60 results on '"Norris JD"'
Search Results
2. Androgen receptor monomers and dimers regulate opposing biological processes in prostate cancer cells.
- Author
-
Safi R, Wardell SE, Watkinson P, Qin X, Lee M, Park S, Krebs T, Dolan EL, Blattler A, Tsuji T, Nayak S, Khater M, Fontanillo C, Newlin MA, Kirkland ML, Xie Y, Long H, Fink EC, Fanning SW, Runyon S, Brown M, Xu S, Owzar K, Norris JD, and McDonnell DP
- Subjects
- Male, Humans, Cell Line, Tumor, Gene Expression Regulation, Neoplastic drug effects, Proto-Oncogene Proteins c-myc metabolism, Proto-Oncogene Proteins c-myc genetics, Protein Multimerization drug effects, Animals, Receptors, Androgen metabolism, Receptors, Androgen genetics, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Prostatic Neoplasms genetics, Prostatic Neoplasms drug therapy, Cell Proliferation drug effects, Androgens metabolism, Androgens pharmacology, Signal Transduction drug effects, TOR Serine-Threonine Kinases metabolism
- Abstract
Most prostate cancers express the androgen receptor (AR), and tumor growth and progression are facilitated by exceptionally low levels of systemic or intratumorally produced androgens. Thus, absolute inhibition of the androgen signaling axis remains the goal of current therapeutic approaches to treat prostate cancer (PCa). Paradoxically, high dose androgens also exhibit considerable efficacy as a treatment modality in patients with late-stage metastatic PCa. Here we show that low levels of androgens, functioning through an AR monomer, facilitate a non-genomic activation of the mTOR signaling pathway to drive proliferation. Conversely, high dose androgens facilitate the formation of AR dimers/oligomers to suppress c-MYC expression, inhibit proliferation and drive a transcriptional program associated with a differentiated phenotype. These findings highlight the inherent liabilities in current approaches used to inhibit AR action in PCa and are instructive as to strategies that can be used to develop new therapeutics for this disease and other androgenopathies., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
3. The Impact of Circulating Tumor Cell HOXB13 RNA Detection in Men with Metastatic Castration-Resistant Prostate Cancer (mCRPC) Treated with Abiraterone or Enzalutamide.
- Author
-
Halabi S, Guo S, Park JJ, Nanus DM, George DJ, Antonarakis ES, Danila DC, Szmulewitz RZ, McDonnell DP, Norris JD, Lu C, Luo J, and Armstrong AJ
- Subjects
- Male, Humans, RNA, Prospective Studies, Retrospective Studies, DNA, Complementary therapeutic use, Receptors, Androgen genetics, Receptors, Androgen metabolism, Nitriles therapeutic use, Biomarkers, Tumor genetics, Biomarkers, Tumor therapeutic use, Homeodomain Proteins genetics, Neoplastic Cells, Circulating pathology, Prostatic Neoplasms, Castration-Resistant drug therapy, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant pathology, Androstenes, Benzamides, Phenylthiohydantoin
- Abstract
Purpose: HOXB13 is an androgen receptor (AR) coregulator specifically expressed in cells of prostatic lineage. We sought to associate circulating tumor cell (CTC) HOXB13 expression with outcomes in men with mCRPC treated with abiraterone or enzalutamide., Experimental Design: We conducted a retrospective analysis of the multicenter prospective PROPHECY trial of mCRPC men (NCT02269982, n = 118) treated with abiraterone/enzalutamide. CTC detection and HOXB13 complementary DNA (cDNA) expression was measured using a modified Adnatest, grouping patients into 3 categories: CTC 0 (undetectable); CTC+ HOXB13 CTC low (<4 copies); or CTC+ HOXB13 CTC high. The HOXB13 threshold was determined by maximally selected rank statistics for prognostic associations with overall survival (OS) and progression-free survival (PFS)., Results: We included 102 men with sufficient CTC HOXB13 cDNA, identifying 25%, 31%, and 44% of patients who were CTC 0, CTC+ HOXB13 low, and CTC+ HOXB13 high, respectively. Median OS were 25.7, 27.8, and 12.1 months whereas the median PFS were 9.0, 7.7, and 3.8 months, respectively. In subgroup analysis among men with CellSearch CTCs ≥5 copies/mL and adjusting for prior abi/enza treatment and Halabi clinical risk score, the multivariate HR for HOXB13 CTC detection was 2.39 (95% CI, 1.06-5.40) for OS and 2.78 (95% CI, 1.38-5.59) for PFS, respectively. Low HOXB13 CTC detection was associated with lower CTC PSA, PSMA, AR-FL, and AR-V7 detection, and more liver/lung metastases (41% vs. 25%)., Conclusions: Higher CTC HOXB13 expression is associated with AR-dependent biomarkers in CTCs and is adversely prognostic in the context of potent AR inhibition in men with mCRPC., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
4. Application Status Among Women Enrolled in a Healthy Start Program in Arkansas for the Special Nutrition Program for Women and Children.
- Author
-
Reece S, McElfish PA, Andersen JA, Ayers BL, Tiwari T, Willis DE, Rowland B, Norris JD, Beasley K, Mendoza Kabua P, and Brown CC
- Subjects
- Infant, Humans, Female, Child, Pregnancy, Arkansas, Cross-Sectional Studies, Nutritional Status, Pregnant Women, Health Promotion, Food Assistance
- Abstract
This study aimed to examine the demographic characteristics of pregnant women in a Healthy Start program who are presumed eligible for the Special Supplemental Nutrition Program for Women, Infants, and Children (WIC), but who have not yet applied for WIC benefits. We used a cross sectional evaluation of data collected from pregnant women (n=203) participating in a Healthy Start program. Data came from surveys administered at enrollment in the Healthy Start program from July 15th, 2019 until January 14th, 2022. The primary outcome was WIC application status, which was determined by whether the woman had applied or was receiving benefits at the time of enrollment. Covariates included race/ethnicity, marital status, insurance, education, income, age, employment, and having previous children/pregnancies. Fisher exact tests and logistic regression were used to examine associations. Approximately 65% of women had not yet applied for WIC benefits. Marshallese women (80.9%) and other NHPI women (80.0%) had the highest need for assistance. In adjusted analyses, White women (p = 0.040) and Hispanic women (p = 0.005) had lower rates of needing assistance applying for WIC than Marshallese women. There were higher rates of needing assistance in applying for women with private insurance or with no insurance and for those with higher incomes. Nearly two out of every three pregnant women who were eligible for WIC had not yet applied for benefits. The findings highlight the need for outreach for all populations that may be eligible, particularly among racial/ethnic minorities and those with higher incomes., (© 2023. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
5. A New Chemotype of Chemically Tractable Nonsteroidal Estrogens Based on a Thieno[2,3- d ]pyrimidine Core.
- Author
-
Sammeta VR, Norris JD, Artham S, Torrice CD, Byemerwa J, Joiner C, Fanning SW, McDonnell DP, and Willson TM
- Abstract
Despite continued interest in the development of nonsteroidal estrogens and antiestrogens, there are only a few chemotypes of estrogen receptor ligands. Using targeted screening in a ligand sensing assay, we identified a phenolic thieno[2,3- d ]pyrimidine with affinity for estrogen receptor α. An efficient three-step synthesis of the heterocyclic core and structure-guided optimization of the substituents resulted in a series of potent nonsteroidal estrogens. The chemical tractability of the thieno[2,3- d ]pyrimidine chemotype will support the design of new estrogen receptor ligands as therapeutic hormones and antihormones., Competing Interests: The authors declare no competing financial interest., (© 2022 American Chemical Society.)
- Published
- 2022
- Full Text
- View/download PDF
6. Next-Generation Endocrine Therapies for Breast Cancer.
- Author
-
McDonnell DP, Wardell SE, Chang CY, and Norris JD
- Subjects
- Estrogen Receptor alpha genetics, Female, Humans, Mutation, Selective Estrogen Receptor Modulators therapeutic use, Antineoplastic Agents, Hormonal therapeutic use, Breast Neoplasms drug therapy
- Abstract
Competing Interests: Donald P. McDonnellEmployment: Duke UniversityStock and Other Ownership Interests: Zentalis, G1 Therapeutics, Viba Therapeutics, Rappta Therapeutics, X-RAD TherapeuticsHonoraria: NovartisConsulting or Advisory Role: Zentalis, G1 therapeutics, Bristol-Myers Squibb, Rappta TherapeuticsResearch Funding: Bristol-Myers Squibb, Novartis, ZentalisPatents, Royalties, Other Intellectual Property: Inventor on two patents (assigned to Duke) licensed to Radius Health covering the use of Rad1901 for Breast cancer. I am an inventor on two patents (assigned to Duke) that covers the use of lasofoxifene for ESR1-mutant breast cancers. Licensed to SermonixTravel, Accommodations, Expenses: Bristol-Myers Squibb Suzanne E. WardellConsulting or Advisory Role: ZentalisResearch Funding: Zentalis, Bristol-Myers SquibbPatents, Royalties, Other Intellectual Property: I am listed as an inventor on a patent for the use of RAD1901 in metastatic breast cancer Ching-Yi ChangResearch Funding: Novartis Institutes for BioMedical Research, Bristol-Myers SquibbPatents, Royalties, Other Intellectual Property: Sermonix. Patent application for the use of lasofoxifene as treatment for breast cancer John D. NorrisConsulting or Advisory Role: G1 Pharmaceuticals, CelgeneResearch Funding: G1 Therapeutics, CelgeneNo other potential conflicts of interest were reported.
- Published
- 2021
- Full Text
- View/download PDF
7. The Dysregulated Pharmacology of Clinically Relevant ESR1 Mutants is Normalized by Ligand-activated WT Receptor.
- Author
-
Andreano KJ, Baker JG, Park S, Safi R, Artham S, Oesterreich S, Jeselsohn R, Brown M, Sammons S, Wardell SE, Chang CY, Norris JD, and McDonnell DP
- Subjects
- Breast Neoplasms genetics, Breast Neoplasms metabolism, Breast Neoplasms pathology, Female, Humans, Ligands, Protein Binding, Tumor Cells, Cultured, Antineoplastic Agents, Hormonal pharmacology, Breast Neoplasms drug therapy, Estrogen Receptor alpha genetics, Estrogen Receptor alpha metabolism, Mutation, Selective Estrogen Receptor Modulators pharmacology
- Abstract
The estrogen receptor (ER/ ESR1 ) is expressed in a majority of breast cancers and drugs that inhibit ER signaling are the cornerstone of breast cancer pharmacotherapy. Currently, aromatase inhibitors are the frontline endocrine interventions of choice although their durability in metastatic disease is limited by activating point mutations within the ligand-binding domain of ESR1 that permit ligand-independent activation of the receptor. It has been suggested that the most commonly occurring ESR1 mutations would likely compromise the clinical activity of selective estrogen receptor downregulators and selective estrogen receptor modulators (SERMs) when used as second-line therapies. It was unclear, however, how these mutations, which are likely coexpressed in cells with ER
WT , may impact response to ER ligands in a clinically meaningful manner. To address this issue, we dissected the molecular mechanism(s) underlying ESR1 -mutant pharmacology in models relevant to metastatic disease. These studies revealed that the response of ESR1 mutations to ligands was dictated primarily by the relative coexpression of ERWT in cells. Specifically, dysregulated pharmacology was only evident in cells in which the mutants were overexpressed relative to ligand-activated ERWT ; a finding that highlights the role of allelism in determining ER-mutant pharmacology. Importantly, we demonstrated that the antagonist activity of the SERM, lasofoxifene, was not impacted by mutant status; a finding that has led to its clinical evaluation as a treatment for patients with advanced ER-positive breast cancer whose tumors harbor ESR1 mutations., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
8. G1T48, an oral selective estrogen receptor degrader, and the CDK4/6 inhibitor lerociclib inhibit tumor growth in animal models of endocrine-resistant breast cancer.
