520 results on '"Katzenellenbogen JA"'
Search Results
2. Increase in chemokine CXCL1 by ER beta ligand treatment is a key mediator in promoting axon myelination
- Author
-
Karim, H, Kim, SH, Lapato, AS, Yasui, N, Katzenellenbogen, JA, and Tiwari-Woodruff, SK
- Subjects
animal diseases - Abstract
Estrogen receptor β (ERβ) ligands promote remyelination in mouse models of multiple sclerosis. Recent work using experimental autoimmune encephalomyelitis (EAE) has shown that ERβ ligands induce axon remyelination, but impact peripheral inflammation to varying degrees. To identify if ERβ ligands initiate a common immune mechanism in remyelination, central and peripheral immunity and pathology in mice given ERβ ligands at peak EAE were assessed. All ERβ ligands induced differential expression of cytokines and chemokines, but increased levels of CXCL1 in the periphery and in astrocytes. Oligodendrocyte CXCR2 binds CXCL1 and has been implicated in normal myelination. In addition, despite extensive immune cell accumulation in the CNS, all ERβ ligands promoted extensive remyelination in mice at peak EAE. This finding highlights a component of the mechanism by which ERβ ligands mediate remyelination. Hence, interplay between the immune system and central nervous system may be responsible for the remyelinating effects of ERβ ligands. Our findings of potential neuroprotective benefits arising from the presence of CXCL1 could have implications for improved therapies for multiple sclerosis.
- Published
- 2018
3. Abstract P4-07-02: Withdrawn
- Author
-
Katzenellenbogen, BS, primary, Guillen, VS, additional, Ziegler, Y, additional, Kim, SH, additional, Laws, MJ, additional, Zhao, Y, additional, Yasuda, MA, additional, Li, Z, additional, El-Ashry, D, additional, and Katzenellenbogen, JA, additional
- Published
- 2019
- Full Text
- View/download PDF
4. Abstract P2-08-07: Anti-proliferative and anti-inflammatory estrogen receptor agents for treatment of endocrine-resistant breast cancer
- Author
-
Josan, JS, primary, Pokludova, K, additional, Devi, S, additional, Srinivasan, S, additional, Katzenellenbogen, JA, additional, and Nettles, KW, additional
- Published
- 2017
- Full Text
- View/download PDF
5. Iterative Catalyst-Controlled Diastereoselective Matteson Homologations Enable the Selective Synthesis of Benzestrol Isomers.
- Author
-
Angle SR, Sharma HA, Choi CK, Carlson KE, Hou Y, Nwachukwu JC, Kim SH, Katzenellenbogen BS, Nettles KW, Katzenellenbogen JA, and Jacobsen EN
- Subjects
- Stereoisomerism, Catalysis, Molecular Structure, Humans, Estrogen Receptor alpha metabolism, Estrogen Receptor alpha chemistry
- Abstract
We report the development of an iterative Matteson homologation reaction with catalyst-controlled diastereoselectivity through the design of a new catalyst. This reaction was applied to the selective synthesis of each stereoisomer of benzestrol, a bioactive compound with estrogenic activity featuring three contiguous stereocenters. The different stereoisomers were assayed to determine their binding affinity for the estrogen receptor α (ERα), and the absolute configuration of the compound having uniquely high activity was determined. This research lays a framework for the catalytic synthesis and study of complete stereoisomeric sets of other bioactive molecules and chemical probes containing contiguous stereocenters.
- Published
- 2024
- Full Text
- View/download PDF
6. Resistance to FOXM1 inhibitors in breast cancer is accompanied by impeding ferroptosis and apoptotic cell death.
- Author
-
Kumar S, Ziegler Y, Plotner BN, Flatt KM, Kim SH, Katzenellenbogen JA, and Katzenellenbogen BS
- Subjects
- Humans, Female, Cell Line, Tumor, Cell Survival drug effects, Cell Proliferation drug effects, Mitochondria metabolism, Mitochondria drug effects, Artemisinins pharmacology, Artemisinins therapeutic use, Cell Movement drug effects, Gene Expression Regulation, Neoplastic drug effects, Forkhead Box Protein M1 metabolism, Forkhead Box Protein M1 genetics, Ferroptosis drug effects, Drug Resistance, Neoplasm drug effects, Apoptosis drug effects, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Breast Neoplasms metabolism
- Abstract
Purpose: Cancer treatments often become ineffective because of acquired drug resistance. To characterize changes in breast cancer cells accompanying development of resistance to inhibitors of the oncogenic transcription factor, FOXM1, we investigated the suppression of cell death pathways, especially ferroptosis, in FOXM1 inhibitor-resistant cells. We also explored whether ferroptosis activators can synergize with FOXM1 inhibitors and can overcome FOXM1 inhibitor resistance., Methods: In estrogen receptor-positive and triple-negative breast cancer cells treated with FOXM1 inhibitor NB73 and ferroptosis activators dihydroartemisinin and JKE1674, alone and in combination, we measured suppression of cell viability, motility, and colony formation, and monitored changes in gene and protein pathway expressions and mitochondrial integrity., Results: Growth suppression of breast cancer cells by FOXM1 inhibitors is accompanied by increased cell death and alterations in mitochondrial morphology and metabolic activity. Low doses of FOXM1 inhibitor strongly synergize with ferroptosis inducers to reduce cell viability, migration, colony formation, and expression of proliferation-related genes, and increase intracellular Fe
+2 and lipid peroxidation, markers of ferroptosis. Acquired resistance to FOXM1 inhibition is associated with increased expression of cancer stem-cell markers and proteins that repress ferroptosis, enabling cell survival and drug resistance. Notably, resistant cells are still sensitive to growth suppression by low doses of ferroptosis activators, effectively overcoming the acquired resistance., Conclusion: Delineating changes in viability and cell death pathways that can overcome drug resistance should be helpful in determining approaches that might best prevent or reverse resistance to therapeutic targeting of FOXM1 and ultimately improve patient clinical outcomes., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
7. Asymmetric allostery in estrogen receptor-α homodimers drives responses to the ensemble of estrogens in the hormonal milieu.
- Author
-
Min CK, Nwachukwu JC, Hou Y, Russo RJ, Papa A, Min J, Peng R, Kim SH, Ziegler Y, Rangarajan ES, Izard T, Katzenellenbogen BS, Katzenellenbogen JA, and Nettles KW
- Subjects
- Allosteric Regulation, Humans, Ligands, Binding Sites, Protein Binding, Protein Conformation, Estrogen Receptor alpha metabolism, Estrogen Receptor alpha chemistry, Protein Multimerization, Molecular Dynamics Simulation, Estrogens metabolism, Estrogens chemistry
- Abstract
The estrogen receptor-α (ER) is thought to function only as a homodimer but responds to a variety of environmental, metazoan, and therapeutic estrogens at subsaturating doses, supporting binding mixtures of ligands as well as dimers that are only partially occupied. Here, we present a series of flexible ER ligands that bind to receptor dimers with individual ligand poses favoring distinct receptor conformations-receptor conformational heterodimers-mimicking the binding of two different ligands. Molecular dynamics simulations showed that the pairs of different ligand poses changed the correlated motion across the dimer interface to generate asymmetric communication between the dimer interface, the ligands, and the surface binding sites for epigenetic regulatory proteins. By examining the binding of the same ligand in crystal structures of ER in the agonist vs. antagonist conformers, we also showed that these allosteric signals are bidirectional. The receptor conformer can drive different ligand binding modes to support agonist vs. antagonist activity profiles, a revision of ligand binding theory that has focused on unidirectional signaling from the ligand to the coregulator binding site. We also observed differences in the allosteric signals between ligand and coregulator binding sites in the monomeric vs. dimeric receptor, and when bound by two different ligands, states that are physiologically relevant. Thus, ER conformational heterodimers integrate two different ligand-regulated activity profiles, representing different modes for ligand-dependent regulation of ER activity., Competing Interests: Competing interests statement:The authors declare no competing interest.
