrossRefPubMedCentral Facciorusso A, Prete VD, Buccino VR, et al. Diagnostic yield of Franseen and Fork-Tip biopsy needles for endoscopic ultrasound-guided tissue acquisition: a meta-analysis. Endosc Int Open. 2019;7:E1221–30.PubMedCrossRefPubMedCentral
67.
go back to reference Asokkumar R, Ka CY, Loh T, et al. Comparison of tissue and molecular yield between fine-needle biopsy (FNB) and fine-needle aspiration (FNA): a randomized study. Endosc Int Open. 2019;7:E955–63.PubMedCrossRefPubMedCentral Asokkumar R, Ka CY, Loh T, et al. Comparison of tissue and molecular yield between fine-needle biopsy (FNB) and fine-needle aspiration (FNA): a randomized study. Endosc Int Open. 2019;7:E955–63.PubMedCrossRefPubMedCentral
68.
go back to reference Cancer Genome Atlas Research Network. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell. 2017;32:185–203.CrossRef Cancer Genome Atlas Research Network. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell. 2017;32:185–203.CrossRef
71.
go back to reference Takahashi K, Yasuda I, Hanaoka T, et al. Comparison of histological sample volumes among various endoscopic ultrasound-guided biopsy needles. J Clin Med. 2021;10:3560.PubMedCrossRefPubMedCentral Takahashi K, Yasuda I, Hanaoka T, et al. Comparison of histological sample volumes among various endoscopic ultrasound-guided biopsy needles. J Clin Med. 2021;10:3560.PubMedCrossRefPubMedCentral
72.
go back to reference Gheorghiu M, Seicean A, Bolboacă SD, et al. Endoscopic ultrasound-guided fine-needle biopsy versus fine-needle aspiration in the diagnosis of focal liver lesions: prospective head-to-head comparison. Diagnostics (Basel). 2022;12:2214.PubMedCrossRef Gheorghiu M, Seicean A, Bolboacă SD, et al. Endoscopic ultrasound-guided fine-needle biopsy versus fine-needle aspiration in the diagnosis of focal liver lesions: prospective head-to-head comparison. Diagnostics (Basel). 2022;12:2214.PubMedCrossRef
73.
go back to reference Noh DH, Choi K, Gu S, et al. Comparison of 22-gauge standard fine needle versus core biopsy needle for endoscopic ultrasound-guided sampling of suspected pancreatic cancer: a randomized crossover trial. Scand J Gastroenterol. 2018;53:94–9.PubMedCrossRef Noh DH, Choi K, Gu S, et al. Comparison of 22-gauge standard fine needle versus core biopsy needle for endoscopic ultrasound-guided sampling of suspected pancreatic cancer: a randomized crossover trial. Scand J Gastroenterol. 2018;53:94–9.PubMedCrossRef
74.
go back to reference Alatawi A, Beuvon F, Grabar S, et al. Comparison of 22G reverse-beveled versus standard needle for endoscopic ultrasound-guided sampling of solid pancreatic lesions. United European Gastroenterol J. 2015;3:343–52.PubMedCrossRefPubMedCentral Alatawi A, Beuvon F, Grabar S, et al. Comparison of 22G reverse-beveled versus standard needle for endoscopic ultrasound-guided sampling of solid pancreatic lesions. United European Gastroenterol J. 2015;3:343–52.PubMedCrossRefPubMedCentral
75.
go back to reference Eso Y, Kou T, Nagai H, et al. Utility of ultrasound-guided liver tumor biopsy for next-generation sequencing-based clinical sequencing. Hepatol Res. 2019;49:579–89.PubMedCrossRef Eso Y, Kou T, Nagai H, et al. Utility of ultrasound-guided liver tumor biopsy for next-generation sequencing-based clinical sequencing. Hepatol Res. 2019;49:579–89.PubMedCrossRef
76.
