Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Courtney L. Andersen is active.

Publication


Featured researches published by Courtney L. Andersen.


Endocrine-related Cancer | 2012

Hormone response in ovarian cancer: time to reconsider as a clinical target?

Francesmary Modugno; R. Laskey; A. Smith; Courtney L. Andersen; Paul Haluska; Steffi Oesterreich

Ovarian cancer is the sixth most common cancer worldwide among women in developed countries and the most lethal of all gynecologic malignancies. There is a critical need for the introduction of targeted therapies to improve outcome. Epidemiological evidence suggests a critical role for steroid hormones in ovarian tumorigenesis. There is also increasing evidence from in vitro studies that estrogen, progestin, and androgen regulate proliferation and invasion of epithelial ovarian cancer cells. Limited clinical trials have shown modest response rates; however, they have consistently identified a small subset of patients that respond very well to endocrine therapy with few side effects. We propose that it is timely to perform additional well-designed trials that should include biomarkers of response.


Clinical Cancer Research | 2017

Active estrogen receptor-alpha signaling in ovarian cancer models and clinical specimens

Courtney L. Andersen; Matthew J. Sikora; M.M. Boisen; Tianzhou Ma; Alec Christie; George C. Tseng; Yongseok Park; Soumya Luthra; Uma Chandran; Paul Haluska; Gina Mantia-Smaldone; Kunle Odunsi; Karen McLean; Adrian V. Lee; Esther Elishaev; Robert P. Edwards; Steffi Oesterreich

Purpose: High-grade serous ovarian cancer (HGSOC) is an aggressive disease with few available targeted therapies. Despite high expression of estrogen receptor-alpha (ERα) in approximately 80% of HGSOC and some small but promising clinical trials of endocrine therapy, ERα has been understudied as a target in this disease. We sought to identify hormone-responsive, ERα-dependent HGSOC. Experimental Design: We characterized endocrine response in HGSOC cells across culture conditions [ two-dimensional (2D), three-dimensional (3D), forced suspension] and in patient-derived xenograft (PDX) explants, assessing proliferation and gene expression. Estrogen-regulated transcriptome data were overlapped with public datasets to develop a comprehensive panel of ERα target genes. Expression of this panel and ERα H-score were assessed in HGSOC samples from patients who received endocrine therapy. Time on endocrine therapy was used as a surrogate for clinical response. Results: Proliferation is ERα-regulated in HGSOC cells in vitro and in vivo, and is partly dependent on 3D context. Transcriptomic studies identified genes shared by cell lines and PDX explants as ERα targets. The selective ERα downregulator (SERD) fulvestrant is more effective than tamoxifen in blocking ERα action. ERα H-score is predictive of efficacy of endocrine therapy, and this prediction is further improved by inclusion of target gene expression, particularly IGFBP3. Conclusions: Laboratory models corroborate intertumor heterogeneity of endocrine response in HGSOC but identify features associated with functional ERα and endocrine responsiveness. Assessing ERα function (e.g., IGFBP3 expression) in conjunction with H-score may help select patients who would benefit from endocrine therapy. Preclinical data suggest that SERDs might be more effective than tamoxifen. Clin Cancer Res; 23(14); 3802–12. ©2017 AACR.


Molecular and Cellular Endocrinology | 2015

Treating gynecologic malignancies with selective estrogen receptor downregulators (SERDs): promise and challenges.

M.M. Boisen; Courtney L. Andersen; Sreeja Sreekumar; Steffi Oesterreich

Endometrial and ovarian cancers are estrogen-dependent gynecologic malignancies. Although many are estrogen receptor (ER) positive, treatment with the selective estrogen receptor modulator (SERM) tamoxifen, a tissue selective partial-agonist, has demonstrated only modest clinical benefit. Selective estrogen receptor downregulators (SERDs) are pure ER antagonists showing a benefit for advanced ER positive breast cancer, which has bolstered their potential use for ER positive gynecologic malignancies. We summarize these preclinical and clinical data, suggesting that a subpopulation of patients with endometrial or ovarian cancer exists in which treatment with SERDs results in improved outcome. However, the full potential of SERDs for a gynecologic malignancies will be realized only when the appropriate predictive biomarkers are identified. Additionally, a further understanding ER signaling in the context of ovarian and endometrial tissues that appear to involve c-Src and other kinase pathways is needed to successfully address the emergence of resistance with rationally designed combination therapies.