- Author
-
Andreano KJ, Wardell SE, Baker JG, Desautels TK, Baldi R, Chao CA, Heetderks KA, Bae Y, Xiong R, Tonetti DA, Gutgesell LM, Zhao J, Sorrentino JA, Thompson DA, Bisi JE, Strum JC, Thatcher GRJ, and Norris JD
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols pharmacology, Apoptosis drug effects, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Proliferation drug effects, Drug Resistance, Neoplasm, Estrogen Antagonists pharmacology, Female, Humans, Mice, Neoplasms, Hormone-Dependent metabolism, Neoplasms, Hormone-Dependent pathology, Protein Kinase Inhibitors pharmacology, Receptors, Estrogen metabolism, Tamoxifen pharmacology, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antibodies, Monoclonal, Humanized pharmacology, Breast Neoplasms drug therapy, Cyclin-Dependent Kinase 4 antagonists & inhibitors, Cyclin-Dependent Kinase 6 antagonists & inhibitors, HIV Antibodies pharmacology, Neoplasms, Hormone-Dependent drug therapy, Selective Estrogen Receptor Modulators pharmacology
- Abstract
Purpose: The combination of targeting the CDK4/6 and estrogen receptor (ER) signaling pathways with palbociclib and fulvestrant is a proven therapeutic strategy for the treatment of ER-positive breast cancer. However, the poor physicochemical properties of fulvestrant require monthly intramuscular injections to patients, which limit the pharmacokinetic and pharmacodynamic activity of the compound. Therefore, an orally available compound that more rapidly reaches steady state may lead to a better clinical response in patients. Here, we report the identification of G1T48, a novel orally bioavailable, non-steroidal small molecule antagonist of ER., Methods: The pharmacological effects and the antineoplastic mechanism of action of G1T48 on tumors was evaluated using human breast cancer cells (in vitro) and xenograft efficacy models (in vivo)., Results: G1T48 is a potent and efficacious inhibitor of estrogen-mediated transcription and proliferation in ER-positive breast cancer cells, similar to the pure antiestrogen fulvestrant. In addition, G1T48 can effectively suppress ER activity in multiple models of endocrine therapy resistance including those harboring ER mutations and growth factor activation. In vivo, G1T48 has robust antitumor activity in a model of estrogen-dependent breast cancer (MCF7) and significantly inhibited the growth of tamoxifen-resistant (TamR), long-term estrogen-deprived (LTED) and patient-derived xenograft tumors with an increased response being observed with the combination of G1T48 and the CDK4/6 inhibitor lerociclib., Conclusions: These data show that G1T48 has the potential to be an efficacious oral antineoplastic agent in ER-positive breast cancer.
- Published
- 2020
- Full Text
- View/download PDF
9. Miniaturization optimized weapon killing power during the social stress of late pre-contact North America (AD 600-1600).
- Author
-
Mika A, Flood K, Norris JD, Wilson M, Key A, Buchanan B, Redmond B, Pargeter J, Bebber MR, and Eren MI
- Subjects
- Archaeology, History, Ancient, Humans, Indians, North American psychology, North America, Population Growth, Warfare psychology, Indians, North American history, Miniaturization, Sociological Factors, Warfare history, Weapons history
- Abstract
Before Europeans arrived to Eastern North America, prehistoric, indigenous peoples experienced a number of changes that culminated in the development of sedentary, maize agricultural lifeways of varying complexity. Inherent to these lifeways were several triggers of social stress including population nucleation and increase, intergroup conflict (warfare), and increased territoriality. Here, we examine whether this period of social stress co-varied with deadlier weaponry, specifically, the design of the most commonly found prehistoric archery component in late pre-contact North America: triangular stone arrow tips (TSAT). The examination of modern metal or carbon projectiles, arrows, and arrowheads has demonstrated that smaller arrow tips penetrate deeper into a target than do larger ones. We first experimentally confirm that this relationship applies to arrow tips made from stone hafted onto shafts made from wood. We then statistically assess a large sample (n = 742) of late pre-contact TSAT and show that these specimens are extraordinarily small. Thus, by miniaturizing their arrow tips, prehistoric people in Eastern North America optimized their projectile weaponry for maximum penetration and killing power in warfare and hunting. Finally, we verify that these functional advantages were selected across environmental and cultural boundaries. Thus, while we cannot and should not rule out stochastic, production economizing, or non-adaptive cultural processes as an explanation for TSAT, overall our results are consistent with the hypothesis that broad, socially stressful demographic changes in late pre-contact Eastern North America resulted in the miniaturization-and augmented lethality-of stone tools across the region., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
10. Correction to: Pharmacokinetic and pharmacodynamic analysis of fulvestrant in preclinical models of breast cancer to assess the importance of its estrogen receptor-α degrader activity in antitumor efficacy.
- Author
-
Wardell SE, Yllanes AP, Chao CA, Bae Y, Andreano KJ, Desautels TK, Heetderks KA, Blitzer JT, Norris JD, and McDonnell DP
- Abstract
The article Pharmacokinetic and pharmacodynamic analysis of fulvestrant in preclinical models of breast cancer to assess the importance of its estrogen receptor-α degrader activity in antitumor efficacy, written by Suzanne E. Wardell, Alexander P. Yllanes, Christina A. Chao, Yeeun Bae, Kaitlyn J. Andreano, Taylor K. Desautels, Kendall A. Heetderks, Jeremy T. Blitzer, John D. Norris, Donald P. McDonnell, was originally published electronically on the publisher's internet portal on September 27, 2019 without open access. With the author(s)' decision to opt for Open Choice the copyright of the article changed on November 16, 2019 to © The Author(s) 2019 and the article is forthwith distributed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The original article has been corrected.
- Published
- 2020
- Full Text
- View/download PDF
11. Pharmacokinetic and pharmacodynamic analysis of fulvestrant in preclinical models of breast cancer to assess the importance of its estrogen receptor-α degrader activity in antitumor efficacy.
- Author
-
Wardell SE, Yllanes AP, Chao CA, Bae Y, Andreano KJ, Desautels TK, Heetderks KA, Blitzer JT, Norris JD, and McDonnell DP
- Subjects
- Administration, Oral, Animals, Antineoplastic Agents, Hormonal administration & dosage, Dose-Response Relationship, Drug, Estrogen Receptor Antagonists administration & dosage, Estrogen Receptor alpha antagonists & inhibitors, Female, Fulvestrant administration & dosage, Mice, Xenograft Model Antitumor Assays, Antineoplastic Agents, Hormonal pharmacokinetics, Breast Neoplasms drug therapy, Estrogen Receptor Antagonists pharmacokinetics, Fulvestrant pharmacokinetics
- Abstract
Purpose: Fulvestrant is a selective estrogen receptor downregulator (SERD) that is approved for first- or second-line use as a single agent or in combination with cyclin dependent kinase or phosphatidylinositol 3-kinase inhibitors for the treatment of metastatic breast cancer. Fulvestrant exhibits exceptionally effective antitumor activity in preclinical models of breast cancer, a success that has been attributed to its robust SERD activity despite modest receptor downregulation in patient tumors. By modeling human exposures in animal models we probe the absolute need for SERD activity., Methods: Three xenograft models of endocrine therapy-resistant breast cancer were used to evaluate the efficacy of fulvestrant administered in doses historically used in preclinical studies in the field or by using a dose regimen intended to model clinical exposure levels. Pharmacokinetic and pharmacodynamic analyses were conducted to evaluate plasma exposure and intratumoral ER downregulation., Results: A clinically relevant 25 mg/kg dose of fulvestrant exhibited antitumor efficacy comparable to the historically used 200 mg/kg dose, but at this lower dose it did not result in robust ER downregulation. Further, the antitumor efficacy of the lower dose of fulvestrant was comparable to that observed for other oral SERDs currently in development., Conclusion: The use of clinically unachievable exposure levels of fulvestrant as a benchmark in preclinical development of SERDs may negatively impact the selection of those molecules that are advanced for clinical development. Further, these studies suggest that antagonist efficacy, as opposed to SERD activity, is likely to be the primary driver of clinical response.
- Published
- 2020
- Full Text
- View/download PDF
12. The Lineage Determining Factor GRHL2 Collaborates with FOXA1 to Establish a Targetable Pathway in Endocrine Therapy-Resistant Breast Cancer.
- Author
-
Cocce KJ, Jasper JS, Desautels TK, Everett L, Wardell S, Westerling T, Baldi R, Wright TM, Tavares K, Yllanes A, Bae Y, Blitzer JT, Logsdon C, Rakiec DP, Ruddy DA, Jiang T, Broadwater G, Hyslop T, Hall A, Laine M, Phung L, Greene GL, Martin LA, Pancholi S, Dowsett M, Detre S, Marks JR, Crawford GE, Brown M, Norris JD, Chang CY, and McDonnell DP
- Subjects
- Animals, Antibodies, Neutralizing immunology, Antibodies, Neutralizing therapeutic use, Cell Adhesion Molecules immunology, Cell Adhesion Molecules metabolism, Drug Resistance, Neoplasm, Estrogen Receptor alpha genetics, Female, GPI-Linked Proteins immunology, GPI-Linked Proteins metabolism, Humans, MCF-7 Cells, Mammary Neoplasms, Experimental drug therapy, Mammary Neoplasms, Experimental genetics, Mice, Mucoproteins immunology, Mucoproteins metabolism, Oncogene Proteins immunology, Oncogene Proteins metabolism, Hepatocyte Nuclear Factor 3-alpha metabolism, Mammary Neoplasms, Experimental metabolism, Transcription Factors metabolism
- Abstract
Notwithstanding the positive clinical impact of endocrine therapies in estrogen receptor-alpha (ERα)-positive breast cancer, de novo and acquired resistance limits the therapeutic lifespan of existing drugs. Taking the position that resistance is nearly inevitable, we undertook a study to identify and exploit targetable vulnerabilities that were manifest in endocrine therapy-resistant disease. Using cellular and mouse models of endocrine therapy-sensitive and endocrine therapy-resistant breast cancer, together with contemporary discovery platforms, we identified a targetable pathway that is composed of the transcription factors FOXA1 and GRHL2, a coregulated target gene, the membrane receptor LYPD3, and the LYPD3 ligand, AGR2. Inhibition of the activity of this pathway using blocking antibodies directed against LYPD3 or AGR2 inhibits the growth of endocrine therapy-resistant tumors in mice, providing the rationale for near-term clinical development of humanized antibodies directed against these proteins., (Copyright © 2019 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
