Abstract

Abstract Epithelial ovarian cancer (EOC) remains the leading cause of death from gynecological malignancy and the fifth leading cause of all cancer-related deaths among American women. The most common and lethal subtype, high-grade serous ovarian cancer (HGSOC) tends to originate from the fallopian tube. Precursor lesions of HGSOC such as secretory cell expansion (SCE), the loss of PAX2, PTEN, p53 mutation and serous tubal intraepithelial carcinoma (STIC) are often found at the fallopian tube epithelium (FTE). However, the mechanism of how neoplastic lesions disseminate in FTE and eventually colonize the ovary is unclear. We are interested in understanding if ovarian cancer initiation in the FTE is mediated by extracellular vesicles (EVs), which are nanosized biovesicles secreted by all living cells. EVs serve as vehicles for nucleic acids, proteins, and lipids. Cancer cells commonly secrete more EVs than healthy cells and shift their EV cargos to promote tumorigenesis. The alteration of molecular content of tumor-derived EV can potentially indicate disease states and serve as a new class of cancer biomarkers. Hence, our hypothesis is that EV cargos secreted by tumor cells contain factors promoting preneoplastic lesions in the FTE. Our goal is to interrogate the role of EVs in the initiation of ovarian cancer in the FTE at the early stage, for which there is no adequate in vitro model. We created a microfluidic model using a 3D dynamic culture system termed PREDICT-MOS to enable the direct interaction of hFTE with EVs and the long-term culture of the fallopian tube explant. We are interested to 1) test the role of EVs to drive preneoplastic transformation of hFTE progenitor cells and 2) to test whether tumor derived-EVs shift EV proteins cargos secreted by hFTE. The hFTE cultured on PREDICT-MOS is directly exposed to OVCAR3-derived EVs and embedded for transcriptomic profiling utilizing GeoMx Digital Spatial Profiler to identify differentially expressed genes in both ciliated and secretory cells within the epithelium. In parallel, we are characterizing the hFTE EV protein content with or without OVCAR3 EV treatment to detect the alteration in hFTE cargos using mass spectrometry proteomics. Our preliminary transcriptomic data from 8 patient fallopian tube samples demonstrates the deregulation of pathways associated with inflammation (TNF and NF-kappa B signaling), immune suppression (PD-1 checkpoint signaling) and epithelium morphogenesis. In addition, we have identified 295 proteins as the results of the first proteomic characterization of hFTE-derived EV proteins, which can serve as a “baseline” to evaluate whether the fallopian tube shifts its sEV cargo during ovarian cancer carcinogenesis. Further validation of transcriptomic findings on translational level using immunohistochemistry and identification of OVCAR3 EV protein cargos that induce the transcriptomic changes are needed to establish the role of EVs in the initiation of HGSOC in the fallopian tube. This project is supported by the Ovarian Cancer Research Fund Alliance Program Grant. Citation Format: Didi Zha, Jared Sipes, Sagar Rayamajhi, Leonidas E. Bantis, Angela Russo, Harsh Pathak, Andrew K. Godwin, Joanna E. Burdette. Modeling the malignant transformation of fallopian tube epithelium driven by extracellular vesicles cargos in an organ-on-chip microphysiological system [abstract]. In: Proceedings of the AACR Special Conference on Ovarian Cancer; 2023 Oct 5-7; Boston, Massachusetts. Philadelphia (PA): AACR; Cancer Res 2024;84(5 Suppl_2):Abstract nr B041.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call