- Published
- 2024
- Full Text
- View/download PDF
8. NB compounds are potent and efficacious FOXM1 inhibitors in high-grade serous ovarian cancer cells.
- Author
-
Liu C, Vorderbruggen M, Muñoz-Trujillo C, Kim SH, Katzenellenbogen JA, Katzenellenbogen BS, and Karpf AR
- Subjects
- Humans, Female, Cell Line, Tumor, Cystadenocarcinoma, Serous drug therapy, Cystadenocarcinoma, Serous pathology, Cystadenocarcinoma, Serous metabolism, Cell Survival drug effects, Neoplasm Grading, Forkhead Box Protein M1 metabolism, Forkhead Box Protein M1 antagonists & inhibitors, Ovarian Neoplasms drug therapy, Ovarian Neoplasms pathology, Ovarian Neoplasms metabolism, Antineoplastic Agents pharmacology, Apoptosis drug effects
- Abstract
Background: Genetic studies implicate the oncogenic transcription factor Forkhead Box M1 (FOXM1) as a potential therapeutic target in high-grade serous ovarian cancer (HGSOC). We evaluated the activity of different FOXM1 inhibitors in HGSOC cell models., Results: We treated HGSOC and fallopian tube epithelial (FTE) cells with a panel of previously reported FOXM1 inhibitors. Based on drug potency, efficacy, and selectivity, determined through cell viability assays, we focused on two compounds, NB-73 and NB-115 (NB compounds), for further investigation. NB compounds potently and selectively inhibited FOXM1 with lesser effects on other FOX family members. NB compounds decreased FOXM1 expression via targeting the FOXM1 protein by promoting its proteasome-mediated degradation, and effectively suppressed FOXM1 gene targets at both the protein and mRNA level. At the cellular level, NB compounds promoted apoptotic cell death. Importantly, while inhibition of apoptosis using a pan-caspase inhibitor rescued HGSOC cells from NB compound-induced cell death, it did not rescue FOXM1 protein degradation, supporting that FOXM1 protein loss from NB compound treatment is specific and not a general consequence of cytotoxicity. Drug washout studies indicated that FOXM1 reduction was retained for at least 72 h post-treatment, suggesting that NB compounds exhibit long-lasting effects in HGSOC cells. NB compounds effectively suppressed both two-dimensional and three-dimensional HGSOC cell colony formation at sub-micromolar concentrations. Finally, NB compounds exhibited synergistic activity with carboplatin in HGSOC cells., Conclusions: NB compounds are potent, selective, and efficacious inhibitors of FOXM1 in HGSOC cells and are worthy of further investigation as HGSOC therapeutics., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
9. ESR1 F404 Mutations and Acquired Resistance to Fulvestrant in ESR1-Mutant Breast Cancer.
- Author
-
Kingston B, Pearson A, Herrera-Abreu MT, Sim LX, Cutts RJ, Shah H, Moretti L, Kilburn LS, Johnson H, Macpherson IR, Ring A, Bliss JM, Hou Y, Toy W, Katzenellenbogen JA, Chandarlapaty S, and Turner NC
- Subjects
- Humans, Female, Fulvestrant pharmacology, Fulvestrant therapeutic use, Estrogen Receptor alpha genetics, Estrogen Receptor alpha metabolism, Mutation, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology, Circulating Tumor DNA genetics
- Abstract
Fulvestrant is used to treat patients with hormone receptor-positive advanced breast cancer, but acquired resistance is poorly understood. PlasmaMATCH Cohort A (NCT03182634) investigated the activity of fulvestrant in patients with activating ESR1 mutations in circulating tumor DNA (ctDNA). Baseline ESR1 mutations Y537S are associated with poor outcomes and Y537C with good outcomes. Sequencing of baseline and EOT ctDNA samples (n = 69) revealed 3/69 (4%) patients acquired novel ESR1 F404 mutations (F404L, F404I, and F404V), in cis with activating mutations. In silico modeling revealed that ESR1 F404 contributes to fulvestrant binding to estrogen receptor-alpha (ERα) through a pi-stacking bond, with mutations disrupting this bond. In vitro analysis demonstrated that single F404L, E380Q, and D538G models were less sensitive to fulvestrant, whereas compound mutations D538G + F404L and E380Q + F404L were resistant. Several oral ERα degraders were active against compound mutant models. We have identified a resistance mechanism specific to fulvestrant that can be targeted by treatments in clinical development., Significance: Novel F404 ESR1 mutations may be acquired to cause overt resistance to fulvestrant when combined with preexisting activating ESR1 mutations. Novel combinations of mutations in the ER ligand binding domain may cause drug-specific resistance, emphasizing the potential of similar drug-specific mutations to impact the efficacy of oral ER degraders in development. This article is featured in Selected Articles from This Issue, p. 201., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
10. Somatic estrogen receptor α mutations that induce dimerization promote receptor activity and breast cancer proliferation.
- Author
-
Irani S, Tan W, Li Q, Toy W, Jones C, Gadiya M, Marra A, Katzenellenbogen JA, Carlson KE, Katzenellenbogen BS, Karimi M, Segu Rajappachetty R, Del Priore IS, Reis-Filho JS, Shen Y, and Chandarlapaty S
- Subjects
- Female, Humans, Cell Proliferation, Dimerization, Estradiol pharmacology, Estradiol metabolism, Estrogen Receptor alpha genetics, Estrogen Receptor alpha metabolism, Ligands, Mutation, Breast Neoplasms genetics, Breast Neoplasms metabolism
- Abstract
Physiologic activation of estrogen receptor α (ERα) is mediated by estradiol (E2) binding in the ligand-binding pocket of the receptor, repositioning helix 12 (H12) to facilitate binding of coactivator proteins in the unoccupied coactivator binding groove. In breast cancer, activation of ERα is often observed through point mutations that lead to the same H12 repositioning in the absence of E2. Through expanded genetic sequencing of breast cancer patients, we identified a collection of mutations located far from H12 but nonetheless capable of promoting E2-independent transcription and breast cancer cell growth. Using machine learning and computational structure analyses, this set of mutants was inferred to act distinctly from the H12-repositioning mutants and instead was associated with conformational changes across the ERα dimer interface. Through both in vitro and in-cell assays of full-length ERα protein and isolated ligand-binding domain, we found that these mutants promoted ERα dimerization, stability, and nuclear localization. Point mutations that selectively disrupted dimerization abrogated E2-independent transcriptional activity of these dimer-promoting mutants. The results reveal a distinct mechanism for activation of ERα function through enforced receptor dimerization and suggest dimer disruption as a potential therapeutic strategy to treat ER-dependent cancers.