13. HOXB13 interaction with MEIS1 modifies proliferation and gene expression in prostate cancer.
- Author
-
Johng D, Torga G, Ewing CM, Jin K, Norris JD, McDonnell DP, and Isaacs WB
- Subjects
- Cell Line, Tumor, Cell Proliferation, Gene Expression, Gene Expression Profiling, Gene Knockdown Techniques, Germ-Line Mutation, Homeodomain Proteins genetics, Humans, Male, Myeloid Ecotropic Viral Integration Site 1 Protein genetics, RNA, Messenger analysis, Homeodomain Proteins metabolism, Myeloid Ecotropic Viral Integration Site 1 Protein metabolism, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology
- Abstract
Background: The recurrent p.Gly84Glu germline mutation (G84E) in HOXB13 is consistently associated with prostate cancer (PCa), although the mechanisms underlying such linkage remain elusive. The majority of the PCa-associated HOXB13 mutations identified are localized to two conserved domains in HOXB13 that have been shown to mediate the interaction with MEIS cofactors belonging to the TALE family of homeodomain transcription factors. In this study, we sought to interrogate the biochemical and functional interactions between HOXB13 and MEIS in prostatic cells with a goal of defining how the HOXB13-MEIS complex impacts PCa pathobiology and define the extent to which the oncogenic activity of G84E is related to its effect on HOXB13-MEIS interaction/function., Methods: HOXB13 and MEIS paralog expression in prostate epithelial cells and PCa cell lines was characterized by qPCR and immunoblot analyses. HOXB13 and MEIS1 co-expression in human prostate tissue was confirmed by IHC, followed by co-IP mapping of HOXB13-MEIS1 interactions. Proliferation of the PCa cell line LAPC4 following shRNA-mediated knockdown of each gene or both genes was assessed using DNA- and metabolic-based assays. Transcriptional targets of HOXB13 and MEIS1 were identified by gene expression profiling and qPCR. Finally, protein stability of HOXB13 in the context of MEIS1 was determined using pulse-chase assays., Results: HOXB13 and MEIS1 are co-expressed and interact in prostate cells. Both of the putative MEIS interacting domains (MID) within HOXB13 were shown to be capable of mediating the interaction between HOXB13 and MEIS1 independently and such interactions were not influenced by the G84E mutation. The inhibitory effect of either HOXB13 or MEIS1 knockdown on cellular proliferation was augmented by knockdown of both genes, and MEIS1 knockdown abolished HOXB13-driven regulation of BCHE and TNFSF10 mRNA expression. Notably, we demonstrated that MEIS1 stabilized the HOXB13 protein in LAPC4 cells., Conclusions: Our study provides evidence for functional HOXB13-MEIS1 interactions in PCa. MEIS1 may contribute to the cancer-promoting actions of HOXB13 in cellular proliferation and gene regulation by prolonging HOXB13 half-life. Our data demonstrates that G84E is not a loss-of-function mutation that interferes with HOXB13 stability or ability to interact with MEIS1., (© 2018 Wiley Periodicals, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
14. Targeting mutant estrogen receptors.
- Author
-
Wardell SE, Norris JD, and McDonnell DP
- Subjects
- Breast Neoplasms drug therapy, Breast Neoplasms genetics, Female, Humans, Mutation genetics, Selective Estrogen Receptor Modulators pharmacology, Selective Estrogen Receptor Modulators therapeutic use, Mutant Proteins metabolism, Receptors, Estrogen genetics
- Abstract
A drug used in hormone replacement therapy can target estrogen receptors that have become resistant to breast cancer treatments., Competing Interests: SW, JN, DM No competing interests declared, (© 2019, Wardell et al.)
- Published
- 2019
- Full Text
- View/download PDF
15. Discovery of LSZ102, a Potent, Orally Bioavailable Selective Estrogen Receptor Degrader (SERD) for the Treatment of Estrogen Receptor Positive Breast Cancer.
- Author
-
Tria GS, Abrams T, Baird J, Burks HE, Firestone B, Gaither LA, Hamann LG, He G, Kirby CA, Kim S, Lombardo F, Macchi KJ, McDonnell DP, Mishina Y, Norris JD, Nunez J, Springer C, Sun Y, Thomsen NM, Wang C, Wang J, Yu B, Tiong-Yip CL, and Peukert S
- Subjects
- Animals, Antineoplastic Agents pharmacokinetics, Biological Availability, Drug Design, Drug Discovery, Female, Humans, MCF-7 Cells, Mice, Mice, Nude, Rats, Rats, Sprague-Dawley, Rats, Wistar, Selective Estrogen Receptor Modulators pharmacokinetics, Thiophenes chemistry, Thiophenes pharmacokinetics, Xenograft Model Antitumor Assays, Antineoplastic Agents chemical synthesis, Antineoplastic Agents pharmacology, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Estrogen Receptor alpha drug effects, Selective Estrogen Receptor Modulators chemical synthesis, Selective Estrogen Receptor Modulators pharmacology, Thiophenes chemical synthesis, Thiophenes pharmacology
- Abstract
In breast cancer, estrogen receptor alpha (ERα) positive cancer accounts for approximately 74% of all diagnoses, and in these settings, it is a primary driver of cell proliferation. Treatment of ERα positive breast cancer has long relied on endocrine therapies such as selective estrogen receptor modulators, aromatase inhibitors, and selective estrogen receptor degraders (SERDs). The steroid-based anti-estrogen fulvestrant (5), the only approved SERD, is effective in patients who have not previously been treated with endocrine therapy as well as in patients who have progressed after receiving other endocrine therapies. Its efficacy, however, may be limited due to its poor physicochemical properties. We describe the design and synthesis of a series of potent benzothiophene-containing compounds that exhibit oral bioavailability and preclinical activity as SERDs. This article culminates in the identification of LSZ102 (10), a compound in clinical development for the treatment of ERα positive breast cancer.
- Published
- 2018
- Full Text
- View/download PDF
16. Neomorphic ERα Mutations Drive Progression in Breast Cancer and Present a Challenge for New Drug Discovery.
- Author
-
McDonnell DP, Norris JD, and Chang CY
- Subjects
- Breast Neoplasms genetics, Cell Line, Tumor, Drug Discovery, Gene Expression Regulation, Neoplastic, Humans, Mutation, Drug Resistance, Neoplasm, Estrogen Receptor alpha genetics
- Abstract
In this issue of Cancer Cell, Jeselsohn et al. dissect the function of several of the most clinically important estrogen receptor alpha mutants associated with endocrine therapy resistance in breast cancer and demonstrate that they manifest disease-relevant neomorphic activities that likely contribute to tumor pathogenesis., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
17. CDK4/6 Therapeutic Intervention and Viable Alternative to Taxanes in CRPC.
- Author
-
Stice JP, Wardell SE, Norris JD, Yllanes AP, Alley HM, Haney VO, White HS, Safi R, Winter PS, Cocce KJ, Kishton RJ, Lawrence SA, Strum JC, and McDonnell DP
- Subjects
- Animals, Antineoplastic Agents pharmacokinetics, Cell Line, Tumor, Drug Resistance, Neoplasm drug effects, Humans, Male, Mice, Nude, Molecular Targeted Therapy methods, Prostatic Neoplasms, Castration-Resistant pathology, Protein Kinase Inhibitors pharmacokinetics, Protein Kinase Inhibitors pharmacology, Taxoids pharmacology, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Cyclin-Dependent Kinase 4 antagonists & inhibitors, Cyclin-Dependent Kinase 6 antagonists & inhibitors, Prostatic Neoplasms, Castration-Resistant drug therapy
- Abstract
Resistance to second-generation androgen receptor (AR) antagonists and CYP17 inhibitors in patients with castration-resistant prostate cancer (CRPC) develops rapidly through reactivation of the androgen signaling axis and has been attributed to AR overexpression, production of constitutively active AR splice variants, or the selection for AR mutants with altered ligand-binding specificity. It has been established that androgens induce cell-cycle progression, in part, through upregulation of cyclin D1 (CCND1) expression and subsequent activation of cyclin-dependent kinases 4 and 6 (CDK4/6). Thus, the efficacy of the newly described CDK4/6 inhibitors (G1T28 and G1T38), docetaxel and enzalutamide, was evaluated as single agents in clinically relevant in vitro and in vivo models of hormone-sensitive and treatment-resistant prostate cancer. CDK4/6 inhibition (CDK4/6i) was as effective as docetaxel in animal models of treatment-resistant CRPC but exhibited significantly less toxicity. The in vivo effects were durable and importantly were observed in prostate cancer cells expressing wild-type AR, AR mutants, and those that have lost AR expression. CDK4/6i was also effective in prostate tumor models expressing the AR-V7 variant or the AR F876L mutation, both of which are associated with treatment resistance. Furthermore, CDK4/6i was effective in prostate cancer models where AR expression was lost. It is concluded that CDK4/6 inhibitors are a viable alternative to taxanes as therapeutic interventions in endocrine therapy-refractory CRPC. Implications: The preclinical efficacy of CDK4/6 monotherapy observed here suggests the need for near-term clinical studies of these agents in advanced prostate cancer. Mol Cancer Res; 15(6); 660-9. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
18. Androgen receptor antagonism drives cytochrome P450 17A1 inhibitor efficacy in prostate cancer.
- Author
-
Norris JD, Ellison SJ, Baker JG, Stagg DB, Wardell SE, Park S, Alley HM, Baldi RM, Yllanes A, Andreano KJ, Stice JP, Lawrence SA, Eisner JR, Price DK, Moore WR, Figg WD, and McDonnell DP
- Subjects
- Active Transport, Cell Nucleus, Animals, Benzamides, Cell Line, Tumor, Cell Proliferation drug effects, Drug Resistance, Neoplasm, Drug Synergism, HEK293 Cells, Humans, Inhibitory Concentration 50, Male, Metribolone pharmacology, Mice, Inbred NOD, Mice, SCID, Nitriles, Phenylthiohydantoin analogs & derivatives, Phenylthiohydantoin pharmacology, Protein Binding, Receptors, Androgen metabolism, Steroid 17-alpha-Hydroxylase metabolism, Testosterone pharmacology, Transcriptional Activation drug effects, Xenograft Model Antitumor Assays, Androgen Receptor Antagonists pharmacology, Antineoplastic Agents, Hormonal pharmacology, Prostatic Neoplasms, Castration-Resistant drug therapy, Steroid 17-alpha-Hydroxylase antagonists & inhibitors
- Abstract
The clinical utility of inhibiting cytochrome P450 17A1 (CYP17), a cytochrome p450 enzyme that is required for the production of androgens, has been exemplified by the approval of abiraterone for the treatment of castration-resistant prostate cancer (CRPC). Recently, however, it has been reported that CYP17 inhibitors can interact directly with the androgen receptor (AR). A phase I study recently reported that seviteronel, a CYP17 lyase-selective inhibitor, ædemonstrated a sustained reduction in prostate-specific antigen in a patient with CRPC, and another study showed seviteronel's direct effects on AR function. This suggested that seviteronel may have therapeutically relevant activities in addition to its ability to inhibit androgen production. Here, we have demonstrated that CYP17 inhibitors, with the exception of orteronel, can function as competitive AR antagonists. Conformational profiling revealed that the CYP17 inhibitor-bound AR adopted a conformation that resembled the unliganded AR (apo-AR), precluding nuclear localization and DNA binding. Further, we observed that seviteronel and abiraterone inhibited the growth of tumor xenografts expressing the clinically relevant mutation AR-F876L and that this activity could be attributed entirely to competitive AR antagonism. The results of this study suggest that the ability of CYP17 inhibitors to directly antagonize the AR may contribute to their clinical efficacy in CRPC.
- Published
- 2017
- Full Text
- View/download PDF
19. Discovery of an Acrylic Acid Based Tetrahydroisoquinoline as an Orally Bioavailable Selective Estrogen Receptor Degrader for ERα+ Breast Cancer.