go back to reference Chun SM, Sung CO, Jeon H, et al. Next-generation sequencing using S1 nuclease for poor-quality formalin-fixed paraffin-embedded tumor specimens. J Mol Diagn. 2018;20:802–11.PubMedCrossRef Chun SM, Sung CO, Jeon H, et al. Next-generation sequencing using S1 nuclease for poor-quality formalin-fixed paraffin-embedded tumor specimens. J Mol Diagn. 2018;20:802–11.PubMedCrossRef
77.
go back to reference Pishvaian MJ, Blais ME, Brody JR, et al. Overall survival in patients with pancreatic cancer receiving matched therapies following molecular profiling: a retrospective analysis of the know your tumor registry trial. Lancet Oncol. 2020;21:508–18.PubMedCrossRefPubMedCentral Pishvaian MJ, Blais ME, Brody JR, et al. Overall survival in patients with pancreatic cancer receiving matched therapies following molecular profiling: a retrospective analysis of the know your tumor registry trial. Lancet Oncol. 2020;21:508–18.PubMedCrossRefPubMedCentral
78.
go back to reference Singhi AD, George B, Greenbowe JR, et al. Real-time targeted genome profile analysis of pancreatic ductal adenocarcinomas identifies genetic alterations that might be targeted with existing drugs or used as biomarkers. Gastroenterology. 2019;156:2242-2253.e4.PubMedCrossRef Singhi AD, George B, Greenbowe JR, et al. Real-time targeted genome profile analysis of pancreatic ductal adenocarcinomas identifies genetic alterations that might be targeted with existing drugs or used as biomarkers. Gastroenterology. 2019;156:2242-2253.e4.PubMedCrossRef
79.
go back to reference Hayashi H, Tanishima S, Fujii K, et al. Genomic testing for pancreatic cancer in clinical practice as real-world evidence. Pancreatology. 2018;18:647–54.PubMedCrossRef Hayashi H, Tanishima S, Fujii K, et al. Genomic testing for pancreatic cancer in clinical practice as real-world evidence. Pancreatology. 2018;18:647–54.PubMedCrossRef
80.
82.
go back to reference Umemoto K, Yamamoto H, Oikawa R, et al. The molecular landscape of pancreatobiliary cancers for novel targeted therapies from real-world genomic profiling. J Natl Cancer Inst. 2022;114:1279–86.PubMedCrossRefPubMedCentral Umemoto K, Yamamoto H, Oikawa R, et al. The molecular landscape of pancreatobiliary cancers for novel targeted therapies from real-world genomic profiling. J Natl Cancer Inst. 2022;114:1279–86.PubMedCrossRefPubMedCentral
83.
go back to reference Javle M, Bekaii-Saab T, Jain A, et al. Biliary cancer: utility of next-generation sequencing for clinical management. Cancer. 2016;122:3838–47.PubMedCrossRef Javle M, Bekaii-Saab T, Jain A, et al. Biliary cancer: utility of next-generation sequencing for clinical management. Cancer. 2016;122:3838–47.PubMedCrossRef
85.
go back to reference Nakamura H, Arai Y, Totoki Y, et al. Genomic spectra of biliary tract cancer. Nat Genet. 2015;47:1003–10.PubMedCrossRef Nakamura H, Arai Y, Totoki Y, et al. Genomic spectra of biliary tract cancer. Nat Genet. 2015;47:1003–10.PubMedCrossRef
86.
go back to reference Guo L, Zhou F, Liu H, et al. Genomic mutation characteristics and prognosis of biliary tract cancer. Am J Transl Res. 2022;14:4990–5002.PubMedPubMedCentral Guo L, Zhou F, Liu H, et al. Genomic mutation characteristics and prognosis of biliary tract cancer. Am J Transl Res. 2022;14:4990–5002.PubMedPubMedCentral
87.