- Published
- 2024
- Full Text
- View/download PDF
11. Reprogramming of endothelial gene expression by tamoxifen inhibits angiogenesis and ERα-negative tumor growth.
- Author
-
Fébrissy C, Adlanmerini M, Péqueux C, Boudou F, Buscato M, Gargaros A, Gilardi-Bresson S, Boriak K, Laurell H, Fontaine C, Katzenellenbogen BS, Katzenellenbogen JA, Guillermet-Guibert J, Arnal JF, Metivier R, and Lenfant F
- Subjects
- Mice, Animals, Estrogen Receptor alpha genetics, Estrogen Receptor alpha metabolism, Endothelial Cells metabolism, Angiogenesis, Gene Expression, Endothelium metabolism, Cell Line, Tumor, Tumor Microenvironment genetics, Tamoxifen pharmacology, Neoplasms
- Abstract
Rationale : 17β-estradiol (E2) can directly promote the growth of ERα-negative cancer cells through activation of endothelial ERα in the tumor microenvironment, thereby increasing a normalized tumor angiogenesis. ERα acts as a transcription factor through its nuclear transcriptional AF-1 and AF-2 transactivation functions, but membrane ERα plays also an important role in endothelium. The present study aims to decipher the respective roles of these two pathways in ERα-negative tumor growth. Moreover, we delineate the actions of tamoxifen, a Selective Estrogen Receptor Modulator (SERM) in ERα-negative tumors growth and angiogenesis, since we recently demonstrated that tamoxifen impacts vasculature functions through complex modulation of ERα activity. Methods: ERα-negative B16K1 cancer cells were grafted into immunocompetent mice mutated for ERα-subfunctions and tumor growths were analyzed in these different models in response to E2 and/or tamoxifen treatment. Furthermore, RNA sequencings were analyzed in endothelial cells in response to these different treatments and validated by RT-qPCR and western blot. Results: We demonstrate that both nuclear and membrane ERα actions are required for the pro-tumoral effects of E2, while tamoxifen totally abrogates the E2-induced in vivo tumor growth, through inhibition of angiogenesis but promotion of vessel normalization. RNA sequencing indicates that tamoxifen inhibits the E2-induced genes, but also initiates a specific transcriptional program that especially regulates angiogenic genes and differentially regulates glycolysis, oxidative phosphorylation and inflammatory responses in endothelial cells. Conclusion: These findings provide evidence that tamoxifen specifically inhibits angiogenesis through a reprogramming of endothelial gene expression via regulation of some transcription factors, that could open new promising strategies to manage cancer therapies affecting the tumor microenvironment of ERα-negative tumors., Competing Interests: Competing Interests: The authors have declared that no competing interest exists., (© The author(s).)
- Published
- 2024
- Full Text
- View/download PDF
12. Endothelial ERα promotes glucose tolerance by enhancing endothelial insulin transport to skeletal muscle.
- Author
-
Sacharidou A, Chambliss K, Peng J, Barrera J, Tanigaki K, Luby-Phelps K, Özdemir İ, Khan S, Sirsi SR, Kim SH, Katzenellenbogen BS, Katzenellenbogen JA, Kanchwala M, Sathe AA, Lemoff A, Xing C, Hoyt K, Mineo C, and Shaul PW
- Subjects
- Animals, Female, Male, Mice, Endothelial Cells, Glucose, Muscle, Skeletal, Receptors, Estrogen, Estrogen Receptor alpha, Insulin
- Abstract
The estrogen receptor (ER) designated ERα has actions in many cell and tissue types that impact glucose homeostasis. It is unknown if these include mechanisms in endothelial cells, which have the potential to influence relative obesity, and processes in adipose tissue and skeletal muscle that impact glucose control. Here we show that independent of impact on events in adipose tissue, endothelial ERα promotes glucose tolerance by enhancing endothelial insulin transport to skeletal muscle. Endothelial ERα-deficient male mice are glucose intolerant and insulin resistant, and in females the antidiabetogenic actions of estradiol (E2) are absent. The glucose dysregulation is due to impaired skeletal muscle glucose disposal that results from attenuated muscle insulin delivery. Endothelial ERα activation stimulates insulin transcytosis by skeletal muscle microvascular endothelial cells. Mechanistically this involves nuclear ERα-dependent upregulation of vesicular trafficking regulator sorting nexin 5 (SNX5) expression, and PI3 kinase activation that drives plasma membrane recruitment of SNX5. Thus, coupled nuclear and non-nuclear actions of ERα promote endothelial insulin transport to skeletal muscle to foster normal glucose homeostasis., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
13. Targeting the oncogenic transcription factor FOXM1 to improve outcomes in all subtypes of breast cancer.
- Author
-
Katzenellenbogen BS, Guillen VS, and Katzenellenbogen JA
- Subjects
- Adult, Humans, Female, Cell Line, Tumor, Forkhead Box Protein M1 genetics, Treatment Outcome, Forkhead Transcription Factors genetics, Gene Expression Regulation, Neoplastic, Cell Proliferation, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms pathology, Antineoplastic Agents therapeutic use
- Abstract
FOXM1 (Forkhead box M1) is an oncogenic transcription factor that is greatly upregulated in breast cancer and many other cancers where it promotes tumorigenesis, and cancer growth and progression. It is expressed in all subtypes of breast cancer and is the factor most associated with risk of poor patient survival, especially so in triple negative breast cancer (TNBC). Thus, new approaches to inhibiting FOXM1 and its activities, and combination therapies utilizing FOXM1 inhibitors in conjunction with known cancer drugs that work together synergistically, could improve cancer treatment outcomes. Targeting FOXM1 might prove especially beneficial in TNBC where few targeted therapies currently exist, and also in suppressing recurrent advanced estrogen receptor (ER)-positive and HER2-positive breast cancers for which treatments with ER or HER2 targeted therapies that were effective initially are no longer beneficial. We present these perspectives and future directions in the context of what is known about FOXM1, its regulation, and its key roles in promoting cancer aggressiveness and metastasis, while being absent or very low in most normal non-regenerating adult tissues. We discuss new inhibitors of FOXM1 and highlight FOXM1 as an attractive target for controlling drug-resistant and difficult-to-suppress breast cancers, and how blocking FOXM1 might improve outcomes for patients with all subtypes of breast cancer., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
14. Coordinated activation of c-Src and FOXM1 drives tumor cell proliferation and breast cancer progression.
- Author
-
Nandi I, Smith HW, Sanguin-Gendreau V, Ji L, Pacis A, Papavasiliou V, Zuo D, Nam S, Attalla SS, Kim SH, Lusson S, Kuasne H, Fortier AM, Savage P, Martinez Ramirez C, Park M, Katzenellenbogen JA, Katzenellenbogen BS, and Muller WJ
- Subjects
- Humans, Female, Forkhead Box Protein M1 genetics, Forkhead Box Protein M1 metabolism, Cell Line, Tumor, Forkhead Transcription Factors metabolism, Cell Proliferation, Cell Cycle genetics, Gene Expression Regulation, Neoplastic, Breast Neoplasms pathology
- Abstract
Activation of the tyrosine kinase c-Src promotes breast cancer progression and poor outcomes, yet the underlying mechanisms are incompletely understood. Here, we have shown that deletion of c-Src in a genetically engineered model mimicking the luminal B molecular subtype of breast cancer abrogated the activity of forkhead box M1 (FOXM1), a master transcriptional regulator of the cell cycle. We determined that c-Src phosphorylated FOXM1 on 2 tyrosine residues to stimulate its nuclear localization and target gene expression. These included key regulators of G2/M cell-cycle progression as well as c-Src itself, forming a positive feedback loop that drove proliferation in genetically engineered and patient-derived models of luminal B-like breast cancer. Using genetic approaches and small molecules that destabilize the FOXM1 protein, we found that targeting this mechanism induced G2/M cell-cycle arrest and apoptosis, blocked tumor progression, and impaired metastasis. We identified a positive correlation between FOXM1 and c-Src expression in human breast cancer and show that the expression of FOXM1 target genes predicts poor outcomes and associates with the luminal B subtype, which responds poorly to currently approved therapies. These findings revealed a regulatory network centered on c-Src and FOXM1 that is a targetable vulnerability in aggressive luminal breast cancers.