- Author
-
Burks HE, Abrams T, Kirby CA, Baird J, Fekete A, Hamann LG, Kim S, Lombardo F, Loo A, Lubicka D, Macchi K, McDonnell DP, Mishina Y, Norris JD, Nunez J, Saran C, Sun Y, Thomsen NM, Wang C, Wang J, and Peukert S
- Subjects
- Acrylates chemistry, Acrylates pharmacokinetics, Acrylates pharmacology, Acrylates therapeutic use, Administration, Oral, Animals, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents pharmacology, Breast metabolism, Breast pathology, Breast Neoplasms metabolism, Breast Neoplasms pathology, Dogs, Drug Discovery, Estrogen Receptor alpha metabolism, Female, Humans, MCF-7 Cells, Mice, Inbred C57BL, Molecular Docking Simulation, Proteolysis drug effects, Tetrahydroisoquinolines pharmacokinetics, Tetrahydroisoquinolines pharmacology, Antineoplastic Agents chemistry, Antineoplastic Agents therapeutic use, Breast drug effects, Breast Neoplasms drug therapy, Estrogen Receptor alpha antagonists & inhibitors, Tetrahydroisoquinolines chemistry, Tetrahydroisoquinolines therapeutic use
- Abstract
Tetrahydroisoquinoline 40 has been identified as a potent ERα antagonist and selective estrogen receptor degrader (SERD), exhibiting good oral bioavailability, antitumor efficacy, and SERD activity in vivo. We outline the discovery and chemical optimization of the THIQ scaffold leading to THIQ 40 and showcase the racemization of the scaffold, pharmacokinetic studies in preclinical species, and the in vivo efficacy of THIQ 40 in a MCF-7 human breast cancer xenograft model.
- Published
- 2017
- Full Text
- View/download PDF
20. MMTV-PyMT and Derived Met-1 Mouse Mammary Tumor Cells as Models for Studying the Role of the Androgen Receptor in Triple-Negative Breast Cancer Progression.
- Author
-
Christenson JL, Butterfield KT, Spoelstra NS, Norris JD, Josan JS, Pollock JA, McDonnell DP, Katzenellenbogen BS, Katzenellenbogen JA, and Richer JK
- Subjects
- Androgen Antagonists administration & dosage, Androgen Antagonists pharmacology, Animals, Cell Line, Tumor, Cell Nucleus metabolism, Cell Proliferation drug effects, Dihydrotestosterone administration & dosage, Dihydrotestosterone pharmacology, Disease Progression, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Lung Neoplasms secondary, Mammary Tumor Virus, Mouse physiology, Mice, Mice, Transgenic, Lung Neoplasms pathology, Mammary Neoplasms, Experimental metabolism, Receptors, Androgen metabolism, Triple Negative Breast Neoplasms metabolism
- Abstract
Triple-negative breast cancer (TNBC) has a faster rate of metastasis compared to other breast cancer subtypes, and no effective targeted therapies are currently FDA-approved. Recent data indicate that the androgen receptor (AR) promotes tumor survival and may serve as a potential therapeutic target in TNBC. Studies of AR in disease progression and the systemic effects of anti-androgens have been hindered by the lack of an AR-positive (AR+) immunocompetent preclinical model. In this study, we identified the transgenic MMTV-PyMT (mouse mammary tumor virus-polyoma middle tumor-antigen) mouse mammary gland carcinoma model of breast cancer and Met-1 cells derived from this model as tools to study the role of AR in breast cancer progression. AR protein expression was examined in late-stage primary tumors and lung metastases from MMTV-PyMT mice as well as in Met-1 cells by immunohistochemistry (IHC). Sensitivity of Met-1 cells to the AR agonist dihydrotestosterone (DHT) and anti-androgen therapy was examined using cell viability, migration/invasion, and anchorage-independent growth assays. Late-stage primary tumors and lung metastases from MMTV-PyMT mice and Met-1 cells expressed abundant nuclear AR protein, while negative for estrogen and progesterone receptors. Met-1 sensitivity to DHT and AR antagonists demonstrated a reliance on AR for survival, and AR antagonists inhibited invasion and anchorage-independent growth. These data suggest that the MMTV-PyMT model and Met-1 cells may serve as valuable tools for mechanistic studies of the role of AR in disease progression and how anti-androgens affect the tumor microenvironment.
- Published
- 2017
- Full Text
- View/download PDF
21. Inhibiting androgen receptor nuclear entry in castration-resistant prostate cancer.
- Author
-
Pollock JA, Wardell SE, Parent AA, Stagg DB, Ellison SJ, Alley HM, Chao CA, Lawrence SA, Stice JP, Spasojevic I, Baker JG, Kim SH, McDonnell DP, Katzenellenbogen JA, and Norris JD
- Subjects
- Androgen Receptor Antagonists chemistry, Antineoplastic Agents chemistry, Cell Proliferation drug effects, Dose-Response Relationship, Drug, Drug Screening Assays, Antitumor, Humans, Male, Prostatic Neoplasms, Castration-Resistant pathology, Structure-Activity Relationship, Androgen Receptor Antagonists pharmacology, Antineoplastic Agents pharmacology, Cell Nucleus drug effects, Cell Nucleus metabolism, Prostatic Neoplasms, Castration-Resistant drug therapy, Prostatic Neoplasms, Castration-Resistant metabolism, Receptors, Androgen metabolism
- Abstract
Clinical resistance to the second-generation antiandrogen enzalutamide in castration-resistant prostate cancer (CRPC), despite persistent androgen receptor (AR) activity in tumors, highlights an unmet medical need for next-generation antagonists. We have identified and characterized tetra-aryl cyclobutanes (CBs) as a new class of competitive AR antagonists that exhibit a unique mechanism of action. These CBs are structurally distinct from current antiandrogens (hydroxyflutamide, bicalutamide, and enzalutamide) and inhibit AR-mediated gene expression, cell proliferation, and tumor growth in several models of CRPC. Conformational profiling revealed that CBs stabilize an AR conformation resembling an unliganded receptor. Using a variety of techniques, it was determined that the AR-CB complex was not recruited to AR-regulated promoters and, like apo AR, remains sequestered in the cytoplasm, bound to heat shock proteins. Thus, we have identified third-generation AR antagonists whose unique mechanism of action suggests that they may have therapeutic potential in CRPC., Competing Interests: Statement. A patent covering this work has been published (Publication No. WO 2015/048246).
- Published
- 2016
- Full Text
- View/download PDF
22. Efficacy of SERD/SERM Hybrid-CDK4/6 Inhibitor Combinations in Models of Endocrine Therapy-Resistant Breast Cancer.
- Author
-
Wardell SE, Ellis MJ, Alley HM, Eisele K, VanArsdale T, Dann SG, Arndt KT, Primeau T, Griffin E, Shao J, Crowder R, Lai JP, Norris JD, McDonnell DP, and Li S
- Subjects
- Animals, Antineoplastic Agents, Hormonal administration & dosage, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Breast Neoplasms genetics, Breast Neoplasms pathology, Cyclin-Dependent Kinase 4 antagonists & inhibitors, Cyclin-Dependent Kinase 6 antagonists & inhibitors, Disease-Free Survival, Drug Resistance, Neoplasm genetics, Estradiol administration & dosage, Estradiol analogs & derivatives, Female, Fulvestrant, Gene Expression Regulation, Neoplastic drug effects, Humans, MCF-7 Cells, Mice, Mutation, Piperazines administration & dosage, Pyridines administration & dosage, Tamoxifen administration & dosage, Xenograft Model Antitumor Assays, Breast Neoplasms drug therapy, Cyclin-Dependent Kinase 4 genetics, Cyclin-Dependent Kinase 6 genetics, Drug Resistance, Neoplasm drug effects, Estrogen Receptor alpha genetics, Selective Estrogen Receptor Modulators administration & dosage
- Abstract
Purpose: Endocrine therapy, using tamoxifen or an aromatase inhibitor, remains first-line therapy for the management of estrogen receptor (ESR1)-positive breast cancer. However, ESR1 mutations or other ligand-independent ESR1 activation mechanisms limit the duration of response. The clinical efficacy of fulvestrant, a selective estrogen receptor downregulator (SERD) that competitively inhibits agonist binding to ESR1 and triggers receptor downregulation, has confirmed that ESR1 frequently remains engaged in endocrine therapy-resistant cancers. We evaluated the activity of a new class of selective estrogen receptor modulators (SERM)/SERD hybrids (SSH) that downregulate ESR1 in relevant models of endocrine-resistant breast cancer. Building on the observation that concurrent inhibition of ESR1 and the cyclin-dependent kinases 4 and 6 (CDK4/6) significantly increased progression-free survival in advanced patients, we explored the activity of different SERD- or SSH-CDK4/6 inhibitor combinations in models of endocrine therapy-resistant ESR1(+) breast cancer., Experimental Design: SERDs, SSHs, and the CDK4/6 inhibitor palbociclib were evaluated as single agents or in combination in established cellular and animal models of endocrine therapy-resistant ESR1(+) breast cancer., Results: The combination of palbociclib with a SERD or an SSH was shown to effectively inhibit the growth of MCF7 cell or ESR1-mutant patient-derived tumor xenografts. In tamoxifen-resistant MCF7 xenografts, the palbociclib/SERD or SSH combination resulted in an increased duration of response as compared with either drug alone., Conclusions: A SERD- or SSH-palbociclib combination has therapeutic potential in breast tumors resistant to endocrine therapies or those expressing ESR1 mutations. See related commentary by DeMichele and Chodosh, p. 4999., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
23. Small-Molecule-Mediated Degradation of the Androgen Receptor through Hydrophobic Tagging.
- Author
-
Gustafson JL, Neklesa TK, Cox CS, Roth AG, Buckley DL, Tae HS, Sundberg TB, Stagg DB, Hines J, McDonnell DP, Norris JD, and Crews CM
- Subjects
- Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Benzamides, Cell Line, Tumor, Cell Proliferation drug effects, Drug Resistance, Neoplasm, Humans, Hydrophobic and Hydrophilic Interactions, Male, Nitriles, Phenylthiohydantoin analogs & derivatives, Phenylthiohydantoin chemistry, Phenylthiohydantoin pharmacology, Point Mutation, Prostate drug effects, Prostate metabolism, Prostatic Neoplasms drug therapy, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Receptors, Androgen genetics, Androgen Receptor Antagonists chemistry, Androgen Receptor Antagonists pharmacology, Proteolysis drug effects, Receptors, Androgen metabolism, Small Molecule Libraries chemistry, Small Molecule Libraries pharmacology
- Abstract
Androgen receptor (AR)-dependent transcription is a major driver of prostate tumor cell proliferation. Consequently, it is the target of several antitumor chemotherapeutic agents, including the AR antagonist MDV3100/enzalutamide. Recent studies have shown that a single AR mutation (F876L) converts MDV3100 action from an antagonist to an agonist. Here we describe the generation of a novel class of selective androgen receptor degraders (SARDs) to address this resistance mechanism. Molecules containing hydrophobic degrons linked to small-molecule AR ligands induce AR degradation, reduce expression of AR target genes and inhibit proliferation in androgen-dependent prostate cancer cell lines. These results suggest that selective AR degradation may be an effective therapeutic prostate tumor strategy in the context of AR mutations that confer resistance to second-generation AR antagonists., (© 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2015
- Full Text
- View/download PDF
24. Oral Selective Estrogen Receptor Downregulators (SERDs), a Breakthrough Endocrine Therapy for Breast Cancer.
- Author
-
McDonnell DP, Wardell SE, and Norris JD
- Subjects
- Animals, Antineoplastic Agents, Hormonal pharmacology, Aromatase Inhibitors chemistry, Aromatase Inhibitors pharmacology, Breast drug effects, Breast metabolism, Breast Neoplasms metabolism, Female, Humans, Selective Estrogen Receptor Modulators pharmacology, Tamoxifen pharmacology, Antineoplastic Agents, Hormonal chemistry, Breast Neoplasms drug therapy, Drug Discovery, Estrogen Receptor alpha metabolism, Selective Estrogen Receptor Modulators chemistry, Tamoxifen analogs & derivatives
- Abstract
Drugs that inhibit estrogen receptor alpha (ERα) or that block the production of estrogens remain frontline interventions in the treatment and management of breast cancer at all stages. However, resistance to endocrine therapies, especially in the setting of advanced disease, remains an impediment to durable clinical responses. Although the mechanisms underlying resistance to existing agents are complex, preclinical studies suggest that selective estrogen receptor downregulators (SERDs), molecules which eliminate ERα expression, may have particular utility in the treatment of breast cancers that have progressed on tamoxifen and/or aromatase inhibitors. The discovery and development of orally bioavailable SERDs provide the opportunity to evaluate the utility of eliminating ERα expression in advanced metastatic breast cancers.
