Oncotarget

Research Papers:

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

Ferga C. Gleeson _, Sarah E. Kerr, Benjamin R. Kipp, Jesse S. Voss, Douglas M. Minot, Zheng Jin Tu, Michael R. Henry, Rondell P. Graham, George Vasmatzis, John C. Cheville, Konstantinos N. Lazaridis and Michael J. Levy

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2016; 7:54526-54536. https://doi.org/10.18632/oncotarget.9440

Metrics: PDF 2132 views  |   HTML 3195 views  |   ?  


Abstract

Ferga C. Gleeson1, Sarah E. Kerr2, Benjamin R. Kipp2, Jesse S. Voss2, Douglas M. Minot2, Zheng Jin Tu3, Michael R. Henry2, Rondell P. Graham2, George Vasmatzis4, John C. Cheville4, Konstantinos N. Lazaridis1,4, Michael J. Levy1

1Division of Gastroenterology & Hepatology, Mayo Clinic Rochester, MN, USA

2Department of Laboratory Medicine & Pathology, Mayo Clinic Rochester, MN, USA

3Division of Biomedical Statics & Informatics, Department of Health Sciences Research, Mayo Clinic Rochester, MN, USA

4Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA

Correspondence to:

Ferga C. Gleeson, email: [email protected]

Keywords: endoscopic ultrasound fine needle aspiration, pancreatic adenocarcinoma, targeted next-generation sequencing, mutation concordance, personalized medicine

Received: April 04, 2016     Accepted: April 24, 2016     Published: May 18, 2016

ABSTRACT

Background & Aims: Less than 10% of registered drug intervention trials for pancreatic ductal adenocarcinoma (PDAC) include a biomarker stratification strategy. The ability to identify distinct mutation subsets via endoscopic ultrasound fine needle aspiration (EUS FNA) molecular cytology could greatly aid clinical trial patient stratification and offer predictive markers. We identified chemotherapy treatment naïve ampullary adenocarcinoma and PDAC patients who underwent EUS FNA to assess multigene mutational frequency and diversity with a surgical resection concordance assessment, where available.

Methods: Following strict cytology smear screening criteria, targeted next generation sequencing (NGS) using a 160 cancer gene panel was performed.

Results: Complete sequencing was achieved in 29 patients, whereby 83 pathogenic alterations were identified in 21 genes. Cytology genotyping revealed that the majority of mutations were identified in KRAS (93%), TP53 (72%), SMAD4 (31%), and GNAS (10%). There was 100% concordance for the following pathogenic alterations: KRAS, TP53, SMAD4, KMT2D, NOTCH2, MSH2, RB1, SMARCA4, PPP2R1A, PIK3R1, SCL7A8, ATM, and FANCD2. Absolute multigene mutational concordance was 83%. Incremental cytology smear mutations in GRIN2A, GATA3 and KDM6A were identified despite re-examination of raw sequence reads in the corresponding resection specimens.

Conclusions: EUS FNA cytology genotyping using a 160 cancer gene NGS panel revealed a broad spectrum of pathogenic alterations. The fidelity of cytology genotyping to that of paired surgical resection specimens suggests that EUS FNA represents a suitable surrogate and may complement the conventional stratification criteria in decision making for therapies and may guide future biomarker driven therapeutic development.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 9440