Abstract

Abstract Despite advances in chemotherapeutic options over the last four decades, ovarian cancer remains the most lethal gynecologic malignancy with a cure rate that is essentially unchanged. A major barrier is lack of treatment-naïve, clinically- relevant in vivo models of epithelial ovarian cancer (EOC). To target this problem, we have developed low-passage in vivo models of EOC derived from patient primary debulking procedures at Mayo Clinic (Rochester, MN). Tumors were obtained from consented patients with suspected ovarian cancer, minced and injected into severe combined immunodeficiency (SCID) mice intraperitoneally for model establishment. To the date, we have heterotransplanted 129 EOC or ovarian malignant mixed mullerian (MMMT) tumors. The most common histologic subtype is serous (70.5%), followed by endometrioid (12.4%), clear cell (6.9%), and others (9.9%). The tumors were injected intraperitoneally into severe combined immunodeficiency (SCID) mice and the average time to moribund (TTM) was 174 +/− 83 days. In general, tumors grew faster with serial passages, TTM for epithelial tumors was 87 +/− 15 days and 19.5 +/− 3 days for MMMT. De novo ascites developed in 17 models and 24 models developed bowel metastasis, two clinically-relevant complications in late-stage ovarian cancer patients. Tumorgrafts were histologically similar to their source patient tumors and were positive for expression epithelial markers (pan-cytokeratin). The engraftment rate was 72.4% (n=87) after ≤ 6 months of lead-time, which is the average time to tumorgraft harvest. Passage of well-established tumorgrafts in SCID mice was successful in 88.6 % (n= 70) of all mice but only 56% (n=16) of athymic nude mice, consistent with prior studies demonstrating improved engraftment rates in more immunodeficient mice. Molecular characterization of tumorgrafts by array comparative genomic hybridization revealed similar gains and losses between the patient, early-passage, and later-passage tumorgraft tissues, indicating the tumorgrafts were genetically similar to their primary tumor. Analysis of tumorgrafts using the Affymetrix Human Plus 2.0 arrays revealed a predominance of previously described molecular subtypes C3, C4, and C5. Current investigations with both standard chemotherapy and novel targeted therapies are ongoing. Taken together, these models are as diverse as the patient population from which they are derived, produce clinically-relevant complications of EOC, maintain genetic fidelity in SCID mice, and are useful preclinical models for testing and development of novel therapeutics for ovarian cancer. High rates of engraftment are feasible. These tumorgrafts may help define predictors of response and eventually, lead to better treatments for patients. This work is funded by the Mayo Ovarian SPORE (CA136393) and the Minnesota Ovarian Cancer Alliance. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 3274. doi:1538-7445.AM2012-3274

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