- Published
- 2015
- Full Text
- View/download PDF
25. Obesity, cholesterol metabolism, and breast cancer pathogenesis.
- Author
-
McDonnell DP, Park S, Goulet MT, Jasper J, Wardell SE, Chang CY, Norris JD, Guyton JR, and Nelson ER
- Subjects
- Breast Neoplasms pathology, Female, Humans, Hydroxycholesterols metabolism, Lipid Metabolism, Obesity pathology, Risk Factors, Breast Neoplasms metabolism, Cholesterol metabolism, Obesity metabolism
- Abstract
Obesity and altered lipid metabolism are risk factors for breast cancer in pre- and post-menopausal women. These pathologic relationships have been attributed in part to the impact of cholesterol on the biophysical properties of cell membranes and to the influence of these changes on signaling events initiated at the membrane. However, more recent studies have indicated that the oxysterol 27-hydroxycholesterol (27HC), and not cholesterol per se, may be the primary biochemical link between lipid metabolism and cancer. The enzyme responsible for production of 27HC from cholesterol, CYP27A1, is expressed primarily in the liver and in macrophages. In addition, significantly elevated expression of this enzyme within breast tumors has also been observed. It is believed that 27HC, acting through the liver X receptor in macrophages and possibly other cells, is involved in maintaining organismal cholesterol homeostasis. It has also been shown recently that 27HC is an estrogen receptor agonist in breast cancer cells and that it stimulates the growth and metastasis of tumors in several models of breast cancer. These findings provide the rationale for the clinical evaluation of pharmaceutical approaches that interfere with cholesterol/27HC synthesis as a means to mitigate the impact of cholesterol on breast cancer pathogenesis. Cancer Res; 74(18); 4976-82. ©2014 AACR., (©2014 American Association for Cancer Research.)
- Published
- 2014
- Full Text
- View/download PDF
26. ELF3 is a repressor of androgen receptor action in prostate cancer cells.
- Author
-
Shatnawi A, Norris JD, Chaveroux C, Jasper JS, Sherk AB, McDonnell DP, and Giguère V
- Subjects
- Animals, Cell Line, Tumor, Cell Movement, DNA-Binding Proteins chemistry, Gene Expression Regulation, Neoplastic, Humans, Male, Mice, Mice, Inbred NOD, Mice, SCID, Neoplasm Transplantation, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Protein Binding, Protein Interaction Domains and Motifs, Proto-Oncogene Proteins chemistry, Proto-Oncogene Proteins c-ets, Repressor Proteins physiology, Response Elements, Transcription Factors chemistry, Transcription, Genetic, Tumor Burden, DNA-Binding Proteins physiology, Prostatic Neoplasms metabolism, Proto-Oncogene Proteins physiology, Receptors, Androgen metabolism, Transcription Factors physiology
- Abstract
The androgen receptor (AR) has a critical role in the development and progression of prostate cancer (PC) and is a major therapeutic target in this disease. The transcriptional activity of AR is modulated by the coregulators with which it interacts, and consequently deregulation of cofactor expression and/or activity impacts the expression of genes whose products can have a role in PC pathogenesis. Here we report that E74-like factor 3 (ELF3), a member of the ETS family of transcription factors, is a repressor of AR transcriptional activity. Exogenous expression of ELF3 represses AR transcriptional activity when assessed using reporter-based transfection assays or when evaluated on endogenous AR target genes. Conversely, ELF3 knock down increases the AR transcriptional activity. Biochemical dissection of this activity indicates that it results from the physical interaction between ELF3 and AR and that this interaction inhibits the recruitment of AR to specific androgen response elements within target gene promoters. Significantly, we observed that depletion of ELF3 expression in LNCaP cells promotes cell migration, whereas increased ELF3 expression severely inhibits tumor growth in vitro and in a mouse xenograft model. Taken together, these results suggest that modulation of ELF3 expression and/or AR/ELF3 interaction may have utility in the treatment of PC.
- Published
- 2014
- Full Text
- View/download PDF
27. A viable foal obtained by equine somatic cell nuclear transfer using oocytes recovered from immature follicles of live mares.
- Author
-
Choi YH, Norris JD, Velez IC, Jacobson CC, Hartman DL, and Hinrichs K
- Subjects
- Animals, Cloning, Organism methods, Embryo Transfer veterinary, Embryonic Development, Female, Pregnancy, Pregnancy Rate, Cloning, Organism veterinary, Horses physiology, Nuclear Transfer Techniques veterinary
- Abstract
The presence of heterogenous mitochondria from the host ooplast affects the acceptance of offspring obtained by somatic cell nuclear transfer. This might be avoided by obtaining oocytes from selected females, but is then complicated by low numbers of available oocytes. We examined the efficiency of equine somatic cell nuclear transfer using oocytes recovered by transvaginal aspiration of immature follicles from 11 mares. Use of metaphase I oocytes as cytoplasts and of scriptaid (a histone deacetylase inhibitor) treatment during oocyte activation were evaluated to determine if these approaches would increase blastocyst production. In experiment 1, blastocyst development was 0/14 for metaphase I oocytes and 4/103 (4%) for metaphase II oocytes. Three blastocysts were transferred to recipient mares, resulting in two pregnancies and one live foal, which died shortly after birth. In experiment 2, blastocyst development was 2/47 (4%) for control oocytes and 1/83 (1%) for scriptaid-treated oocytes. No foals were born from two blastocysts transferred in the control group. The blastocyst from the scriptaid treatment resulted in birth of a live foal. In conclusion, this is apparently the first report of production of a viable cloned foal from oocytes collected from immature follicles of live mares, supporting the possibility of cloning using oocytes from selected mares., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
28. Effects of repeated transvaginal aspiration of immature follicles on mare health and ovarian status.
- Author
-
Velez IC, Arnold C, Jacobson CC, Norris JD, Choi YH, Edwards JF, Hayden SS, and Hinrichs K
- Subjects
- Animals, Female, Oocyte Retrieval adverse effects, Oocyte Retrieval methods, Pregnancy, Horses physiology, Oocyte Retrieval veterinary, Ovarian Follicle physiology, Suction veterinary, Ultrasonography veterinary
- Abstract
Reasons for Performing Study: Transvaginal ultrasound-guided follicle aspiration (TVA) is performed clinically but there is little information available on complications associated with this procedure., Objectives: It is possible that TVA is associated with damage to the ovary and may induce peritonitis or peritoneal adhesions. This study was conducted to determine the effect of repeated TVA on mare health and ovarian status., Methods: Thirty-two mares were used for oocyte recovery via repeated TVA over a 3 year period; different mares were used each year. In Year 1, ovarian status was monitored in 11 mares by transrectal palpation and ultrasonography. In Year 2, 6 of 11 mares underwent abdominocentesis and were examined by laparoscopy after one TVA and again after multiple TVAs. In Year 3, 10 mares underwent multiple TVAs with either a 15 or a 12 gauge needle and the ovaries were removed for examination., Results: Four hundred and twenty-seven aspiration sessions (390 via TVA and 37 via needle placement through the flank) and 3202 follicle punctures (3161 TVA and 41 flank) were performed. One mare developed an ovarian abscess. Transient rectal bleeding was evident after 16% of TVA sessions. No adhesions were found on laparoscopic or gross examination of ovaries and there were minimal changes on histological evaluation., Conclusions: Follicle aspiration carries a small possibility (< 0.5%) of ovarian abscess formation. There is a possibility of rectal abrasion or puncture but little gross or histological damage to the ovary., Potential Relevance: These results provide a basis for using prophylactic administration of antibiotics after TVA and for advising mare owners of the rare but potential complications associated with the procedure.
- Published
- 2012
- Full Text
- View/download PDF
29. Evaluation of foal production following intracytoplasmic sperm injection and blastocyst culture of oocytes from ovaries collected immediately before euthanasia or after death of mares under field conditions.
- Author
-
Hinrichs K, Choi YH, Norris JD, Love LB, Bedford-Guaus SJ, Hartman DL, and Velez IC
- Subjects
- Animals, Euthanasia, Animal, Female, Blastocyst physiology, Embryo Culture Techniques veterinary, Horses physiology, Oocytes physiology, Ovary physiology, Sperm Injections, Intracytoplasmic veterinary
- Abstract
Objective: To evaluate the efficiency of foal production following intracytoplasmic sperm injection (ICSI) and blastocyst culture of oocytes from mares that died or were euthanized under field conditions., Design: Prospective case series., Animals: 16 mares (age, 3 to 19 years) that died or were euthanized for various causes., Procedures: Ovaries were collected immediately before euthanasia (n = 10) or after death (6). Ovaries were transported to the laboratory for oocyte recovery (15 mares), or oocytes were recovered at a remote location and shipped to the laboratory (1). Oocytes underwent ICSI, and presumptive zygotes were cultured for 7 to 10 days. Blastocysts were shipped to embryo transfer facilities for transcervical transfer to recipient mares., Results: Ovaries were processed 30 minutes to 12 hours (mean ± SD, 4.6 ± 3.3 hours) after mares' deaths. A mean of 14.1 ± 8.6 oocytes/mare were cultured, and 110 of 225 (49%) matured. Twenty-one blastocysts developed after ICSI and were transferred to recipient mares. Thirteen pregnancies were established; 10 healthy foals were produced from 6 donor mares. The number of blastocysts produced per mare and number of live foals produced per mare were significantly correlated with the number of oocytes recovered., Conclusions and Clinical Relevance: Foals were produced from mares after death or euthanasia under field conditions. Proportions of foals born overall (10 foals/16 mares) and mares from which ≥ 1 foal was produced (6/16) were greater than those reported following recovery and oviductal transfer of oocytes to inseminated recipients after death of donor mares under field conditions.
- Published
- 2012
- Full Text
- View/download PDF
30. The homeodomain protein HOXB13 regulates the cellular response to androgens.
- Author
-
Norris JD, Chang CY, Wittmann BM, Kunder RS, Cui H, Fan D, Joseph JD, and McDonnell DP
- Subjects
- Amino Acid Sequence, Blotting, Western, Cell Line, Cell Line, Tumor, Cell Movement drug effects, Cell Proliferation drug effects, Chromatin Immunoprecipitation, Cluster Analysis, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Gene Expression Profiling, Gene Expression Regulation drug effects, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Homeodomain Proteins genetics, Humans, Lipid Metabolism drug effects, Male, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Protein Binding drug effects, RNA, Small Interfering genetics, Receptors, Androgen genetics, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Response Elements genetics, Reverse Transcriptase Polymerase Chain Reaction, Transfection, Homeodomain Proteins metabolism, Metribolone pharmacology, Receptors, Androgen metabolism
- Abstract
HOXB13 is a member of the homeodomain family of sequence-specific transcription factors and, together with the androgen receptor (AR), plays a critical role in the normal development of the prostate gland. We demonstrate here that, in prostate cancer cells, HOXB13 is a key determinant of the response to androgens. Specifically, it was determined that HOXB13 interacts with the DNA-binding domain of AR and inhibits the transcription of genes that contain an androgen-response element (ARE). In contrast, the AR:HOXB13 complex confers androgen responsiveness to promoters that contain a specific HOXB13-response element. Further, HOXB13 and AR synergize to enhance the transcription of genes that contain a HOX element juxtaposed to an ARE. The profound effects of HOXB13 knockdown on androgen-regulated proliferation, migration, and lipogenesis in prostate cancer cells highlight the importance of the observed changes in gene expression.