Gynecologic Oncology | 2016

High expression of orphan nuclear receptor NR4A1 in a subset of ovarian tumors with worse outcome

Evan Delgado; M.M. Boisen; R. Laskey; Rui Chen; Chi Song; Jad Sallit; Zachary A. Yochum; Courtney L. Andersen; Matthew J. Sikora; Jacob Wagner; Stephen Safe; Esther Elishaev; Adrian V. Lee; Robert P. Edwards; Paul Haluska; George C. Tseng; Mark E. Schurdak; Steffi Oesterreich

OBJECTIVE Nuclear receptors (NRs) play a vital role in the development and progression of several cancers including breast and prostate. Using TCGA data, we sought to identify critical nuclear receptors in high grade serous ovarian cancers (HGSOC) and to confirm these findings using in vitro approaches. METHODS In silico analysis of TCGA data was performed to identify relevant NRs in HGSOC. Ovarian cancer cell lines were screened for NR expression and functional studies were performed to determine the significance of these NRs in ovarian cancers. NR expression was analyzed in ovarian cancer tissue samples using immunohistochemistry to identify correlations with histology and stage of disease. RESULTS The NR4A family of NRs was identified as a potential driver of ovarian cancer pathogenesis. Overexpression of NR4A1 in particular correlated with worse progression free survival. Endogenous expression of NR4A1 in normal ovarian samples was relatively high compared to that of other tissue types, suggesting a unique role for this orphan receptor in the ovary. Expression of NR4A1 in HGSOC cell lines as well as in patient samples was variable. NR4A1 primarily localized to the nucleus in normal ovarian tissue while co-localization within the cytoplasm and nucleus was noted in ovarian cancer cell lines and patient tissues. CONCLUSIONS NR4A1 is highly expressed in a subset of HGSOC samples from patients that have a worse progression free survival. Studies to target NR4A1 for therapeutic intervention should include HGSOC.


Hormones and Cancer | 2018

The Evolution of Estrogen Receptor Signaling in the Progression of Endometriosis to Endometriosis-Associated Ovarian Cancer

Courtney L. Andersen; M.M. Boisen; Matthew J. Sikora; Tianzhou Ma; George C. Tseng; Swati Suryawanshi; Anda M. Vlad; Esther Elishaev; Robert P. Edwards; Steffi Oesterreich

To investigate changes in estrogen receptor alpha (ERα) signaling during progression of endometriosis to endometriosis-associated ovarian cancer (EAOC) as a driver of malignant transformation. We procured tissue samples of normal endometrium, endometriosis (benign, atypical, concurrent with EAOC), and EAOC. We evaluated expression of a 236-gene signature of estrogen signaling. ANOVA and unsupervised clustering were used to identify gene expression profiles across disease states. These profiles were compared to profiles of estrogen regulation in cancer models from the Gene Expression Omnibus (GEO). Gene Set Enrichment Analysis (GSEA) was performed to determine whether gene expression in EAOC was consistent with ERα activity. ANOVA revealed 158 differentially expressed genes (q < 0.05) and unsupervised clustering identified five distinct gene clusters. The estrogen signaling profile of EAOC was not consistent with activated ERα in pre-clinical models. Gene set enrichment analysis did not identify signatures of activated ERα in EAOC but instead identified expression patterns consistent with loss of ERα function and development of endocrine resistance. Gene expression data suggest that ERα signaling becomes inactivated throughout the progression of endometriosis to EAOC. The gene expression pattern in EAOC is more consistent with profiles of endocrine resistance.


Cancer Research | 2016

Abstract 862: WNT4 mediates endocrine response and resistance in invasive lobular carcinoma cell lines and patient tumor explants

Matthew J. Sikora; Courtney L. Andersen; Caroline M. Alexander; Priscilla M. McAuliffe; Steffi Oesterreich