- Published
- 2023
- Full Text
- View/download PDF
15. Effective combination treatments for breast cancer inhibition by FOXM1 inhibitors with other targeted cancer drugs.
- Author
-
Guillen VS, Ziegler Y, Gopinath C, Kumar S, Dey P, Plotner BN, Dawson NZ, Kim SH, Katzenellenbogen JA, and Katzenellenbogen BS
- Subjects
- Humans, Female, Forkhead Box Protein M1 genetics, Caspase 3 genetics, Neoplasm Recurrence, Local drug therapy, Cell Line, Tumor, Cell Proliferation, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms metabolism, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms metabolism, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use
- Abstract
Purpose: Few targeted treatment options currently exist for patients with advanced, often recurrent breast cancers, both triple-negative breast cancer (TNBC) and hormone receptor-positive breast cancer. Forkhead box M1 (FOXM1) is an oncogenic transcription factor that drives all cancer hallmarks in all subtypes of breast cancer. We previously developed small-molecule inhibitors of FOXM1 and to further exploit their potential as anti-proliferative agents, we investigated combining FOXM1 inhibitors with drugs currently used in the treatment of breast and other cancers and assessed the potential for enhanced inhibition of breast cancer., Methods: FOXM1 inhibitors alone and in combination with other cancer therapy drugs were assessed for their effects on suppression of cell viability and cell cycle progression, induction of apoptosis and caspase 3/7 activity, and changes in related gene expressions. Synergistic, additive, or antagonistic interactions were evaluated using ZIP (zero interaction potency) synergy scores and the Chou-Talalay interaction combination index., Results: The FOXM1 inhibitors displayed synergistic inhibition of proliferation, enhanced G2/M cell cycle arrest, and increased apoptosis and caspase 3/7 activity and associated changes in gene expression when combined with several drugs across different pharmacological classes. We found especially strong enhanced effectiveness of FOXM1 inhibitors in combination with drugs in the proteasome inhibitor class for ER-positive and TNBC cells and with CDK4/6 inhibitors (Palbociclib, Abemaciclib, and Ribociclib) in ER-positive cells., Conclusion: The findings suggest that the combination of FOXM1 inhibitors with several other drugs might enable dose reduction in both agents and provide enhanced efficacy in treatment of breast cancer., (© 2023. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
16. Stringing along the estrogen receptor to engage with DNA.
- Author
-
Katzenellenbogen JA
- Subjects
- Receptors, Estrogen genetics, DNA genetics
- Published
- 2023
- Full Text
- View/download PDF
17. [ 18 F]Tosyl fluoride as a versatile [ 18 F]fluoride source for the preparation of 18 F-labeled radiopharmaceuticals.
- Author
-
Zhou D, Chu W, Xu J, Schwarz S, and Katzenellenbogen JA
- Subjects
- Radiopharmaceuticals, Positron-Emission Tomography methods, Fluorine Radioisotopes, Fluorides, Fluorine Compounds
- Abstract
Positron emission tomography (PET) is an in vivo imaging technology that utilizes positron-emitting radioisotope-labeled compounds as PET radiotracers that are commonly used in clinic and in various research areas, including oncology, cardiology, and neurology. Fluorine-18 is the most widely used PET-radionuclide and commonly produced by proton bombardment of
18 O-enriched water in a cyclotron. The [18 F]fluoride thus obtained generally requires processing by azeotropic drying in order to completely remove H2 O before it can be used for nucleophilic radiofluorination. In general, the drying step is important in facilitating the radiofluorination reactions and the preparation of18 F-labeled PET radiotracers. In this communication, we have demonstrated the feasibility of using [18 F]tosyl fluoride ([18 F]TsF) as a versatile [18 F]fluoride source for radiofluorination to bypass the azeotropic drying step, and we have developed a continuous flow solid-phase radiosynthesis strategy to generate [18 F]TsF in a form that is excellent for radiofluorination. [18 F]TsF shows high reactivity in radiofluorination and provides the features suitable for preparing PET radiotracers on a small scale and exploring novel radiolabeling technologies. Thus, using [18 F]TsF as a [18 F]fluoride source is a promising strategy that facilitates radiofluorination and provides a convenient and efficient solution for the preparation of18 F-labeled radiopharmaceuticals that is well matched to the emerging trends in PET imaging technologies., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
18. Solid phase radiosynthesis of an olaparib derivative using 4-[ 18 F] fluorobenzoic acid and in vivo evaluation in breast and prostate cancer xenograft models for PARP-1 expression.
- Author
-
Xu J, Chen H, Rogers BE, Katzenellenbogen JA, and Zhou D
- Subjects
- Animals, Male, Humans, Poly (ADP-Ribose) Polymerase-1 metabolism, Radiopharmaceuticals chemistry, Tissue Distribution, Heterografts, Positron-Emission Tomography methods, Poly(ADP-ribose) Polymerase Inhibitors, Prostatic Neoplasms diagnostic imaging
- Abstract
Introduction: Solid-phase synthesis and conjugation reactions of acids and amines using coupling reagents are common in organic synthesis, but rare in
18 F radiochemistry. 4-[18 F]Fluorobenzoic acid (FBA) is a useful building block, but is seldom used directly with coupling reagents for the preparation of18 F radiopharmaceuticals. To overcome the inconveniences associated with using [18 F]FBA in conjugation reactions, we have developed a non-covalent solid-phase synthesis (SPS) strategy for the radiosynthesis of [18 F]PARPi, a derivative of olaparib as a Poly (ADP-ribose) polymerase-1 (PARP-1) radioligand., Methods: Fluoro-, bromo- and iodo-benzoic derivatives of olaparib were synthesized, and their PARP-1 affinities were measured using a recently developed cell culture-based competitive assay. To produce [18 F]PARPi, [18 F]FBA was radiosynthesized and purified using a cation-exchange cartridge, and then trapped by an anion-exchange resin cartridge, on which the solid-phase radiosynthesis was carried out to produce the desired product. [18 F]PARPi was evaluated in vivo in breast and prostate xenograft tumor models by microPET imaging, biodistribution and autoradiography., Results: The best derivatives of olaparib were identified as compound 4, 7 and 8. [18 F]4 ([18 F]PARPi) was radiosynthesized in high radiochemical yield, high molar activity and high radiochemical purity using this SPS strategy. The in vivo evaluation of [18 F]PARPi demonstrates the PARP-1 specific uptake of [18 F]PARPi in the animal models., Conclusions: This method is simple and efficient, having great potential for the synthesis of radiopharmaceuticals starting from [18 F]FBA or other radiolabeled aromatic acids. Using [18 F]PARPi prepared by this method, we demonstrated the promise of [18 F]PARPi in the nuclear imaging of PARP-1 expression., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
19. Estrogen Receptor Beta 1: A Potential Therapeutic Target for Female Triple Negative Breast Cancer.
- Author
-
Dey P, Wang A, Ziegler Y, Kumar S, Yan S, Kim SH, Katzenellenbogen JA, and Katzenellenbogen BS
- Subjects
- Humans, Female, Animals, Mice, Epithelial-Mesenchymal Transition genetics, Ligands, Cell Line, Tumor, Estrogen Receptor beta genetics, Estrogen Receptor beta metabolism, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms metabolism
- Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer characterized by the absence of estrogen receptor alpha, progesterone receptor, and HER2. These receptors often serve as targets in breast cancer treatment. As a result, TNBCs are difficult to treat and have a high propensity to metastasize to distant organs. For these reasons, TNBCs are responsible for over 50% of all breast cancer mortalities while only accounting for 15% to 20% of breast cancer cases. However, estrogen receptor beta 1 (ERβ1), an isoform of the ESR2 gene, has emerged as a potential therapeutic target in the treatment of TNBCs. Using an in vivo xenograft preclinical mouse model with human TNBC, we found that expression of ERβ1 significantly reduced both primary tumor growth and metastasis. Moreover, TNBCs with elevated levels of ERβ1 showed reduction in epithelial to mesenchymal transition markers and breast cancer stem cell markers, and increases in the expression of genes associated with inhibition of cancer cell invasiveness and metastasis, suggesting possible mechanisms underlying the antitumor activity of ERβ1. Gene expression analysis by quantitative polymerase chain reaction and RNA-seq revealed that treatment with chloroindazole, an ERβ-selective agonist ligand, often enhanced the suppressive activity of ERβ1 in TNBCs in vivo or in TNBC cells in culture, suggesting the potential utility of ERβ1 and ERβ ligand in improving TNBC treatment. The findings enable understanding of the mechanisms by which ERβ1 impedes TNBC growth, invasiveness, and metastasis and consideration of ways by which treatments involving ERβ might improve TNBC patient outcome., (© The Author(s) 2022. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