- Published
- 2009
- Full Text
- View/download PDF
31. Induction of Kruppel-like factor 5 expression by androgens results in increased CXCR4-dependent migration of prostate cancer cells in vitro.
- Author
-
Frigo DE, Sherk AB, Wittmann BM, Norris JD, Wang Q, Joseph JD, Toner AP, Brown M, and McDonnell DP
- Subjects
- Cell Line, Tumor, Cell Movement, Cell Proliferation, Chemokine CXCL12 metabolism, Chromatin Immunoprecipitation, Humans, In Vitro Techniques, Male, RNA, Messenger metabolism, RNA, Small Interfering metabolism, Androgens metabolism, Gene Expression Regulation, Neoplastic, Kruppel-Like Transcription Factors biosynthesis, Prostatic Neoplasms metabolism, Receptors, Androgen metabolism, Receptors, CXCR4 metabolism
- Abstract
Advanced prostate cancers preferentially metastasize to bone, suggesting that this tissue produces factors that provide a suitable microenvironment for prostate cancer cells. Recently, it has become clear that even in antiandrogen-resistant cancers, the androgen receptor (AR)-signaling axis is required for prostate cancer progression. Therefore, we hypothesized that AR may be involved in the regulation of pathways that are responsible for the homing of prostate cancer cells to select microenvironments. In support of this hypothesis, we have determined that chemokine (C-X-C motif) receptor 4 (CXCR4), the receptor for the chemokine CXCL12, is up-regulated in prostate cancer cells in response to androgens. Given that the levels of CXCL12 are elevated at sites of known prostate cancer metastases such as bone, these results suggest that androgens may influence prostate cancer metastasis. Specifically, we demonstrate that androgens increase the levels of both CXCR4 mRNA and functional protein in LNCaP prostate cancer cells. Importantly, androgens enhanced the migration of LNCaP cells toward a CXCL12 gradient, an effect that could be blocked by the specific CXCR4 antagonist AMD3100. Interestingly, CXCR4 is not directly regulated by androgens but rather is positively up-regulated by Krüppel-like factor 5 (KLF5), a transcription factor that we have shown to be an early, direct target of AR. Further, KLF5 is both required and sufficient for androgen-mediated CXCR4 expression and migration toward CXCL12. Taken together, these findings demonstrate that AR can utilize the CXCL12/CXCR4 axis through induction of KLF5 expression to promote prostate cancer progression and highlight the potential utility of CXCR4 antagonists as prostate cancer therapeutics.
- Published
- 2009
- Full Text
- View/download PDF
32. Inhibition of prostate cancer cell growth by second-site androgen receptor antagonists.
- Author
-
Joseph JD, Wittmann BM, Dwyer MA, Cui H, Dye DA, McDonnell DP, and Norris JD
- Subjects
- Allosteric Regulation drug effects, Animals, Antineoplastic Agents chemistry, Cell Line, Tumor, Cell Proliferation drug effects, Drug Resistance, Neoplasm drug effects, Drug Screening Assays, Antitumor, Humans, Ligands, Male, Mice, Molecular Conformation, Prostatic Neoplasms genetics, Receptors, Androgen, Transcription, Genetic drug effects, Androgen Receptor Antagonists, Antineoplastic Agents pharmacology, Prostatic Neoplasms pathology
- Abstract
The impact of ligand binding on nuclear receptor (NR) structure and the ability of target cells to distinguish between different receptor-ligand complexes are key determinants of the pharmacological activity of NR ligands. However, until relatively recently, these mechanistic insights have not been used in a prospective manner to develop screens for NR modulators with specific therapeutic activities. Driven by the need for unique androgen receptor (AR) antagonists that retain activity in hormone-refractory prostate cancer, we developed and applied a conformation-based screen to identify AR antagonists that were mechanistically distinct from existing drugs of this class. Two molecules were identified by using this approach, D36 and D80, which interact with AR in a unique manner and allosterically inhibit AR agonist activity. Unlike the clinically important antiandrogens, casodex and hydroxyflutamide, both D36 and D80 block androgen action in cellular models of hormone-refractory prostate cancer. Mechanistically, these compounds further distinguish themselves from classical AR antagonists in that they do not promote AR nuclear translocation and quantitatively inhibit the association of AR with DNA even under conditions of overexpression. Although the therapeutic potential of these antiandrogens is apparent, it is the demonstration that it is possible, to modulate the interaction of cofactors with agonist-activated AR, using second-site modulators, that has the greatest potential with respect to the therapeutic exploitation of AR and other NRs.
- Published
- 2009
- Full Text
- View/download PDF
33. Differential presentation of protein interaction surfaces on the androgen receptor defines the pharmacological actions of bound ligands.
- Author
-
Norris JD, Joseph JD, Sherk AB, Juzumiene D, Turnbull PS, Rafferty SW, Cui H, Anderson E, Fan D, Dye DA, Deng X, Kazmin D, Chang CY, Willson TM, and McDonnell DP
- Subjects
- Cell Line, Tumor, Cell Proliferation drug effects, Humans, Protein Binding, Protein Conformation drug effects, Protein Interaction Mapping, Receptors, Androgen chemistry, Receptors, Androgen genetics, Structure-Activity Relationship, Transcription, Genetic drug effects, Ligands, Receptors, Androgen metabolism
- Abstract
The pharmacological activity of different nuclear receptor ligands is reflected by their impact on receptor structure. Thus, we asked whether differential presentation of protein-protein interaction surfaces on the androgen receptor (AR), a surrogate assay of receptor conformation, could be used in a prospective manner to define the pharmacological activity of bound ligands. To this end, we identified over 150 proteins/polypeptides whose ability to interact with AR is influenced in a differential manner by ligand binding. The most discriminatory of these protein-AR interactions were used to develop a robust compound-profiling tool that enabled the separation of ligands into functionally distinguishable classes. Importantly, the ligands within each class exhibited similar pharmacological activities, a result that highlights the relationship between receptor structure and activity and provides direction for the discovery of novel AR modulators.
- Published
- 2009
- Full Text
- View/download PDF
34. Development of a small-molecule serum- and glucocorticoid-regulated kinase-1 antagonist and its evaluation as a prostate cancer therapeutic.
- Author
-
Sherk AB, Frigo DE, Schnackenberg CG, Bray JD, Laping NJ, Trizna W, Hammond M, Patterson JR, Thompson SK, Kazmin D, Norris JD, and McDonnell DP
- Subjects
- Benzoates pharmacology, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Cell Growth Processes physiology, Cell Line, Tumor, HeLa Cells, Humans, Immediate-Early Proteins biosynthesis, Immediate-Early Proteins genetics, Immediate-Early Proteins metabolism, Male, Metribolone pharmacology, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Protein Serine-Threonine Kinases biosynthesis, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, RNA, Messenger biosynthesis, RNA, Messenger genetics, RNA, Small Interfering genetics, Receptors, Androgen metabolism, Up-Regulation, Immediate-Early Proteins antagonists & inhibitors, Prostatic Neoplasms drug therapy, Prostatic Neoplasms enzymology, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Androgens, through their actions on the androgen receptor (AR), are required for the development of the prostate and contribute to the pathologic growth dysregulation observed in prostate cancers. Consequently, androgen ablation has become an essential component of the pharmacotherapy of prostate cancer. In this study, we explored the utility of targeting processes downstream of AR as an alternate approach for therapy. Specifically, we show that the serum and glucocorticoid-regulated kinase 1 (SGK1) gene is an androgen-regulated target gene in cellular models of prostate cancer. Furthermore, functional serum- and glucocorticoid-regulated kinase 1 (SGK1) protein, as determined by the phosphorylation of its target Nedd4-2, was also increased with androgen treatment. Importantly, we determined that RNA interference-mediated knockdown of SGK1 expression attenuates the androgen-mediated growth of the prostate cancer cell line LNCaP. Given these findings, we explored the utility of SGK1 as a therapeutic target in prostate cancer by developing and evaluating a small-molecule inhibitor of this enzyme. From these studies emerged GSK650394, a competitive inhibitor that quantitatively blocks the effect of androgens on LNCaP cell growth. Thus, in addition to androgen ablation, inhibition of pathways downstream of AR is likely to have therapeutic utility in prostate cancer.
- Published
- 2008
- Full Text
- View/download PDF
35. Linking ligand-induced alterations in androgen receptor structure to differential gene expression: a first step in the rational design of selective androgen receptor modulators.
- Author
-
Kazmin D, Prytkova T, Cook CE, Wolfinger R, Chu TM, Beratan D, Norris JD, Chang CY, and McDonnell DP
- Subjects
- Amino Acid Sequence, Androgens, Binding Sites, Cell Line, Gene Expression, Gene Expression Profiling, Humans, In Vitro Techniques, Ligands, Models, Molecular, Molecular Sequence Data, Oligonucleotide Array Sequence Analysis, Peptide Library, Protein Array Analysis, Protein Conformation, Receptors, Androgen chemistry, Recombinant Proteins agonists, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Thermodynamics, Receptors, Androgen genetics, Receptors, Androgen metabolism
- Abstract
We have previously identified a family of novel androgen receptor (AR) ligands that, upon binding, enable AR to adopt structures distinct from that observed in the presence of canonical agonists. In this report, we describe the use of these compounds to establish a relationship between AR structure and biological activity with a view to defining a rational approach with which to identify useful selective AR modulators. To this end, we used combinatorial peptide phage display coupled with molecular dynamic structure analysis to identify the surfaces on AR that are exposed specifically in the presence of selected AR ligands. Subsequently, we used a DNA microarray analysis to demonstrate that differently conformed receptors facilitate distinct patterns of gene expression in LNCaP cells. Interestingly, we observed a complete overlap in the identity of genes expressed after treatment with mechanistically distinct AR ligands. However, it was differences in the kinetics of gene regulation that distinguished these compounds. Follow-up studies, in cell-based assays of AR action, confirmed the importance of these alterations in gene expression. Together, these studies demonstrate an important link between AR structure, gene expression, and biological outcome. This relationship provides a firm underpinning for mechanism-based screens aimed at identifying SARMs with useful clinical profiles.
