Genomic correlates of response to capecitabine and temozolomide (CAPTEM) in pancreatic neuroendocrine tumors.

Authors

null

Patrick Lee

Cedars-Sinai Medical Center, Los Angeles, CA

Patrick Lee , Edik Matthew Blais , Jun Gong , Arsen Osipov , Natalie Moshayedi , Shant Thomassian , Camille Ng , Jennifer W. Chuy , Lynn McCormick Matrisian , Emanuel Petricoin III, Michael J. Pishvaian , Andrew Eugene Hendifar

Organizations

Cedars-Sinai Medical Center, Los Angeles, CA, University of Virginia Health System, Charlottesville, VA, City of Hope, Duarte, CA, Johns Hopkins University School of Medicine, Department of Oncology, Balimore, MD, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, Pancreatic Cancer Action Network, Manhattan Beach, CA, Perthera, Holliston, MA, Georgetown University Medical Center, Washington, DC, Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA

Research Funding

Other

Background: Despite the frequent use of capecitabine and temozolomide (CAPTEM) to treat metastatic, well-differentiated pancreatic neuroendocrine tumors (PNETs), no reliable genomic predictors of response currently exist. PNETs commonly harbor mutations in MEN1, ATRX, DAXX, and the PI3K/AKT/mTOR pathway. We sought to determine whether the mutational status of these genes correlates with response to CAPTEM. Methods: A retrospective cohort of PNET cases seen at Cedars-Sinai Medical Center or from Perthera’s Real-World Evidence Database included 23 patients who were treated with CAPTEM in 1st or 2nd line and had targeted next-generation sequencing (NGS) of their tumors available. Genomic alterations were correlated with progression-free survival (PFS) using multivariate Cox regression analysis. Results: We analyzed 23 PNET patients, 4 (17.4%) of whom had documented functional tumors. We identified MEN1 mutations as positively associated with CAPTEM response, but this effect was less pronounced for the subset with co-occurring DAXX mutations, which are commonly found alongside MEN1 alterations. With and without accounting for line of therapy, we found that PFS on CAPTEM was significantly longer in MEN1-mutated, DAXX-wildtype tumors compared to other mutation profiles (P< 0.01, see Table). ATRX (67%) and PTEN (33%) alterations were also enriched in the MEN1-mutated/DAXX-wildtype subset; however, other PI3K/AKT/mTOR alterations were common across all MEN1-mutated cases. Conclusions: We describe a novel genomic signature (MEN1 mut/DAXX wt) that correlates with PNET response to CAPTEM therapy and is exploratory in nature. Prospective validation of these associations is warranted while taking into account other therapies, histopathologic factors, and other genomic correlates.

Exploratory PFS analyses on 1st/2nd line CAPTEM comparing MEN1-mutated/DAXX-wildtype PNETs versus other genomic profiles.

PNET Subset
mPFS [95% CI]
Univariate
p-value (HR [95% CI])
Multivariate
p-value (HR [95% CI])
Line of Therapy
p-value (HR [95% CI])
MEN1-mutated
&DAXX-wildtype (n=9)
16.3m [12.6-N/R]
0.009932
(0.16 [0.04-0.64])
0.009722
(0.14 [0.03-0.63])
0.6245 (0.75 [0.24-2.33])
MEN1-wildtype
or DAXX-mutated (n=14)
7.4m [7.1-N/R]

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—Gastroesophageal, Pancreatic, and Hepatobiliary

Track

Gastrointestinal Cancer—Gastroesophageal, Pancreatic, and Hepatobiliary

Sub Track

Neuroendocrine/Carcinoid

Citation

J Clin Oncol 40, 2022 (suppl 16; abstr 4124)

DOI

10.1200/JCO.2022.40.16_suppl.4124

Abstract #

4124

Poster Bd #

110

Abstract Disclosures