20. Endogenous DOPA inhibits melanoma through suppression of CHRM1 signaling.
- Author
-
Doepner M, Lee I, Natale CA, Brathwaite R, Venkat S, Kim SH, Wei Y, Vakoc CR, Capell BC, Katzenellenbogen JA, Katzenellenbogen BS, Feigin ME, and Ridky TW
- Abstract
Melanoma risk is 30 times higher in people with lightly pigmented skin versus darkly pigmented skin. Using primary human melanocytes representing the full human skin pigment continuum and preclinical melanoma models, we show that cell-intrinsic differences between dark and light melanocytes regulate melanocyte proliferative capacity and susceptibility to malignant transformation, independent of melanin and ultraviolet exposure. These differences result from dihydroxyphenylalanine (DOPA), a melanin precursor synthesized at higher levels in melanocytes from darkly pigmented skin. We used both high-throughput pharmacologic and genetic in vivo CRISPR screens to determine that DOPA limits melanocyte and melanoma cell proliferation by inhibiting the muscarinic acetylcholine receptor M
1 (CHRM1) signaling. Pharmacologic CHRM1 antagonism in melanoma leads to depletion of c-Myc and FOXM1, both of which are proliferation drivers associated with aggressive melanoma. In preclinical mouse melanoma models, pharmacologic inhibition of CHRM1 or FOXM1 inhibited tumor growth. CHRM1 and FOXM1 may be new therapeutic targets for melanoma.- Published
- 2022
- Full Text
- View/download PDF
21. FOXM1 regulates glycolysis and energy production in multiple myeloma.
- Author
-
Cheng Y, Sun F, Thornton K, Jing X, Dong J, Yun G, Pisano M, Zhan F, Kim SH, Katzenellenbogen JA, Katzenellenbogen BS, Hari P, and Janz S
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation, Forkhead Box Protein M1 genetics, Forkhead Box Protein M1 metabolism, Forkhead Transcription Factors genetics, Forkhead Transcription Factors metabolism, Gene Expression Regulation, Gene Expression Regulation, Neoplastic, Glycolysis, Humans, Mice, Transcription Factors metabolism, Multiple Myeloma genetics, Multiple Myeloma metabolism
- Abstract
The transcription factor, forkhead box M1 (FOXM1), has been implicated in the natural history and outcome of newly diagnosed high-risk myeloma (HRMM) and relapsed/refractory myeloma (RRMM), but the mechanism with which FOXM1 promotes the growth of neoplastic plasma cells is poorly understood. Here we show that FOXM1 is a positive regulator of myeloma metabolism that greatly impacts the bioenergetic pathways of glycolysis and oxidative phosphorylation (OxPhos). Using FOXM1-deficient myeloma cells as principal experimental model system, we find that FOXM1 increases glucose uptake, lactate output, and oxygen consumption in myeloma. We demonstrate that the novel 1,1-diarylethylene small-compound FOXM1 inhibitor, NB73, suppresses myeloma in cell culture and human-in-mouse xenografts using a mechanism that includes enhanced proteasomal FOXM1 degradation. Consistent with the FOXM1-stabilizing chaperone function of heat shock protein 90 (HSP90), the HSP90 inhibitor, geldanamycin, collaborates with NB73 in slowing down myeloma. These findings define FOXM1 as a key driver of myeloma metabolism and underscore the feasibility of targeting FOXM1 for new approaches to myeloma therapy and prevention., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
22. Estradiol promotes cell survival and induces Greb1 expression in granulosa cell tumors of the ovary through an ERα-dependent mechanism.
- Author
-
Cluzet V, Devillers MM, Petit F, Pierre A, Giton F, Airaud E, L'Hôte D, Leary A, Genestie C, Treilleux I, Mayeur A, Katzenellenbogen JA, Kim SH, Cohen-Tannoudji J, Chauvin S, and Guigon CJ
- Subjects
- Aged, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Estrogen Receptor alpha genetics, Estrogen Receptor alpha metabolism, Estrogen Receptor beta agonists, Estrogen Receptor beta genetics, Estrogen Receptor beta metabolism, Female, Gene Expression Regulation, Neoplastic, Granulosa Cell Tumor genetics, Granulosa Cell Tumor pathology, Humans, Middle Aged, Neoplasm Proteins genetics, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Receptors, Estrogen genetics, Receptors, Estrogen metabolism, Receptors, G-Protein-Coupled agonists, Receptors, G-Protein-Coupled genetics, Receptors, G-Protein-Coupled metabolism, Signal Transduction, Up-Regulation, Estradiol pharmacology, Estrogen Receptor alpha agonists, Granulosa Cell Tumor metabolism, Neoplasm Proteins metabolism, Ovarian Neoplasms metabolism
- Abstract
Granulosa cell tumor (GCT) is a form of ovarian tumor characterized by its tendency to recur years after surgical ablation. Little is known about the mechanisms involved in GCT development and progression. GCTs can produce estradiol (E2), but whether this hormone could play a role in this cancer through its nuclear receptors, i.e. ERα and ERβ, remains unknown. Here, we addressed this issue by cell-based and molecular studies on human GCTs and GCT cell lines. Importantly, we observed that E2 significantly increased the growth of GCT cells by promoting cell survival. The use of selective agonists of each type of receptor, together with Esr1 (ERα) or Esr2 (ERβ)-deleted GCT cells, revealed that E2 mediated its effects through ERα-dependent genomic mechanisms and ERβ/ERα-dependent extra-nuclear mechanisms. Notably, the expression of Greb1, a prototypical ER target gene, was dose-dependently upregulated by E2 specifically through ERα in GCT cells. Accordingly, using GCTs from patients, we found that GREB1 mRNA abundance was positively correlated to intra-tumoral E2 concentrations. Tissue microarray analyses showed that there were various combinations of ER expression in primary and recurrent GCTs, and that ERα expression persisted only in combination with ERβ in ~40% of recurrent tumors. Altogether, this study demonstrates that E2 can promote the progression of GCTs, with a clear dependence on ERα. In addition to demonstrating that GCTs can be classified as a hormone-related cancer, our results also highlight that the nature of ER forms present in recurrent GCTs could underlie the variable efficiency of endocrine therapies. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd., (© 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
23. Exploration of alcohol-enhanced Cu-mediated radiofluorination toward practical labeling.
- Author
-
Zhou D, Chu W, and Katzenellenbogen JA
- Subjects
- Copper chemistry, Fluorides, Isotope Labeling, Radiochemistry, Fluorine Radioisotopes chemistry, Radiopharmaceuticals chemistry
- Abstract
Copper-mediated nucleophilic radiofluorination using boronic precursors is a promising, general method to label aromatic compounds with [
18 F]fluoride. However, in various reports, large amounts of precursor (60 μmol) were needed to achieve high radiochemical conversions (RCCs), which is neither ideal nor practical for the preparation of18 F radiopharmaceuticals. To investigate this matter, we studied alcohol-enhanced Cu-mediated nucleophilic radiofluorination using a variety of model reactions in which we varied the concentration of [18 F]fluoride (no carrier added or isotope diluted) and the amount of precursor, base, and Cu(OTF)2 (Py)4 . We found that lower amounts of precursors (e.g., 15 μmol) could be used and that the amount of base (e.g., K2 CO3 or KHCO3 ) played a critical and limiting role in the labeling reactions. Greater than one-equivalent of base and sufficient amounts of precursors and Cu(OTf)2 (Py)4 were required to achieve good to high RCCs. The RCCs were also dependent on the overall concentration of the labeling reactions, with low reaction volumes and high concentrations of reagents being preferred. Our findings will help to improve the design of radiolabeling protocols using alcohol-enhanced copper-mediated radiofluorination of boronic precursors for the preparation of18 F labeled radiopharmaceuticals and other radiohalogen-labeled compounds., (© 2021 John Wiley & Sons, Ltd.)- Published
- 2022
- Full Text
- View/download PDF
24. Transcription Regulation and Genome Rewiring Governing Sensitivity and Resistance to FOXM1 Inhibition in Breast Cancer.
