American Association for Cancer Research
Browse
15357163mct181224-sup-211169_3_supp_5501801_pr2qnn.pdf (905.07 kB)

Supplemental Methods and Supplemental Figures S1-S3 from KIT-Dependent and KIT-Independent Genomic Heterogeneity of Resistance in Gastrointestinal Stromal Tumors — TORC1/2 Inhibition as Salvage Strategy

Download (905.07 kB)
journal contribution
posted on 2023-04-03, 16:09 authored by Thomas Mühlenberg, Julia Ketzer, Michael C. Heinrich, Susanne Grunewald, Adrian Marino-Enriquez, Marcel Trautmann, Wolfgang Hartmann, Eva Wardelmann, Jürgen Treckmann, Karl Worm, Stefanie Bertram, Thomas Herold, Hans-Ulrich Schildhaus, Hanno Glimm, Albrecht Stenzinger, Benedikt Brors, Peter Horak, Peter Hohenberger, Stefan Fröhling, Jonathan A. Fletcher, Sebastian Bauer

Supplemental Methods; Supplemental Figure S1: Cancer-related mutations in the DKTK-Master patient cohort; Supplemental Figure S2: T1-PTEN cells were treated with increasing concentrations of approved KIT-inhibitors for 72h and analyzed by SRB assay; Supplemental Figure S3: Dose-combination studies with sapanisertib

Funding

DKTK Joint Funding Program

GIST Cancer Research Fund

VA Merit Review

History

ARTICLE ABSTRACT

Sporadic gastrointestinal stromal tumors (GIST), characterized by activating mutations of KIT or PDGFRA, favorably respond to KIT inhibitory treatment but eventually become resistant. The development of effective salvage treatments is complicated by the heterogeneity of KIT secondary resistance mutations. Recently, additional mutations that independently activate KIT-downstream signaling have been found in pretreated patients—adding further complexity to the scope of resistance. We collected genotyping data for KIT from tumor samples of pretreated GIST, providing a representative overview on the distribution and incidence of secondary KIT mutations (n = 80). Analyzing next-generation sequencing data of 109 GIST, we found that 18% carried mutations in KIT-downstream signaling intermediates (NF1/2, PTEN, RAS, PIK3CA, TSC1/2, AKT, BRAF) potentially mediating resistance to KIT inhibitors. Notably, we found no apparent other driver mutations in refractory cases that were analyzed by whole exome/genome sequencing (13/109). Using CRISPR/Cas9 methods, we generated a panel of GIST cell lines harboring mutations in KIT, PTEN, KRAS, NF1, and TSC2. We utilized this panel to evaluate sapanisertib, a novel mTOR kinase inhibitor, as a salvage strategy. Sapanisertib had potent antiproliferative effects in all cell lines, including those with KIT-downstream mutations. Combinations with KIT or MEK inhibitors completely abrogated GIST-survival signaling and displayed synergistic effects. Our isogenic cell line panel closely approximates the genetic heterogeneity of resistance observed in heavily pretreated patients with GIST. With the clinical development of novel, broad spectrum KIT inhibitors, emergence of non-KIT–related resistance may require combination treatments with inhibitors of KIT-downstream signaling such as mTOR or MEK.