Invasive lobular carcinoma (ILC) is a breast cancer subtype affecting ∼30,000 U.S. women annually. Over 90% of ILC are estrogen receptor (ER)-positive; however, endocrine therapy may have poorer efficacy in a subset of ILC patients versus invasive ductal carcinoma (IDC) patients. This prompted us to assess global ER activity in ILC cell lines MDA MB 134VI (MM134) and SUM44PE (44PE) to identify novel mediators of ER signaling. These analyses identified the Wnt ligand WNT4 as an ILC-specific ER target gene, with an ILC-specific ER binding site (ERBS) at the WNT4 locus. Considering the critical role of WNT4 in normal mammary gland expansion, we hypothesize that ILC cells utilize WNT4 signaling to drive endocrine response and resistance. We assessed whether WNT4 is necessary for ILC cell growth using siRNA. WNT4 knockdown completely blocked estrogen-induced growth in ILC cells but not IDC cells. In parallel, the WNT4 ERBS was only occupied in ILC cells in response to estrogen, but progesterone-induced WNT4 in IDC was not associated with this ERBS. This suggests that, via the ILC-specific WNT4 ERBS, ILC cells drive estrogen-regulated proliferation by hijacking a developmental Wnt pathway. Wnt pathways typically activate â-catenin; however, we observed â-catenin dysfunction in ILC cells and that WNT4 cannot activate â-catenin. Thus, WNT4 signals in ILC cells via a novel non-canonical pathway. Using long-term estrogen-deprived (LTED) variants of MM134 and 44PE (4 and 2 lines, respectively), we assessed WNT4 in ILC endocrine resistance. WNT4 is over-expressed, but uncoupled from ER, in all MM134-LTED. Conversely, WNT4 is reduced in 44PE-LTED but remains ER-regulated; ER occupies the WNT4 ERBS only in 44PE-LTED cells and not MM134-LTED. Using siRNA, MM134-LTED (high WNT4) are growth-inhibited by WNT4 knockdown, while 44PE-LTED (low WNT4) are insensitive. However, WNT4 knockdown sensitizes 44PE-LTED to endocrine therapy. Taken together, uncoupling and upregulating WNT4 or WNT4/ER cross-talk may represent convergent endocrine resistance mechanisms in ILC. To assess the role of WNT4 in patient ILC, we examined WNT4 protein in archival breast tumors and observed that WNT4 is frequently expressed in ILC and IDC tumors. We also examined WNT4 regulation and endocrine response in patient tumor explants. We observed ER regulation of WNT4 directly in ILC tissues that correlated with sensitivity to fulvestrant but resistance to tamoxifen; this may mimic clinical endocrine resistance. Ongoing studies are assessing WNT4 as a biomarker and mediator of endocrine resistance in ILC. Clinical observations suggest that ER regulates unique pathways in ILC. We identified WNT4 as a downstream effector of endocrine signaling in ILC, with critical roles in both estrogen-induced growth and endocrine resistance. WNT4 signaling may represent a novel target to modulate endocrine response specifically for ILC patients. Citation Format: Matthew J. Sikora, Courtney L. Andersen, Caroline M. Alexander, Priscilla M. McAuliffe, Steffi Oesterreich. WNT4 mediates endocrine response and resistance in invasive lobular carcinoma cell lines and patient tumor explants. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 862.


Cancer Research | 2014

Abstract 2103: Utilizing ER target genes as biomarkers of endocrine response in serous ovarian carcinoma

Courtney L. Andersen; Matthew J. Sikora; Soumya Luthra; Uma Chandran; Paul Haluska; Steffi Oesterreich

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Ovarian cancer is the most common and most lethal gynecologic malignancy. Despite clear histologic and molecular differences between disease subtypes, all patients receive cytotoxic chemotherapy and survival has not improved significantly for several decades. Identifying avenues for targeted therapy is critical to improving clinical outcomes. Given the vast heterogeneity of ovarian cancer, therapeutic strategies will vary based on subtype and specific biomarkers. Estrogen receptor-alpha (ER) is expressed in ∼70% of serous ovarian tumors and epidemiologic evidence indicates estrogen contributes to the disease etiology. Further, clinical data suggest that a subset of patients with serous carcinoma benefit from endocrine therapy. However, the full potential of endocrine therapy in ovarian cancer has been understudied. We hypothesize that a subset of serous ovarian tumors are dependent upon estrogen and that expression of ER target genes will identify patients who can be successfully treated with endocrine therapy. We characterized response to estradiol (E2) in four ER+ serous ovarian cancer cell lines, focusing on cell proliferation, survival, and gene expression. Proliferation was evaluated in 2-D and in a 3-D matrigel system. Survival was assayed in low-attachment conditions. We also evaluated response to anti-estrogens 4-hydroxytamoxifen (4OHT) and fulvestrant (ICI). Expression of canonical ER targets (e.g. GREB1, IGFBP3) was measured by qPCR. Gene expression microarrays after E2 treatment were used to identify genome-wide effects of ER. Studies of endocrine response in vivo are ongoing in patient-derived xenograft models. E2 promotes growth of PEO1 and PEO4 cells in 2-D and 3-D culture, with PEO4 cells exhibiting greater E2 dependence. Preliminary results indicate that E2 also promotes survival in low-attachment conditions. Growth and survival are both decreased by treatment with fulvestrant or 4-hydroxytamoxifen. Conversely, OVCA432 and OVSAHO cells do not respond to treatment with ER ligands. Microarrays identified E2-regulated genes in PEO1 and PEO4 cells that may be utilized as biomarkers of ER activity. Several of these ER targets are differentially expressed between E2-responsive and E2-independent cell lines including DEPTOR, GREB1, GFRA2, and EPHB3. We are currently overlapping our array results with publicly available datasets to develop a gene signature of E2 response, which we will use to profile ER signaling in clinical specimens and patient derived xenografts. Our results suggest that a subset of serous ovarian cancers are endocrine responsive. These studies will determine if expression of specific ER targets correlates with endocrine response and thus if they can be harnessed to predict clinical response to endocrine therapy. Citation Format: Courtney L. Andersen, Matthew J. Sikora, Soumya Luthra, Uma Chandran, Paul Haluska, Steffi Oesterreich. Utilizing ER target genes as biomarkers of endocrine response in serous ovarian carcinoma. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2103. doi:10.1158/1538-7445.AM2014-2103