- Published
- 2006
- Full Text
- View/download PDF
36. Structural basis for an unexpected mode of SERM-mediated ER antagonism.
- Author
-
Wu YL, Yang X, Ren Z, McDonnell DP, Norris JD, Willson TM, and Greene GL
- Subjects
- Amino Acid Sequence, Cinnamates chemistry, Humans, Hydrophobic and Hydrophilic Interactions, Ligands, Molecular Sequence Data, Protein Structure, Tertiary, Selective Estrogen Receptor Modulators chemistry, Stilbenes chemistry, Structure-Activity Relationship, Cinnamates pharmacology, Estrogen Receptor alpha antagonists & inhibitors, Selective Estrogen Receptor Modulators pharmacology, Stilbenes pharmacology
- Abstract
Tamoxifen is effective for the prevention and treatment of estrogen-dependent breast cancers, but is associated with an increased incidence of endometrial tumors. We report the crystal structure of the estrogen receptor alpha (ERalpha) ligand binding domain (LBD) bound to the structurally similar compound GW5638, which has therapeutic potential and does not stimulate the uterus. Like tamoxifen, GW5638 relocates the carboxy-terminal helix (H12) to the known coactivator-docking site in the ERalpha LBD. However, GW5638 repositions residues in H12 through specific contacts with the N terminus of this helix. In contrast to tamoxifen, the resulting increase in exposed hydrophobic surface of ERalpha LBD correlates with a significant destabilization of ERalpha in MCF-7 cells. Thus, the GW5638-ERalpha LBD structure reveals an unexpected mode of SERM-mediated ER antagonism, in which the stability of ERalpha is decreased through an altered position of H12. This dual mechanism of antagonism may explain why GW5638 can inhibit tamoxifen-resistant breast tumors.
- Published
- 2005
- Full Text
- View/download PDF
37. Single-step purification of full-length human androgen receptor.
- Author
-
Juzumiene D, Chang CY, Fan D, Hartney T, Norris JD, and McDonnell DP
- Abstract
The full-length human androgen receptor with an N-terminal biotin acceptor peptide tag was overexpressed in Spodoptera frugiperda cells in the presence of 1 microM dihydrotestosterone. Site-specific biotinylation of BAP was achieved in vivo by co-expression of E. coli biotin holoenzyme synthetase. The androgen receptor was purified by single-step affinity chromatography using Streptavidin Mutein Matrix under native conditions. The resultant protein was active, stable, 95% homogeneous, and we obtained sufficient yield for use in functional and structural studies.
- Published
- 2005
- Full Text
- View/download PDF
38. Application of random peptide phage display to the study of nuclear hormone receptors.
- Author
-
Chang CY, Norris JD, Jansen M, Huang HJ, and McDonnell DP
- Subjects
- Animals, Base Sequence, DNA chemistry, Enzyme-Linked Immunosorbent Assay, Estrogen Receptor alpha, Genetic Vectors, Humans, Ligands, Molecular Sequence Data, Protein Conformation, Receptors, Cytoplasmic and Nuclear analysis, Receptors, Estrogen chemistry, Transcription, Genetic, Biochemistry methods, Peptide Library, Receptors, Cytoplasmic and Nuclear chemistry
- Published
- 2003
- Full Text
- View/download PDF
39. Identification of a negative regulatory surface within estrogen receptor alpha provides evidence in support of a role for corepressors in regulating cellular responses to agonists and antagonists.
- Author
-
Huang HJ, Norris JD, and McDonnell DP
- Subjects
- Amino Acid Motifs, Amino Acid Sequence, Binding Sites genetics, Cell Line, Estradiol metabolism, Estradiol pharmacology, Estrogen Receptor Modulators metabolism, Estrogen Receptor Modulators pharmacology, Estrogen Receptor alpha, Fulvestrant, HeLa Cells, Humans, In Vitro Techniques, Molecular Sequence Data, Mutagenesis, Site-Directed, Peptide Library, Receptors, Estrogen agonists, Receptors, Estrogen antagonists & inhibitors, Receptors, Estrogen genetics, Repressor Proteins metabolism, Signal Transduction, Tamoxifen metabolism, Tamoxifen pharmacology, Transcription, Genetic, Estradiol analogs & derivatives, Receptors, Estrogen chemistry
- Abstract
Several lines of evidence have indicated that the estrogen receptor (ER) can recruit the corepressors, nuclear receptor corepressor (NCoR) and silencing mediator of retinoid and thyroid receptors (SMRT), to target genes in the presence of tamoxifen, suggesting a possible role for NCoR/SMRT in regulating ER pharmacology. However, a tamoxifen-dependent, direct interaction between NCoR/SMRT and ER in vitro has not been demonstrated. To investigate the possible involvement of different corepressors in the actions of antiestrogen-bound ER, we have constructed a phage display library that expresses 23-amino acid peptides containing the canonical CoRNR box motif in an otherwise random background. Screening of the CoRNR box library with apo-ER or ER treated with tamoxifen or ICI 182,780 led to the isolation of peptides whose ability to interact with ER was influenced by the nature of the bound ligand. Using a series of ERalpha mutants, we found that helix 12 was not required for the binding of CoRNR box peptides, whereas disruption of helixes 3 and 5 had a marked effect on peptide binding. One mutant, ER-L372R, lost the ability to interact with CoRNR box-containing peptides without affecting its binding to LXXLL motif-containing peptides. The estradiol- and tamoxifen-mediated transcriptional activity of ER-L372R was dramatically increased by 11- and 3-fold, respectively, compared with that of wild-type ERalpha. The ICI 182,780-mediated repressional activity of this mutant was also reduced by 4-fold compared with that of wild-type ERalpha. These results suggest that leucine 372 may be an important part of the interaction surface on ER that is responsible for corepressor binding. In addition, our data suggest that corepressors, other than NCoR/SMRT, may be involved in ER signaling.
- Published
- 2002
- Full Text
- View/download PDF
40. Elucidation of the molecular mechanism of action of selective estrogen receptor modulators.
- Author
-
McDonnell DP, Wijayaratne A, Chang CY, and Norris JD
- Subjects
- Estradiol pharmacology, Female, Humans, Tamoxifen pharmacology, Receptors, Estrogen drug effects, Selective Estrogen Receptor Modulators pharmacology
- Abstract
The term selective estrogen receptor modulator (SERM) describes a group of pharmaceuticals that manifest estrogen receptor (ER) agonist activity in some tissues but opposes estrogen action in others. Although the name describing this class of drugs is new, the concept is not, as compounds exhibiting tissue-selective ER agonist/antagonist properties have been available for nearly 40 years. What is new is the idea that it may be possible to capitalize on the paradoxical activities of SERMs and develop them as target organ-selective ER agonists for the treatment of osteoporosis and other estrogenopathies. This realization has provided the impetus for research in this area, the progress of which is described in this review.
- Published
- 2002
- Full Text
- View/download PDF
41. Connections and regulation of the human estrogen receptor.
- Author
-
McDonnell DP and Norris JD
- Subjects
- Acetyltransferases metabolism, Enhancer Elements, Genetic, Estrogen Receptor alpha, Estrogen Receptor beta, Histone Acetyltransferases, Humans, Models, Biological, Protein Binding, Receptors, Estrogen chemistry, Receptors, Estrogen genetics, Receptors, Progesterone metabolism, Trans-Activators metabolism, Transcription Factors metabolism, Transcriptional Activation, Estrogens metabolism, Receptors, Estrogen metabolism, Saccharomyces cerevisiae Proteins, Signal Transduction, Transcription, Genetic
- Abstract
Estrogen regulates a plethora of functionally dissimilar processes in a broad range of tissues. Recent progress in the study of the molecular mechanism of action of estrogen(s) has revealed why different cells can respond to the same hormone in a different manner. Three of these findings are of particular importance: (i) There are two genetically and functionally distinct estrogen receptors that have distinct expression patterns in vivo; (ii) the positive and negative transcriptional activities of these receptors require them to engage transcription cofactors (coactivators or corepressors) in target cells; and (iii) not all cofactors are functionally equivalent, nor are they expressed in the same manner in all cells. Thus, although the estrogen receptor is required for a cell to respond to an estrogenic stimulus, the nature and extent of that response are determined by the proteins, pathways, and processes with which the receptor interacts.
- Published
- 2002
- Full Text
- View/download PDF
42. A negative coregulator for the human ER.
- Author
-
Norris JD, Fan D, Sherk A, and McDonnell DP
- Subjects
- Amino Acid Sequence, Binding Sites, Blotting, Northern, Consensus Sequence, Estradiol pharmacology, Estrogen Antagonists pharmacology, Estrogen Receptor alpha, Gene Expression, HeLa Cells, Humans, Molecular Sequence Data, Mutagenesis, RNA metabolism, RNA Splicing Factors, RNA-Binding Proteins chemistry, RNA-Binding Proteins genetics, Receptors, Estrogen drug effects, Repressor Proteins chemistry, Repressor Proteins genetics, Tamoxifen pharmacology, Tissue Distribution, Transcription, Genetic drug effects, Transfection, Tumor Cells, Cultured, RNA-Binding Proteins physiology, Receptors, Estrogen physiology, Repressor Proteins physiology
- Abstract
ERalpha is a ligand-activated transcription factor and a key regulator of the processes involved in cellular proliferation and differentiation. In addition, aberrant ERalpha activity is linked to several pathological conditions including breast cancer. A complex network of coregulatory proteins is largely believed to determine the transcriptional activity of ERalpha. We report here the isolation of a protein, denoted RTA for repressor of tamoxifen transcriptional activity, which contains an RNA recognition motif and interacts with the receptor N-terminal activation domain. RTA interacts with RNA in vitro, and its overexpression inhibits the partial agonist activity manifest by the antiestrogen tamoxifen while minimally affecting E2-activated transcription. Mutation of the RNA recognition motif alters RNA binding specificity and results in a dominant negative form of RTA that leads to derepression of ERalpha transcriptional activity, allowing all classes of antiestrogens to manifest partial agonist activity and enhancing agonist efficacy. These findings suggest a role for RNA binding proteins as coregulatory factors of the nuclear receptor family and reveal a novel mechanism by which antiestrogens can manifest agonist activities in some tissues.
- Published
- 2002
- Full Text
- View/download PDF
43. Definition of the molecular and cellular mechanisms underlying the tissue-selective agonist/antagonist activities of selective estrogen receptor modulators.
- Author
-
McDonnell DP, Connor CE, Wijayaratne A, Chang CY, and Norris JD
- Subjects
- Animals, Drug Resistance, Estrogen Antagonists pharmacology, Estrogen Receptor beta, Female, Humans, Protein Conformation, Receptors, Estrogen chemistry, Receptors, Estrogen physiology, Selective Estrogen Receptor Modulators metabolism, Selective Estrogen Receptor Modulators therapeutic use, Structure-Activity Relationship, Tamoxifen pharmacology, Selective Estrogen Receptor Modulators pharmacology
- Abstract
The term selective estrogen receptor modulators describes a group of pharmaceuticals that function as estrogen receptor (ER) agonists in some tissues but that oppose estrogen action in others. Although the name for this class of drugs has been adopted only recently, the concept is not new, as compounds exhibiting tissue-selective ER agonist/antagonist properties have been around for nearly 40 years. What is new is the idea that it may be possible to capitalize on the paradoxical activities of these drugs and develop them as target organ-selective ER agonists for the treatment of osteoporosis and other estrogenopathies. This realization has provided the impetus for research in this area, the progress of which is discussed in this review.
- Published
- 2002
- Full Text
- View/download PDF
44. Capitalizing on the complexities of estrogen receptor pharmacology in the quest for the perfect SERM.
- Author
-
McDonnell DP, Chang CY, and Norris JD
- Subjects
- Amino Acid Sequence, Animals, Binding Sites, Estradiol pharmacology, Female, Humans, Ligands, Molecular Sequence Data, Peptide Fragments chemistry, Receptors, Estrogen antagonists & inhibitors, Receptors, Estrogen physiology, Sequence Alignment, Tamoxifen pharmacology, Receptors, Estrogen agonists, Selective Estrogen Receptor Modulators chemistry, Selective Estrogen Receptor Modulators pharmacology
- Abstract
The term Selective Estrogen Receptor Modulators (SERMs) has been used of late to describe a group of pharmaceuticals that manifest estrogen receptor (ER) agonist activity in some tissues, but that oppose estrogen action in others. Whereas the name describing this class of drugs is new, the concept is not. Indeed, compounds exhibiting tissue-selective ER agonist/antagonist properties have been around for nearly 40 years. What is new is the idea that it may be possible to capitalize on the paradoxical activities of these drugs and develop them as treatments for estrogenopathies where it is desirable to direct therapy to a specific estrogen-responsive target organ. This realization has provided the impetus for research in this area and has pushed the development and clinical use of this class of drugs. The objective of this review is to describe how the medical need for SERMs arose and how recent studies of the mechanism of action of the currently available drugs are paving the way for the development of novel drugs with improved selectivity.
