Identification of tumorigenic cells and therapeutic targets in pancreatic neuroendocrine tumors

GW Krampitz, BM George… - Proceedings of the …, 2016 - National Acad Sciences
GW Krampitz, BM George, SB Willingham, JP Volkmer, K Weiskopf, N Jahchan
Proceedings of the National Academy of Sciences, 2016National Acad Sciences
Pancreatic neuroendocrine tumors (PanNETs) are a type of pancreatic cancer with limited
therapeutic options. Consequently, most patients with advanced disease die from tumor
progression. Current evidence indicates that a subset of cancer cells is responsible for tumor
development, metastasis, and recurrence, and targeting these tumor-initiating cells is
necessary to eradicate tumors. However, tumor-initiating cells and the biological processes
that promote pathogenesis remain largely uncharacterized in PanNETs. Here we profile …
Pancreatic neuroendocrine tumors (PanNETs) are a type of pancreatic cancer with limited therapeutic options. Consequently, most patients with advanced disease die from tumor progression. Current evidence indicates that a subset of cancer cells is responsible for tumor development, metastasis, and recurrence, and targeting these tumor-initiating cells is necessary to eradicate tumors. However, tumor-initiating cells and the biological processes that promote pathogenesis remain largely uncharacterized in PanNETs. Here we profile primary and metastatic tumors from an index patient and demonstrate that MET proto-oncogene activation is important for tumor growth in PanNET xenograft models. We identify a highly tumorigenic cell population within several independent surgically acquired PanNETs characterized by increased cell-surface protein CD90 expression and aldehyde dehydrogenase A1 (ALDHA1) activity, and provide in vitro and in vivo evidence for their stem-like properties. We performed proteomic profiling of 332 antigens in two cell lines and four primary tumors, and showed that CD47, a cell-surface protein that acts as a “don’t eat me” signal co-opted by cancers to evade innate immune surveillance, is ubiquitously expressed. Moreover, CD47 coexpresses with MET and is enriched in CD90hi cells. Furthermore, blocking CD47 signaling promotes engulfment of tumor cells by macrophages in vitro and inhibits xenograft tumor growth, prevents metastases, and prolongs survival in vivo.
National Acad Sciences