- Author
-
Ziegler Y, Guillen VS, Kim SH, Katzenellenbogen JA, and Katzenellenbogen BS
- Abstract
Forkhead box M1 (FOXM1), an oncogenic transcription factor associated with aggressiveness and highly expressed in many cancers, is an emerging therapeutic target. Using novel 1,1-diarylethylene-diammonium small molecule FOXM1 inhibitors, we undertook transcriptomic, protein, and functional analyses to identify mechanisms by which these compounds impact breast cancer growth and survival, and the changes that occur in estrogen receptor (ERα)-positive and triple negative breast cancer cells that acquire resistance upon long-term treatment with the inhibitors. In sensitive cells, these compounds regulated FOXM1 gene networks controlling cell cycle progression, DNA damage repair, and apoptosis. Resistant cells showed transcriptional alterations that reversed the expression of many genes in the FOXM1 network and rewiring that enhanced inflammatory signaling and upregulated HER2 or EGFR growth factor pathways. ERα-positive breast cancer cells that developed resistance showed greatly reduced ERα levels and responsiveness to fulvestrant and a 10-fold increased sensitivity to lapatinib, suggesting that targeting rewired processes in the resistant state may provide benefits and prolong anticancer effectiveness. Improved understanding of how FOXM1 inhibitors suppress breast cancer and how cancer cells can defeat their effectiveness and acquire resistance should be helpful in directing further studies to move these agents towards translation into the clinic.
- Published
- 2021
- Full Text
- View/download PDF
25. Determination of molar activity of [ 18 F]fluoride by HPLC via sulfonyl derivatization.
- Author
-
Zhou D, Schwarz S, and Katzenellenbogen JA
- Abstract
The molar activity of [
18 F]fluoride was determined by HPLC of sulfonyl fluorides, which have high UV absorbance and are formed exclusively from sulfonyl chlorides and [18 F]fluoride in aqueous solution. The measurable limit of sulfonyl fluorides is as low as 0.1 ppm, allowing measurements up to the theoretical molar activity of [18 F]fluoride. The method was validated with standard solutions of fluoride, and provides a convenient way to measure molar activity of [18 F]fluoride using only HPLC without specialized equipment., (Copyright © 2021 Elsevier Ltd. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
26. Pathway Preferential Estrogens Prevent Hepatosteatosis Due to Ovariectomy and High-Fat Diets.
- Author
-
Zuo Q, Chen KL, Arredondo Eve A, Liu YJ, Kim SH, Katzenellenbogen BS, Katzenellenbogen JA, and Madak-Erdogan Z
- Subjects
- Animals, Body Weight drug effects, Collagen metabolism, Estrogen Receptor alpha metabolism, Estrogens pharmacology, Fatty Liver complications, Fatty Liver genetics, Hep G2 Cells, Hepatocytes metabolism, Humans, Inflammation pathology, Ligands, Lipid Metabolism, Liver metabolism, Metabolomics, Mice, Inbred C57BL, Mice, Obese, Obesity complications, Obesity genetics, Organ Size drug effects, Transcriptome genetics, Weight Gain, Mice, Diet, High-Fat, Estrogens therapeutic use, Fatty Liver etiology, Fatty Liver prevention & control, Ovariectomy
- Abstract
About 20-30% of premenopausal women have metabolic syndrome, and the number is almost double in postmenopausal women, and these women have an increased risk of hepatosteatosis. Postmenopausal women with metabolic syndrome are often treated with hormone replacement therapy (HRT), but estrogens in currently available HRTs increase the risk of breast and endometrial cancers and Cardiovascular Disease. Therefore, there is a critical need to find safer alternatives to HRT to improve postmenopausal metabolic health. Pathway preferential estrogen 1 (PaPE-1) is a novel estrogen receptor ligand that has been shown to favorably affect metabolic tissues without adverse effects on reproductive tissues. In this study, we have examined the effects of PaPE-1 on metabolic health, in particular, examining its effects on the liver transcriptome and on plasma metabolites in two different mouse models: diet-induced obesity (DIO) and leptin-deficient (ob/ob) mice. PaPE-1 significantly decreased liver weight and lipid accumulation in both DIO and ob/ob models and lowered the expression of genes associated with fatty acid metabolism and collagen deposition. In addition, PaPE-1 significantly increased the expression of mitochondrial genes, particularly ones associated with the electron transport chain, suggesting an increase in energy expenditure. Integrated pathway analysis using transcriptomics and metabolomics data showed that PaPE-1 treatment lowered inflammation, collagen deposition, and pathways regulating fatty acid metabolism and increased metabolites associated with glutathione metabolism. Overall, our findings support a beneficial metabolic role for PaPE-1 and suggest that PaPE-1 may protect postmenopausal women from fatty liver disease without increasing reproductive cancer risk.
- Published
- 2021
- Full Text
- View/download PDF
27. Defining the Energetic Basis for a Conformational Switch Mediating Ligand-Independent Activation of Mutant Estrogen Receptors in Breast Cancer.
- Author
-
Mayne CG, Toy W, Carlson KE, Bhatt T, Fanning SW, Greene GL, Katzenellenbogen BS, Chandarlapaty S, Katzenellenbogen JA, and Tajkhorshid E
- Subjects
- Breast Neoplasms genetics, Breast Neoplasms metabolism, Crystallography, X-Ray, Female, Humans, Ligands, Models, Molecular, Protein Binding, Receptors, Estrogen genetics, Receptors, Estrogen metabolism, Tumor Cells, Cultured, Breast Neoplasms pathology, Mutation, Protein Conformation, Receptors, Estrogen chemistry
- Abstract
Although most primary estrogen receptor (ER)-positive breast cancers respond well to endocrine therapies, many relapse later as metastatic disease due to endocrine therapy resistance. Over one third of these are associated with mutations in the ligand-binding domain (LBD) that activate the receptor independent of ligand. We have used an array of advanced computational techniques rooted in molecular dynamics simulations, in concert with and validated by experiments, to characterize the molecular mechanisms by which specific acquired somatic point mutations give rise to ER constitutive activation. By comparing structural and energetic features of constitutively active mutants and ligand-bound forms of ER-LBD with unliganded wild-type (WT) ER, we characterize a spring force originating from strain in the Helix 11-12 loop of WT-ER, opposing folding of Helix 12 into the active conformation and keeping WT-ER off and disordered, with the ligand-binding pocket open for rapid ligand binding. We quantify ways in which this spring force is abrogated by activating mutations that latch (Y537S) or relax (D538G) the folded form of the loop, enabling formation of the active conformation without ligand binding. We also identify a new ligand-mediated hydrogen-bonding network that stabilizes the active, ligand-bound conformation of WT-ER LBD, and similarly stabilizes the active conformation of the ER mutants in the hormone-free state. IMPLICATIONS: Our investigations provide deep insight into the energetic basis for the structural mechanisms of receptor activation through mutation, exemplified here with ER in endocrine-resistant metastatic breast cancers, with potential application to other dysregulated receptor signaling due to driver mutations., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
