Identification of an optimal circulating tumor DNA (ctDNA) shedding threshold to detect actionable driver mutations in colorectal and pancreatic adenocarcinoma.

Authors

Bennett Caughey

Bennett Adam Caughey

Division of Medical Oncology, Duke University School of Medicine, Durham, NC

Bennett Adam Caughey , Kumiko Umemoto , Michelle Green , Masafumi Ikeda , Rachel D'Anna , Makoto Ueno , Donna Niedzwiecki , Hiroya Taniguchi , Daniel Walden , Yoshito Komatsu , Katherine I. Zhou , Taito Esaki , Ryne Ramaker , Tadamichi Denda , Michael Datto , Hideaki Bando , Tanios S. Bekaii-Saab , Takayuki Yoshino , John H Strickler , Yoshiaki Nakamura

Organizations

Division of Medical Oncology, Duke University School of Medicine, Durham, NC, Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan, Department of Pathology, Duke University School of Medicine, Durham, NC, National Cancer Center Hospital East, Kashiwa, Japan, Duke University, Durham, NC, Kanagawa Cancer Center, Yokohama, Japan, Alliance Statistics and Data Management Center and Department of Biostatistics and Bioinformations, Duke University, Durham, NC, Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan, Mayo Clinic Arizona, Phoenix, AZ, Hokkaido University Hospital, Sapporo, Japan, University of Chicago, Chicago, IL, Department of Gastrointestinal and Medical Oncology, NHO Kyushu Cancer Center, Fukuoka, Japan, Division of Gastroenterology, Chiba Cancer Center, Chiba, Japan, Aichi Cancer Center Hospital, Aichi, Japan, Division of Hematology/Oncology, Mayo Clinic, Phoenix, AZ

Research Funding

Other
SCRUM-Japan Funds

Background: In colorectal cancer (CRC), mutations in KRAS, NRAS, and BRAF predict resistance to anti-EGFR therapies. In pancreatic ductal adenocarcinoma (PDAC), ~90% of patients harbor KRAS mutations, while KRAS wild-type tumors often have clinically actionable fusion alterations. Thus, in both cancers, the accurate ascertainment of RAS and BRAF driver status is essential. Sequencing of cell-free DNA (cfDNA) from plasma allows convenient assessment of a tumor’s molecular profile, but sensitivity can be limited by low ctDNA shedding. We sought to establish a ctDNA shedding threshold at which actionable driver mutations can be reliably detected. Methods: Molecular reports and matched clinical data were obtained from the Duke Molecular Registry of Tumors and the SCRUM-Japan GOZILA and GI-SCREEN. CRC or PDAC patients with a pathogenic KRAS, NRAS, or BRAF activating point mutation (“driver”) present on tissue next-generation sequencing (NGS) assays and who also had cfDNA assay available were included. Tissue NGS included Foundation One CDx and Oncomine Comprehensive Assay. Guardant 360 (G360) was the sole plasma cfDNA assay. 131 CRC and 24 PDAC cases with 189 total G360 assays met criteria and were included. Samples were analyzed according to detection of the driver mutation and the maximum mutant allele frequency (MAF) of non-driver mutations on G360. An optimal cut-point for max MAF was explored among the CRC and PDAC patients using a maximally selected Wilcoxon rank statistic method. Results: 76.8% of driver mutations were in KRAS, 22.6% in BRAF, and 1.9% in NRAS with an overlap of 1 BRAF and 1 NRAS mutation with a KRAS mutation. Overall sensitivity of G360 for drivers was 83.0% for CRC and 54.2% for PDAC. No variants were detected on G360 in 9.1% of CRC and 37.5% of PDAC. Sensitivity for driver mutations increased with higher maximum non-driver MAF, with MAF > 1% predicting sensitivity > 98% (Table). Optimal cut-point analysis identified MAF of > 0.34% (p < 0.0001), above which the driver was identified in 97% of patients and below which only 27%. Conclusions: In our study, non-driver MAF > 1% on cfDNA NGS predicts high sensitivity for RAS and BRAF mutations and thus is adequate to guide clinical decisions such as anti-EGFR therapy in CRC, evaluation for fusions in PDAC, and validity in clinical trials. MAF ≤0.34% is a clear threshold to consider an assay inadequate and thus seek alternative testing. Sensitivity rises for MAF between 0.35 and 1% but will require greater patient numbers to establish clinically relevant sensitivity thresholds. These results will be updated with additional data for final presentation.

Non-driver max MAF range
Driver detection sensitivity (95% CI)
N
0% (no variants)
7.7% (0.95-25.1)
26
> 0 to ≤0.5%
64.5% (45.4-80.77)
31
> 0.5 to ≤1%
85.7% (57.2-98.2)
14
> 1 to ≤5%
100% (86.8-100)
26
> 5 to ≤10%
95.8% (78.9-99.9)
24
> 10%
98.5% (92.1-99.96)
68

Disclaimer

This material on this page is ©2024 American Society of Clinical Oncology, all rights reserved. Licensing available upon request. For more information, please contact licensing@asco.org

Abstract Details

Meeting

2022 ASCO Annual Meeting

Session Type

Poster Session

Session Title

Gastrointestinal Cancer—Colorectal and Anal

Track

Gastrointestinal Cancer—Colorectal and Anal

Sub Track

Colorectal Cancer–Advanced Disease

Citation

J Clin Oncol 40, 2022 (suppl 16; abstr 3571)

DOI

10.1200/JCO.2022.40.16_suppl.3571

Abstract #

3571

Poster Bd #

365

Abstract Disclosures

Similar Abstracts

Abstract

2024 ASCO Gastrointestinal Cancers Symposium

Association of candidate alterations with primary resistance to KRAS G12D targeting in colorectal cancer.

First Author: Khalid Jazieh

Abstract

2023 ASCO Gastrointestinal Cancers Symposium

Mutation status of full RAS and BRAF in 169 Moroccan patients with colorectal cancer.

First Author: Sara El Zaitouni

Abstract

2023 ASCO Gastrointestinal Cancers Symposium

Circulating tumor DNA–based genomic landscape of KRAS wild-type pancreatic adenocarcinoma.

First Author: Brendon Fusco

Abstract

2023 ASCO Annual Meeting

Frequency of KRAS, NRAS, and BRAF mutations in colorectal cancer in an Argentinian population.

First Author: Agustín Barbier