Cancer Research | 2013

Abstract 94: Identifying biomarkers of estrogen response in models of serous ovarian cancer.

Courtney L. Andersen; Matthew J. Sikora; Paul Haluska; Steffi Oesterreich

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Ovarian cancer is the fifth leading cause of cancer death among women in the United States. Despite advances in treatment for other cancers, ovarian cancer patient prognosis has not improved significantly since the advent of platinum-based chemotherapy. It is a highly heterogeneous disease with at least four distinct histological subtypes: serous, mucinous, endometrioid, and clear cell. However, no subtype- or biomarker-specific therapies are approved for ovarian cancer. Identifying targeted therapies is crucial to improving clinical outcomes. Specific therapies will likely vary based on tumor histology and biomarkers. Expression of estrogen receptor alpha (ER) is a well established biomarker of response to endocrine therapies in breast cancer. Despite the success of anti-estrogen therapies in treating breast cancer, little attention has been given to their potential in ovarian cancer. However, epidemiologic studies show that approximately 70% of epithelial ovarian tumors express ER, particularly serous and endometrioid subtypes, and that estrogen plays a role in ovarian cancer etiology. Further, clinical data suggest that a subset of ovarian cancer patients can be successfully treated with endocrine therapy. We hypothesize that some ovarian tumors require estrogen for growth and that predictive biomarkers will identify patients who will benefit from endocrine therapy. Using cell line models, we examined the response of ovarian cancer cells to estrogen (E2) and anti-estrogens with regard to gene expression and cell proliferation. E2 induces expression of canonical ER target genes (e.g. GREB1) in these cells, suggesting that they are estrogen-responsive. We observe that 4-hydroxytamoxifen and fulvestrant inhibit growth of ovarian cancer cells including the chemo-resistant line PEO4. Gene expression microarray studies identified additional E2-regulated genes in ovarian cancer cells that may be utilized as biomarkers of ER activity. We are expanding our studies to in vivo models (human primary tumor xenografts) to evaluate the role of E2 in driving tumor progression and determine the efficacy of anti-estrogens in treating serous ovarian cancer. There is a critical need for targeted therapies in ovarian cancer. Our studies will determine if endocrine responsiveness correlates with gene expression of specific biomarkers, which may lead to the identification of predictive clinical markers of response to endocrine therapy. Citation Format: Courtney L. Andersen, Matthew J. Sikora, Paul Haluska, Steffi Oesterreich. Identifying biomarkers of estrogen response in models of serous ovarian cancer. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 94. doi:10.1158/1538-7445.AM2013-94


Drug Response and Resistance to Therapy | 2018

Abstract B23: The evolution of estrogen receptor signaling in the progression of endometriosis to endometriosis-associated ovarian cancer

M.M. Boisen; Courtney L. Andersen; Matt Sikora; Tianzhou Ma; George Tseng; Anda M. Vlad; Esther Elishaev; Uma Chandran; Robert A. Edwards; Steffi Oesterreich


Drug Response and Resistance to Therapy | 2018

Abstract B25: Hormone receptor expression and response in ovarian tumors of low malignant potential

Sarah Taylor; Courtney L. Andersen; Rohit Bhargava; George Tseng; Steffi Oesterreich; Robert P. Edwards; Adrian V. Lee

Collaboration


Dive into the Courtney L. Andersen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

M.M. Boisen

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Uma Chandran

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Adrian V. Lee

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Anda M. Vlad

University of Pittsburgh

View shared research outputs
Researchain Logo
Decentralizing Knowledge