- Published
- 2001
- Full Text
- View/download PDF
45. Circumventing tamoxifen resistance in breast cancers using antiestrogens that induce unique conformational changes in the estrogen receptor.
- Author
-
Connor CE, Norris JD, Broadwater G, Willson TM, Gottardis MM, Dewhirst MW, and McDonnell DP
- Subjects
- Amino Acid Sequence, Animals, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Division drug effects, Cell Division physiology, Drug Interactions, Drug Resistance, Neoplasm, Estrogen Receptor alpha, Estrogen Receptor beta, Female, Humans, Mice, Mice, Inbred BALB C, Mice, Nude, Molecular Sequence Data, Neoplasms, Hormone-Dependent metabolism, Neoplasms, Hormone-Dependent pathology, Protein Conformation, Receptors, Estrogen metabolism, Xenograft Model Antitumor Assays, Breast Neoplasms drug therapy, Cinnamates pharmacology, Estrogen Receptor Modulators pharmacology, Neoplasms, Hormone-Dependent drug therapy, Receptors, Estrogen drug effects, Stilbenes pharmacology, Tamoxifen pharmacology
- Abstract
Tamoxifen inhibits estrogen receptor (ER) transcriptional activity by competitively inhibiting estradiol binding and inducing conformational changes in the receptor that may prevent its interaction with coactivators. In bone, the cardiovascular system, and some breast tumors, however, tamoxifen exhibits agonist activity, suggesting that the tamoxifen-ER complex is not recognized identically in all cells. We used phage display to demonstrate that the antiestrogen GW5638 induces a unique structural change in the ER. The biological significance of this conformational change was revealed in studies that demonstrated that tamoxifen-resistant breast tumor explants are not cross-resistant to GW5638. Because of these properties, this drug is currently being developed as a potential therapeutic for tamoxifen-resistant breast cancers.
- Published
- 2001
46. Estrogen receptor-cofactor interactions as targets for novel drug discovery.
- Author
-
Norris JD, Chang C, and McDonnell DP
- Subjects
- Amino Acid Sequence, Animals, Binding Sites, Drug Design, Female, Humans, Ligands, Molecular Sequence Data, Receptors, Estrogen chemistry, Receptors, Estrogen drug effects, Estrogens physiology, Receptors, Estrogen physiology
- Published
- 2001
- Full Text
- View/download PDF
47. Development of peptide antagonists that target estrogen receptor-cofactor interactions.
- Author
-
McDonnell DP, Chang CY, and Norris JD
- Subjects
- Amino Acid Motifs, Amino Acid Sequence, Animals, Binding Sites, Estrogen Antagonists metabolism, Estrogen Antagonists therapeutic use, Estrogen Receptor Modulators metabolism, Estrogen Receptor Modulators pharmacology, Estrogen Receptor Modulators therapeutic use, Humans, Molecular Sequence Data, Peptides chemistry, Peptides metabolism, Peptides therapeutic use, Protein Binding drug effects, Receptors, Estrogen agonists, Receptors, Estrogen chemistry, Substrate Specificity, Tamoxifen antagonists & inhibitors, Tamoxifen pharmacology, Transcription Factors antagonists & inhibitors, Transcription Factors metabolism, Transcriptional Activation drug effects, Estrogen Antagonists pharmacology, Peptides pharmacology, Receptors, Estrogen antagonists & inhibitors, Receptors, Estrogen metabolism
- Abstract
We have developed a series of high-affinity peptide antagonists that inhibit the transcriptional activity of both subtypes of the human estrogen receptor (ERalpha and ERbeta). We believe that it will be possible to develop these peptides, or corresponding peptidomimetic derivatives, into pharmaceuticals for use in the treatment of breast cancer and other estrogenopathies. It is anticipated that drugs of this type could be used in combination with classical antiestrogens, such as tamoxifen, to achieve a complete blockage of ER-transcriptional activity. Although ER has been the primary target of our studies to date, it is likely that the insights gained from this work will apply to other nuclear receptors and transcription factors.
- Published
- 2000
- Full Text
- View/download PDF
48. Modulation of estrogen receptor-alpha transcriptional activity by the coactivator PGC-1.
- Author
-
Tcherepanova I, Puigserver P, Norris JD, Spiegelman BM, and McDonnell DP
- Subjects
- Estrogen Receptor alpha, Humans, Mutation, Receptors, Cytoplasmic and Nuclear metabolism, Receptors, Estrogen chemistry, Receptors, Estrogen genetics, Signal Transduction, Trans-Activators genetics, Transcription Factors genetics, Transcription, Genetic, Tumor Cells, Cultured, Receptors, Estrogen metabolism, Trans-Activators metabolism, Transcription Factors metabolism
- Abstract
A transcriptional coactivator of the peroxisome proliferator-activated receptor-gamma (PPARgamma), PPARgamma-coactivator-1(PGC-1) interacts in a constitutive manner with the hinge domain of PPARgamma and enhances its transcriptional activity. In this study we demonstrate that PGC-1 is a coactivator of estrogen receptor-alpha (ERalpha)-dependent transcriptional activity. However the mechanism by which PGC-1 interacts with ERalpha is different from that of PPARgamma. Specifically, it was determined that the carboxyl terminus of PGC-1 interacts in a ligand-independent manner with the ERalpha hinge domain. In addition, an LXXLL motif within the amino terminus of PGC-1 was shown to interact in an agonist-dependent manner with the AF2 domain within the carboxyl terminus of ERalpha. The ability of PGC-1 to associate with and potentiate the transcriptional activity of an ERalpha-AF2 mutant that is unable to interact with the p160 class of coactivators suggests that this coactivator may have a unique role in estrogen signaling. It is concluded from these studies that PGC-1 is a bona fide ERalpha coactivator, which may serve as a convergence point between PPARgamma and ERalpha signaling.
- Published
- 2000
- Full Text
- View/download PDF
49. Comparative analyses of mechanistic differences among antiestrogens.
- Author
-
Wijayaratne AL, Nagel SC, Paige LA, Christensen DJ, Norris JD, Fowlkes DM, and McDonnell DP
- Subjects
- Blood Proteins metabolism, Breast Neoplasms pathology, Cell Division drug effects, Cinnamates pharmacology, Drug Stability, Estradiol analogs & derivatives, Estradiol pharmacology, Estrogen Receptor alpha, Fulvestrant, Gene Expression drug effects, Humans, Protein Binding, Protein Conformation drug effects, Raloxifene Hydrochloride pharmacology, Receptors, Estrogen chemistry, Receptors, Estrogen genetics, Stilbenes pharmacology, Tamoxifen analogs & derivatives, Tamoxifen pharmacology, Transcription, Genetic, Tumor Cells, Cultured, Estrogen Antagonists pharmacology
- Abstract
Antiestrogens such as tamoxifen are one of the most effective methods of treating estrogen receptor (ERalpha) positive breast cancers; however, the effectiveness of this therapy is limited by the almost universal development of resistance to the drug. If antiestrogens are recognized differently by the cell as it has been suggested, then in disease conditions where tamoxifen fails to function effectively, a mechanistically different antiestrogen might yield successful results. Although many antiestrogens have been developed, a direct comparison of their mechanisms of action is lacking, thus limiting their utility. Therefore, to determine if there are mechanistic differences among available antiestrogens, we have carried out a comprehensive analysis of the molecular mechanisms of action of 4-hydroxy-tamoxifen (40HT), idoxifene, raloxifene, GW7604, and ICI 182,780. Using a novel set of peptides that recognize different surfaces on ERalpha, we have found that following binding to ERalpha, each ligand induces a distinct ERalpha-ligand conformation. Furthermore, transcriptional assays indicate that each ERalpha-ligand complex is recognized distinctly by the transcription machinery, and consequently, antiestrogens vary in their ability to inhibit estradiol- and 40HT-mediated activities. Relative binding assays have shown that the affinity of these ligands for ERalpha is not always representative of their inhibitory activity. Using this assay, we have also shown that the pharmacology of each antiestrogen is influenced differently by hormone binding proteins. Furthermore, GW7604, like ICI 182,780, but unlike the other antiestrogens evaluated, decreases the stability of the receptor. Overall, our results indicate that there are clear mechanistic distinctions among each of the antiestrogens studied. However, GW7604 and ICI 182,780 differ more significantly from tamoxifen than idoxifene and raloxifene. These data, which reveal differences among antiestrogens, should assist in the selection of compounds for the clinical regulation of ERalpha function.
- Published
- 1999
- Full Text
- View/download PDF
50. Dissection of the LXXLL nuclear receptor-coactivator interaction motif using combinatorial peptide libraries: discovery of peptide antagonists of estrogen receptors alpha and beta.
- Author
-
Chang Cy, Norris JD, Grøn H, Paige LA, Hamilton PT, Kenan DJ, Fowlkes D, and McDonnell DP
- Subjects
- Amino Acid Sequence, Binding Sites, Estrogen Receptor alpha, Estrogen Receptor beta, HeLa Cells, Humans, Ligands, Molecular Sequence Data, Peptide Library, Peptides metabolism, Receptors, Cytoplasmic and Nuclear metabolism, Receptors, Estrogen genetics, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Transcription, Genetic, Tumor Cells, Cultured, Receptors, Estrogen metabolism
- Abstract
Recruitment of transcriptional coactivators following ligand activation is a critical step in nuclear receptor-mediated target gene expression. Upon binding an agonist, the receptor undergoes a conformational change which facilitates the formation of a specific coactivator binding pocket within the carboxyl terminus of the receptor. This permits the alpha-helical LXXLL motif within some coactivators to interact with the nuclear receptors. Until recently, the LXXLL motif was thought to function solely as a docking module; however, it now appears that sequences flanking the core motif may play a role in determining receptor selectivity. To address this issue, we used a combinatorial phage display approach to evaluate the role of flanking sequences in influencing these interactions. We sampled more than 10(8) variations of the core LXXLL motif with estradiol-activated estrogen receptor alpha (ERalpha) as a target and found three different classes of peptides. All of these peptides interacted with ERalpha in an agonist-dependent manner and disrupted ERalpha-mediated transcriptional activity when introduced into target cells. Using a series of ERalpha-mutants, we found that these three classes of peptides showed different interaction patterns from each other, suggesting that not all LXXLL motifs are the same and that receptor binding selectivity can be achieved by altering sequences flanking the LXXLL core motif. Most notable in this regard was the discovery of a peptide which, when overexpressed in cells, selectively disrupted ERbeta- but not ERalpha-mediated reporter gene expression. This novel ERbeta-specific antagonist may be useful in identifying and characterizing the ERbeta-regulated process in estradiol-responsive cells. In conclusion, using a combinatorial approach to define cofactor-receptor interactions, we have clearly been able to demonstrate that not all LXXLL motifs are functionally equivalent, a finding which suggests that it may be possible to target receptor-LXXLL interactions to develop receptor-specific antagonists.
- Published
- 1999
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.