go back to reference Silverman IM, Hollebecque A, Friboulet L, et al. Clinicogenomic analysis of FGFR2-rearranged cholangiocarcinoma identifies correlates of response and mechanisms of resistance to pemigatinib. Cancer Discov. 2021;11:326–39.PubMedCrossRef Silverman IM, Hollebecque A, Friboulet L, et al. Clinicogenomic analysis of FGFR2-rearranged cholangiocarcinoma identifies correlates of response and mechanisms of resistance to pemigatinib. Cancer Discov. 2021;11:326–39.PubMedCrossRef
88.
go back to reference Chakrabarti S, Kamgar M, Mahipal A. Targeted therapies in advanced biliary tract cancer: an evolving paradigm. Cancers (Basel). 2020;12:2039.PubMedCrossRef Chakrabarti S, Kamgar M, Mahipal A. Targeted therapies in advanced biliary tract cancer: an evolving paradigm. Cancers (Basel). 2020;12:2039.PubMedCrossRef
89.
90.
go back to reference Lowery MA, Ptashkin R, Jordan E, et al. Comprehensive molecular profiling of intrahepatic and extrahepatic cholangiocarcinomas: potential targets for intervention. Clin Cancer Res. 2018;24:4154–61.PubMedCrossRefPubMedCentral Lowery MA, Ptashkin R, Jordan E, et al. Comprehensive molecular profiling of intrahepatic and extrahepatic cholangiocarcinomas: potential targets for intervention. Clin Cancer Res. 2018;24:4154–61.PubMedCrossRefPubMedCentral
91.
go back to reference Aitcheson G, Mahipal A, John BV. Targeting FGFR in intrahepatic cholangiocarcinoma [iCCA]: leading the way for precision medicine in biliary tract cancer [BTC]? Expert Opin Investig Drugs. 2021;30:463–77.PubMedCrossRef Aitcheson G, Mahipal A, John BV. Targeting FGFR in intrahepatic cholangiocarcinoma [iCCA]: leading the way for precision medicine in biliary tract cancer [BTC]? Expert Opin Investig Drugs. 2021;30:463–77.PubMedCrossRef
92.
go back to reference Prete MG, Cammarota A, D’Alessio A. Current options and future directions of systemic therapy for advanced biliary tract cancer. Explor Target Antitumor Ther. 2021;2:416–33.PubMedCrossRefPubMedCentral Prete MG, Cammarota A, D’Alessio A. Current options and future directions of systemic therapy for advanced biliary tract cancer. Explor Target Antitumor Ther. 2021;2:416–33.PubMedCrossRefPubMedCentral
93.
go back to reference Takada K, Kubo T, Kikuchi J, et al. Effect of comprehensive cancer genomic profiling on therapeutic strategies and clinical outcomes in patients with advanced biliary tract cancer: a prospective multicenter study. Front Oncol. 2022;12: 988527.PubMedCrossRefPubMedCentral Takada K, Kubo T, Kikuchi J, et al. Effect of comprehensive cancer genomic profiling on therapeutic strategies and clinical outcomes in patients with advanced biliary tract cancer: a prospective multicenter study. Front Oncol. 2022;12: 988527.PubMedCrossRefPubMedCentral
94.
go back to reference Silverman IM, Li M, Murugesan K, et al. Validation and characterization of FGFR2 rearrangements in cholangiocarcinoma with comprehensive genomic profiling. J Mol Diagn. 2022;24:351–64.PubMedCrossRef Silverman IM, Li M, Murugesan K, et al. Validation and characterization of FGFR2 rearrangements in cholangiocarcinoma with comprehensive genomic profiling. J Mol Diagn. 2022;24:351–64.PubMedCrossRef
95.
go back to reference Okawa Y, Ebata N, Kim NKD, et al. Actionability evaluation of biliary tract cancer by genome transcriptome analysis and Asian cancer knowledgebase. Oncotarget. 2021;12:1540–52.PubMedCrossRefPubMedCentral Okawa Y, Ebata N, Kim NKD, et al. Actionability evaluation of biliary tract cancer by genome transcriptome analysis and Asian cancer knowledgebase. Oncotarget. 2021;12:1540–52.PubMedCrossRefPubMedCentral
96.
