CC BY-NC-ND 4.0 · Endosc Int Open 2018; 06(10): E1278-E1282
DOI: 10.1055/a-0650-4447
Case report
Owner and Copyright © Georg Thieme Verlag KG 2018

EUS fine-needle pancreatic core biopsy can determine eligibility for tumor-agnostic immunotherapy

Ferga C. Gleeson
1   Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota, United States
,
Michael J. Levy
1   Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota, United States
,
Anja C. Roden
2   Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, United States
,
Lisa A. Boardman
1   Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota, United States
,
Frank A. Sinicrope
1   Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota, United States
3   Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, United States
,
Robert R. McWilliams
3   Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, United States
,
Lizhi Zhang
2   Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, United States
› Author Affiliations
Further Information

Publication History

submitted 09 April 2018

accepted after revision 14 May 2018

Publication Date:
08 October 2018 (online)

Abstract

Background and study aims The US FDA recently approved a cancer treatment with pembrolizumab based upon the tumor biomarker status of deficient mismatch repair (dMMR) rather than a specific disease-based approach. We sought to determine if endoscopic ultrasound-guided fine-needle biopsy (EUS-FNB) could determine dMMR and quantification of PD-L1 expression to potentially guide the delivery of tumor agnostic immunotherapy.

Patients and methods Immunohistochemistry was performed on archived pancreas core biopsy specimens. Tumors with absent nuclear staining of DNA mismatch repair proteins represented dMMR. Tumors were considered to have any or high PD-L1 expression, if expressed in ≥ 1 % or ≥ 50 % of tumor cells.

Results Histologic specimen adequacy for MMR status assessment was satisfactory in 97.2 % of tumors. dMMR and high PD-L1 expression was identified in 3 % and 8.1 % of the cohort.

Conclusion In the setting of tumor type agnostic immunotherapy, it is projected that at least 3 % of malignant pancreas lesions will be sensitive to pembrolizumab and up to 8 % sensitive to the family of immune checkpoint inhibitors. This highlights the expanding role of EUS-FNB in the field of precision immuno-oncology.

 
  • References

  • 1 US Food and Drug Administration. FDA approves first cancer treatment for any solid tumor with a specific genetic feature. https://www.fda.gov/newsevents/newsroom/pressannouncements/ucm560167.htm
  • 2 Vanderwalde A, Spetzler D, Xiao N. et al. Microsatellite instability status determined by next-generation sequencing and compared with PD-L1 and tumor mutational burden in 11,348 patients. Cancer Med 2018; 7: 746-756
  • 3 Hu ZI, Shia J, Stadler ZK. et al. Evaluating mismatch repair deficiency in pancreatic adenocarcinoma: challenges and recommendations. Clin Cancer Res 2018; 24: 1326-1336
  • 4 Gleeson FC, Zhang L, Roden AC. et al. EUS fine-needle biopsy can determine PD-L1 immune biomarker status in treatment naïve pancreatic ductal adenocarcinoma. Clin Gastroenterol Hepatol 2018; 3 DOI: 10.1016/j.cgh.2018.01.001.
  • 5 Lee BS, Cho CM, Jung MK. et al. comparison of histologic core portions acquired from a core biopsy needle and a conventional needle in solid mass lesions: a prospective randomized trial. Gut Liver 2017; 15: 559-566
  • 6 Lee YN, Moon JH, Kim HK. et al. Core biopsy needle versus standard aspiration needle for endoscopic ultrasound-guided sampling of solid pancreatic masses: a randomized parallel-group study. Endoscopy 2014; 46: 1056-1062
  • 7 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: 3533-3539
  • 8 Hedenström P, Demir A, Khodakaram K. et al. EUS-guided reverse bevel fine-needle biopsy sampling and open tip fine-needle aspiration in solid pancreatic lesions - a prospective, comparative study. Scand J Gastroenterol 2018; 53: 231-237
  • 9 Strand DS, Jeffus SK, Sauer BG. et al. EUS-guided 22-gauge fine-needle aspiration versus core biopsy needle in the evaluation of solid pancreatic neoplasms. Diagn Cytopathol 2014; 42: 751-758
  • 10 Bang JY, Hebert-Magee S, Trevino J. et al. Randomized trial comparing the 22-gauge aspiration and 22-gauge biopsy needles for EUS-guided sampling of solid pancreatic mass lesions. Gastrointest Endosc 2012; 76: 321-327
  • 11 Iacobuzio-Donahue CA, Ryu B, Hruban RH. et al. Exploring the host desmoplastic response to pancreatic carcinoma: gene expression of stromal and neoplastic cells at the site of primary invasion. Am J Pathol 2002; 160: 91-99
  • 12 Wood LD, Hruban RH. Pathology and molecular genetics of pancreatic neoplasms. Cancer J 2012; 18: 492-501
  • 13 Roy-Chowdhuri S, Chen H, Singh RR. et al. Concurrent fine needle aspirations and core needle biopsies: a comparative study of substrates for next-generation sequencing in solid organ malignancies. Mod Pathol 2017; 30: 499-508
  • 14 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: 318-327
  • 15 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: 1474-1480
  • 16 Goldstein JB, Wu W, Borras E. et al. Can microsatellite status of colorectal cancer be reliably assessed after neoadjuvant therapy?. Clin Cancer Res 2017; 1: 5246-5254
  • 17 Pelekanou V, Carvajal-Hausdorf DE, Altan M. et al. Effect of neoadjuvant chemotherapy on tumor-infiltrating lymphocytes and PD-L1 expression in breast cancer and its clinical significance. Breast Cancer Res 2017; 7: 91