28. Dual-mechanism estrogen receptor inhibitors.
- Author
-
Min J, Nwachukwu JC, Min CK, Njeri JW, Srinivasan S, Rangarajan ES, Nettles CC, Sanabria Guillen V, Ziegler Y, Yan S, Carlson KE, Hou Y, Kim SH, Novick S, Pascal BD, Houtman R, Griffin PR, Izard T, Katzenellenbogen BS, Katzenellenbogen JA, and Nettles KW
- Subjects
- Breast Neoplasms metabolism, Breast Neoplasms pathology, Crystallography, X-Ray, Female, Humans, Protein Binding, Protein Conformation, Structure-Activity Relationship, Tumor Cells, Cultured, Breast Neoplasms drug therapy, Estrogen Antagonists chemistry, Estrogen Antagonists pharmacology, Estrogen Receptor alpha antagonists & inhibitors
- Abstract
Efforts to improve estrogen receptor-α (ER)-targeted therapies in breast cancer have relied upon a single mechanism, with ligands having a single side chain on the ligand core that extends outward to determine antagonism of breast cancer growth. Here, we describe inhibitors with two ER-targeting moieties, one of which uses an alternate structural mechanism to generate full antagonism, freeing the side chain to independently determine other critical properties of the ligands. By combining two molecular targeting approaches into a single ER ligand, we have generated antiestrogens that function through new mechanisms and structural paradigms to achieve antagonism. These dual-mechanism ER inhibitors (DMERIs) cause alternate, noncanonical structural perturbations of the receptor ligand-binding domain (LBD) to antagonize proliferation in ER-positive breast cancer cells and in allele-specific resistance models. Our structural analyses with DMERIs highlight marked differences from current standard-of-care, single-mechanism antiestrogens. These findings uncover an enhanced flexibility of the ER LBD through which it can access nonconsensus conformational modes in response to DMERI binding, broadly and effectively suppressing ER activity., Competing Interests: Competing interest statement: J.A.K. is a founder and stockholder of Radius Health Inc. and a consultant of Celcuity Inc. B.S.K. is a consultant of Celcuity Inc.
- Published
- 2021
- Full Text
- View/download PDF
29. Radiosynthesis and Evaluation of Talazoparib and Its Derivatives as PARP-1-Targeting Agents.
- Author
-
Zhou D, Chen H, Mpoy C, Afrin S, Rogers BE, Garbow JR, Katzenellenbogen JA, and Xu J
- Abstract
Poly (ADP-ribose) polymerase-1 (PARP-1) is a critical enzyme in the DNA repair process and the target of several FDA-approved inhibitors. Several of these inhibitors have been radiolabeled for non-invasive imaging of PARP-1 expression or targeted radiotherapy of PARP-1 expressing tumors. In particular, derivatives of olaparib and rucaparib, which have reduced trapping potency by PARP-1 compared to talazoparib, have been radiolabeled for these purposes. Here, we report the first radiosynthesis of [
18 F]talazoparib and its in vitro and in vivo evaluation. Talazoparib ( 3a″ ) and its bromo- or iodo-derivatives were synthesized as racemic mixtures ( 3a , 3b and 3c ), and these compounds exhibit high affinity to PARP-1 ( Ki for talazoparib ( 3a″ ): 0.65 ± 0.07 nM; 3a : 2.37 ± 0.56 nM; 3b : 1.92 ± 0.41 nM; 3c : 1.73 ± 0.43 nM; known PARP-1 inhibitor Olaparib: 1.87 ± 0.10 nM; non-PARP-1 compound Raclopride: >20,000 nM) in a competitive binding assay using a tritium-labeled PARP-1 radioligand [3 H]WC-DZ for screening. [18 F]Talazoparib ( 3a″ ) was radiosynthesized via a multiple-step procedure with good radiochemical and chiral purities (98%) and high molar activity (28 GBq/μmol). The preliminary biodistribution studies in the murine PC-3 tumor model showed that [18 F]talazoparib had a good level of tumor uptake that persisted for over 8 h (3.78 ± 0.55 %ID/gram at 4 h and 4.52 ± 0.32 %ID/gram at 8 h). These studies show the potential for the bromo- and iodo- derivatives for PARP-1 targeted radiotherapy studies using therapeutic radionuclides.- Published
- 2021
- Full Text
- View/download PDF
30. Alleviation of extensive visual pathway dysfunction by a remyelinating drug in a chronic mouse model of multiple sclerosis.
- Author
-
Sekyi MT, Lauderdale K, Atkinson KC, Golestany B, Karim H, Feri M, Soto JS, Diaz C, Kim SH, Cilluffo M, Nusinowitz S, Katzenellenbogen JA, and Tiwari-Woodruff SK
- Subjects
- Animals, Evoked Potentials, Visual drug effects, Inflammation pathology, Mice, Mice, Inbred C57BL, Multiple Sclerosis, Nerve Degeneration pathology, Azo Compounds pharmacology, Encephalomyelitis, Autoimmune, Experimental pathology, Naphthalenes pharmacology, Remyelination drug effects, Visual Pathways drug effects, Visual Pathways pathology
- Abstract
Visual deficits are among the most prevalent symptoms in patients with multiple sclerosis (MS). To understand deficits in the visual pathway during MS and potential treatment effects, we used experimental autoimmune encephalomyelitis (EAE), the most commonly used animal model of MS. The afferent visual pathway was assessed in vivo using optical coherence tomography (OCT), electroretinography (ERG), and visually evoked cortical potentials (VEPs). Inflammation, demyelination, and neurodegeneration were examined by immunohistochemistry ex vivo. In addition, an immunomodulatory, remyelinating agent, the estrogen receptor β ligand chloroindazole (IndCl), was tested for its therapeutic potential in the visual pathway. EAE produced functional deficits in visual system electrophysiology, including suppression of ERG and VEP waveform amplitudes and increased signal latencies. Therapeutic IndCl rescued overall visual system latency by VEP but had little impact on amplitude or ERG findings relative to vehicle. Faster VEP conduction in IndCl-treated mice was associated with enhanced myelin basic protein signal in all visual system structures examined. IndCl preserved retinal ganglion cells (RGCs) and oligodendrocyte density in the prechiasmatic white matter, but similar retinal nerve fiber layer thinning by OCT was noted in vehicle and IndCl-treated mice. Although IndCl differentially attenuated leukocyte and astrocyte staining signal throughout the structures analyzed, axolemmal varicosities were observed in all visual fiber tracts of mice with EAE irrespective of treatment, suggesting impaired axonal energy homeostasis. These data support incomplete functional recovery of VEP amplitude with IndCl, as fiber tracts displayed persistent axon pathology despite remyelination-induced decreases in latencies, evidenced by reduced optic nerve g-ratio in IndCl-treated mice. Although additional studies are required, these findings demonstrate the dynamics of visual pathway dysfunction and disability during EAE, along with the importance of early treatment to mitigate EAE-induced axon damage., (© 2020 The Authors. Brain Pathology published by John Wiley & Sons Ltd on behalf of International Society of Neuropathology.)
- Published
- 2021
- Full Text
- View/download PDF
31. Association of PET-based estradiol-challenge test for breast cancer progesterone receptors with response to endocrine therapy.
- Author
-
Dehdashti F, Wu N, Ma CX, Naughton MJ, Katzenellenbogen JA, and Siegel BA
- Subjects
- Breast Neoplasms drug therapy, Female, Humans, Postmenopause, Prospective Studies, Receptors, Estrogen genetics, Receptors, Progesterone genetics, Breast Neoplasms metabolism, Estradiol therapeutic use, Positron-Emission Tomography methods, Receptors, Estrogen metabolism, Receptors, Progesterone metabolism
- Abstract
Estrogen receptor (ER) testing of breast cancer imperfectly predicts response to endocrine therapy (ET). We hypothesize that a brief estradiol challenge will increase tumor progesterone receptor (PgR) levels only in tumors with functional ER. In this prospective, phase 2, single-center, single-arm trial (NCT02455453), we report the association of response to ET with change in tumor uptake of the progestin analog, 21-[
18 F]fluorofuranylnorprogesterone (FFNP), before and after a one-day estradiol challenge. In 43 postmenopausal women with advanced ER+ breast cancer, we show a post-challenge increase in tumor FFNP uptake only in 28 subjects with clinical benefit from ET (responders), but not in 15 without clinical benefit (nonresponders) (p < 0.0001), indicating 100% sensitivity and specificity. We further show significantly longer survival (p < 0.0001) in the responding subjects. Our results demonstrate that change in tumor FFNP uptake after estradiol challenge is highly predictive of response to ET in women with ER+ breast cancer.- Published