go back to reference Kanai M. Current clinical practice of precision medicine using comprehensive genomic profiling tests in biliary tract cancer in Japan. Curr Oncol. 2022;29:7272–84.PubMedCrossRefPubMedCentral Kanai M. Current clinical practice of precision medicine using comprehensive genomic profiling tests in biliary tract cancer in Japan. Curr Oncol. 2022;29:7272–84.PubMedCrossRefPubMedCentral
97.
go back to reference Moura DTH, Moura EGH, Bernardo WM, et al. Endoscopic retrograde cholangiopancreatography versus endoscopic ultrasound for tissue diagnosis of malignant biliary stricture: systematic review and meta-analysis. Endosc Ultrasound. 2018;7:10–9.PubMedCrossRef Moura DTH, Moura EGH, Bernardo WM, et al. Endoscopic retrograde cholangiopancreatography versus endoscopic ultrasound for tissue diagnosis of malignant biliary stricture: systematic review and meta-analysis. Endosc Ultrasound. 2018;7:10–9.PubMedCrossRef
98.
go back to reference Moura DTH, Moura EGH, Matuguma SE, et al. EUS-FNA versus ERCP for tissue diagnosis of suspect malignant biliary strictures: a prospective comparative study. Endosc Int Open. 2018;6:E769–77.PubMedCrossRefPubMedCentral Moura DTH, Moura EGH, Matuguma SE, et al. EUS-FNA versus ERCP for tissue diagnosis of suspect malignant biliary strictures: a prospective comparative study. Endosc Int Open. 2018;6:E769–77.PubMedCrossRefPubMedCentral
99.
go back to reference Lee YN, Moon JH, Choi HJ, et al. Tissue acquisition for diagnosis of biliary strictures using peroral cholangioscopy or endoscopic ultrasound-guided fine-needle aspiration. Endoscopy. 2019;51:50–9.PubMedCrossRef Lee YN, Moon JH, Choi HJ, et al. Tissue acquisition for diagnosis of biliary strictures using peroral cholangioscopy or endoscopic ultrasound-guided fine-needle aspiration. Endoscopy. 2019;51:50–9.PubMedCrossRef
100.
go back to reference Hirata K, Kuwatani M, Suda G, et al. A novel approach for the genetic analysis of biliary tract cancer specimens obtained through endoscopic ultrasound-guided fine needle aspiration using targeted amplicon sequencing. Clin Transl Gastroenterol. 2019;10: e00022.PubMedCrossRefPubMedCentral Hirata K, Kuwatani M, Suda G, et al. A novel approach for the genetic analysis of biliary tract cancer specimens obtained through endoscopic ultrasound-guided fine needle aspiration using targeted amplicon sequencing. Clin Transl Gastroenterol. 2019;10: e00022.PubMedCrossRefPubMedCentral
101.
go back to reference Kai Y, Ikezawa K, Takada R, et al. Success rate of microsatellite instability examination and complete response with pembrolizumab in biliary tract cancer. JGH Open. 2021;5:712–6.PubMedCrossRefPubMedCentral Kai Y, Ikezawa K, Takada R, et al. Success rate of microsatellite instability examination and complete response with pembrolizumab in biliary tract cancer. JGH Open. 2021;5:712–6.PubMedCrossRefPubMedCentral
102.
go back to reference Maruki Y, Morizane C, Arai Y, et al. Molecular detection and clinicopathological characteristics of advanced/recurrent biliary tract carcinomas harboring the FGFR2 rearrangements: a prospective observational study (PRELUDE Study). J Gastroenterol. 2021;56:250–60.PubMedCrossRef Maruki Y, Morizane C, Arai Y, et al. Molecular detection and clinicopathological characteristics of advanced/recurrent biliary tract carcinomas harboring the FGFR2 rearrangements: a prospective observational study (PRELUDE Study). J Gastroenterol. 2021;56:250–60.PubMedCrossRef
103.
go back to reference Choi HJ, Moon JH, Kim HK, et al. KRAS mutation analysis by next-generation sequencing in endoscopic ultrasound-guided sampling for solid liver masses. J Gastroenterol Hepatol. 2017;32:154–62.PubMedCrossRef Choi HJ, Moon JH, Kim HK, et al. KRAS mutation analysis by next-generation sequencing in endoscopic ultrasound-guided sampling for solid liver masses. J Gastroenterol Hepatol. 2017;32:154–62.PubMedCrossRef
104.
go back to reference Kawakami H, Kuwatani M, Etoh K, et al. Endoscopic retrograde cholangiography versus peroral cholangioscopy to evaluate intraepithelial tumor spread in biliary cancer. Endoscopy. 2009;41:959–64.PubMedCrossRef Kawakami H, Kuwatani M, Etoh K, et al. Endoscopic retrograde cholangiography versus peroral cholangioscopy to evaluate intraepithelial tumor spread in biliary cancer. Endoscopy. 2009;41:959–64.PubMedCrossRef
105.
go back to reference Singhi AD, Nikiforova MN, Chennat J, et al. Integrating next-generation sequencing to endoscopic retrograde cholangiopancreatography (ERCP)-obtained biliary specimens improves the detection and management of patients with malignant bile duct strictures. Gut. 2020;69:52–61.PubMedCrossRef Singhi AD, Nikiforova MN, Chennat J, et al. Integrating next-generation sequencing to endoscopic retrograde cholangiopancreatography (ERCP)-obtained biliary specimens improves the detection and management of patients with malignant bile duct strictures. Gut. 2020;69:52–61.PubMedCrossRef
106.
go back to reference Kuwatani M, Kawakubo K, Sakamoto N, et al. Promising genomic testing for biliary tract cancer using endoscopic ultrasound-guided fine-needle aspiration/biopsy Specimens. Diagnostics (Basel). 2022;12:900.PubMedCrossRef Kuwatani M, Kawakubo K, Sakamoto N, et al. Promising genomic testing for biliary tract cancer using endoscopic ultrasound-guided fine-needle aspiration/biopsy Specimens. Diagnostics (Basel). 2022;12:900.PubMedCrossRef
107.
go back to reference Fujii T, Uchiyama T, Matsuoka M, et al. Evaluation of DNA and RNA quality from archival formalin-fixed paraffin-embedded tissue for next-generation sequencing—retrospective study in Japanese single institution. Pathol Int. 2020;70:602–11.PubMedCrossRef Fujii T, Uchiyama T, Matsuoka M, et al. Evaluation of DNA and RNA quality from archival formalin-fixed paraffin-embedded tissue for next-generation sequencing—retrospective study in Japanese single institution. Pathol Int. 2020;70:602–11.PubMedCrossRef
108.
go back to reference Balla A, Hampel KJ, Sharma MK, et al. Comprehensive validation of cytology specimens for next-generation sequencing and clinical practice experience. J Mol Diagn. 2018;20:812–21.PubMedCrossRef Balla A, Hampel KJ, Sharma MK, et al. Comprehensive validation of cytology specimens for next-generation sequencing and clinical practice experience. J Mol Diagn. 2018;20:812–21.PubMedCrossRef
109.
go back to reference Santos GC, Saieg MA. Preanalytic specimen triage: smears, cell blocks, cytospin preparations, transport media, and cytobanking. Cancer Cytopathol. 2017;125:455–64.CrossRef Santos GC, Saieg MA. Preanalytic specimen triage: smears, cell blocks, cytospin preparations, transport media, and cytobanking. Cancer Cytopathol. 2017;125:455–64.CrossRef
110.
go back to reference Wani S, Muthusamy VR, McGrath CM, et al. AGA white paper: optimizing endoscopic ultrasound-guided tissue acquisition and future directions. Clin Gastroenterol Hepatol. 2018;16:318–27.PubMedCrossRef Wani S, Muthusamy VR, McGrath CM, et al. AGA white paper: optimizing endoscopic ultrasound-guided tissue acquisition and future directions. Clin Gastroenterol Hepatol. 2018;16:318–27.PubMedCrossRef
111.
go back to reference Huggett JF, Cowen S, Foy CA. Considerations for digital PCR as an accurate molecular diagnostic tool. Clin Chem. 2015;61:79–88.PubMedCrossRef Huggett JF, Cowen S, Foy CA. Considerations for digital PCR as an accurate molecular diagnostic tool. Clin Chem. 2015;61:79–88.PubMedCrossRef
112.
go back to reference Matsumoto K, Kato H, Nouso K, et al. Evaluation of local recurrence of pancreatic cancer by KRAS mutation analysis using washes from endoscopic ultrasound-guided fine-needle aspiration. Dig Dis Sci. 2020;65:2907–13.PubMedCrossRef Matsumoto K, Kato H, Nouso K, et al. Evaluation of local recurrence of pancreatic cancer by KRAS mutation analysis using washes from endoscopic ultrasound-guided fine-needle aspiration. Dig Dis Sci. 2020;65:2907–13.PubMedCrossRef
113.
114.
go back to reference Llovet JM, Kelley RK, Villanueva A, et al. Hepatocellular carcinoma. Nat Rev Dis Primers. 2021;7:6.PubMedCrossRef Llovet JM, Kelley RK, Villanueva A, et al. Hepatocellular carcinoma. Nat Rev Dis Primers. 2021;7:6.PubMedCrossRef
115.
go back to reference Nakagawa H, Fujita M, Fujimoto A, et al. Genome sequencing analysis of liver cancer for precision medicine. Semin Cancer Biol. 2019;55:120–7.PubMedCrossRef Nakagawa H, Fujita M, Fujimoto A, et al. Genome sequencing analysis of liver cancer for precision medicine. Semin Cancer Biol. 2019;55:120–7.PubMedCrossRef
117.
go back to reference Takano Y, Noda J, Yamawaki M, et al. Comparative study of an ultrasound-guided percutaneous biopsy and endoscopic ultrasound-guided fine-needle aspiration for liver tumors. Intern Med. 2021;60:1657–64.PubMedCrossRefPubMedCentral Takano Y, Noda J, Yamawaki M, et al. Comparative study of an ultrasound-guided percutaneous biopsy and endoscopic ultrasound-guided fine-needle aspiration for liver tumors. Intern Med. 2021;60:1657–64.PubMedCrossRefPubMedCentral
118.
go back to reference Ozeki Y, Kanogawa N, Ogasawara S, et al. Liver biopsy technique in the era of genomic cancer therapies: a single-center retrospective analysis. Int J Clin Oncol. 2022;27:1459–66.PubMedCrossRef Ozeki Y, Kanogawa N, Ogasawara S, et al. Liver biopsy technique in the era of genomic cancer therapies: a single-center retrospective analysis. Int J Clin Oncol. 2022;27:1459–66.PubMedCrossRef
Metadata
Title
Current status and issues in genomic analysis using EUS-FNA/FNB specimens in hepatobiliary–pancreatic cancers
Authors
Yoshinori Ozono
Hiroshi Kawakami
Naomi Uchiyama
Hiroshi Hatada
Souichiro Ogawa
Publication date
12-09-2023
Publisher
Springer Nature Singapore
Published in
Journal of Gastroenterology / Issue 11/2023
Print ISSN: 0944-1174
Electronic ISSN: 1435-5922
DOI
https://doi.org/10.1007/s00535-023-02037-z

Other articles of this Issue 11/2023

Journal of Gastroenterology 11/2023 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.