- 2021
- Full Text
- View/download PDF
32. Contrasting activities of estrogen receptor beta isoforms in triple negative breast cancer.
- Author
-
Yan S, Dey P, Ziegler Y, Jiao X, Kim SH, Katzenellenbogen JA, and Katzenellenbogen BS
- Subjects
- Humans, Prognosis, Proto-Oncogene Mas, RNA, Messenger, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Estrogen Receptor beta genetics, Estrogen Receptor beta metabolism, Protein Isoforms genetics, Protein Isoforms metabolism, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms genetics
- Abstract
Purpose: Triple negative breast cancer (TNBC), an aggressive subtype of breast cancer, lacks the three major receptors for predicting outcome or targeting therapy. Hence, our aim was to evaluate the potential of estrogen receptor beta (ERβ) as a possible endocrine therapy target in TNBC., Methods: The expression and prognostic effect of ERβ isoforms were analyzed using TCGA breast tumor data, and the expression of ERβ isoform mRNA and protein in TNBC cell lines was assayed. Endogenous ERβ2 and ERβ5 were knocked down with siRNA, and ERβ2, ERβ5, and ERβ1 were upregulated using a doxycycline-inducible lentiviral system. Cell proliferation, migration and invasion, and specific gene expressions were evaluated., Results: ERβ2 and ERβ5 were the predominant endogenous forms of ERβ in TNBC tumors and cell lines. High ERβ2 predicted worse clinical outcome. Knockdown of endogenous ERβ2/ERβ5 in cell lines suppressed proliferation, migration and invasion, and downregulated proto-oncogene survivin expression. ERβ2/ERβ5 upregulation did the reverse, increasing survivin and these cell activities. ERβ1 was barely detectable in TNBC cell lines, but its upregulation reduced survivin, increased tumor suppressor expression (E-cadherin and cystatins), and suppressed proliferation, migration and invasion in both ligand-independent and dependent manners, suggesting the possible translational benefit of ERβ ligands., Conclusions: ERβ2/ERβ5 and ERβ1 exhibit sharply contrasting activities in TNBC cells. Our findings imply that delineating the absolute amounts and relative ratios of the different ERβ isoforms might have prognostic and therapeutic relevance, and could enable better selection of optimal approaches for treatment of this often aggressive form of breast cancer.
- Published
- 2021
- Full Text
- View/download PDF
33. William C. Eckelman: An anchor of stability with a quiet voice that nurtured a new field.
- Author
-
Katzenellenbogen JA, Kilbourn MR, Coenen HH, and Volkert W
- Subjects
- History, 20th Century, History, 21st Century, Nuclear Medicine
- Published
- 2021
- Full Text
- View/download PDF
34. The quest for improving the management of breast cancer by functional imaging: The discovery and development of 16α-[ 18 F]fluoroestradiol (FES), a PET radiotracer for the estrogen receptor, a historical review.
- Author
-
Katzenellenbogen JA
- Subjects
- Humans, Animals, Radioactive Tracers, History, 20th Century, History, 21st Century, Female, Breast Neoplasms diagnostic imaging, Breast Neoplasms metabolism, Positron-Emission Tomography methods, Receptors, Estrogen metabolism, Receptors, Estrogen antagonists & inhibitors, Estradiol analogs & derivatives, Estradiol metabolism
- Abstract
Introduction: 16α-[
18 F]Fluoroestradiol (FES), a PET radiotracer for the estrogen receptor (ER) in breast cancer, was the first receptor-targeted PET radiotracer for oncology and is continuing to prove its value in clinical research, antiestrogen development, and breast cancer care. The story of its conception, design, evaluation and use in clinical studies parallels the evolution of the whole field of receptor-targeted radiotracers, one greatly influenced by the research and intellectual contributions of William C. Eckelman., Methods and Results: The development of methods for efficient production of fluorine-18, for conversion of [18 F]fluoride ion into chemically reactive form, and for its rapid and efficient incorporation into suitable estrogen precursor molecules at high molar activity, were all methodological underpinnings required for the preparation of FES. FES binds to ER with very high affinity, and its in vivo uptake by ER-dependent target tissues in animal models was efficient and selective, findings that preceded its use for PET imaging in patients with breast cancer., Advances in Knowledge and Implications for Patient Care: Comparisons between ER levels measured by FES-PET imaging of breast tumors with tissue-specimen ER quantification by IHC and other methods show that imaging provided improved prediction of benefit from endocrine therapies. Serial imaging of ER by FES-PET, before and after dosing patients with antiestrogens, is used to determine the efficacious dose for established antiestrogens and to facilitate clinical development of new ER antagonists. Beyond FES imaging, PET-based hormone challenge tests, which evaluate the functional status of ER by monitoring rapid changes in tumor metabolic or transcriptional activity after a brief estrogen challenge, provide highly sensitive and selective predictions of whether or not there will be a favorable response to endocrine therapies. There is sufficient interest in the clinical applications of FES that FDA approval is being sought for its wider use in breast cancer., Conclusions: FES was the first PET probe for a receptor in cancer, and its development and clinical applications in breast cancer parallel the conceptual evolution of the whole field of receptor-binding radiotracers., (Copyright © 2020 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
35. A mutant form of ERα associated with estrogen insensitivity affects the coupling between ligand binding and coactivator recruitment.
- Author
-
Li Y, Coons LA, Houtman R, Carlson KE, Martin TA, Mayne CG, Melchers D, Jefferson TB, Ramsey JT, Katzenellenbogen JA, and Korach KS
- Subjects
- Binding Sites genetics, Drug Resistance genetics, Estrogen Receptor alpha chemistry, Estrogen Receptor alpha metabolism, Estrogens pharmacology, Gene Expression Regulation, HEK293 Cells, Hep G2 Cells, Humans, Kinetics, Ligands, Molecular Dynamics Simulation, Nuclear Receptor Coactivator 1 metabolism, Nuclear Receptor Coactivator 3 metabolism, Protein Binding, Protein Domains, Estrogen Receptor alpha genetics, Estrogens metabolism, Mutation, Missense, Nuclear Receptor Coactivator 1 genetics, Nuclear Receptor Coactivator 3 genetics
- Abstract
A homozygous missense mutation in the gene encoding the estrogen receptor α (ERα) was previously identified in a female patient with estrogen insensitivity syndrome. We investigated the molecular features underlying the impaired transcriptional response of this mutant (ERα-Q375H) and four other missense mutations at this position designed to query alternative mechanisms. The identity of residue 375 greatly affected the sensitivity of the receptor to agonists without changing the ligand binding affinity. Instead, the mutations caused changes in the affinity of coactivator binding and alterations in the balance of coactivator and corepressor recruitment. Comparisons among the transcriptional regulatory responses of these six ERα genotypes to a set of ER agonists showed that both steric and electrostatic factors contributed to the functional deficits in gene regulatory activity of the mutant ERα proteins. ERα-coregulator peptide binding in vitro and RIME (rapid immunoprecipitation mass spectrometry of endogenous) analysis in cells showed that the degree of functional impairment paralleled changes in receptor-coregulator binding interactions. These findings uncover coupling between ligand binding and coregulator recruitment that affects the potency rather than the efficacy of the receptor response without substantially altering ligand binding affinity. This highlights a molecular mechanism for estrogen insensitivity syndrome involving mutations that perturb a bidirectional allosteric coupling between ligand binding and coregulator binding that determines receptor transcriptional output., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
36. Suppression of Tumor Growth, Metastasis, and Signaling Pathways by Reducing FOXM1 Activity in Triple Negative Breast Cancer.
- Author
-
Dey P, Wang A, Ziegler Y, Kim SH, El-Ashry D, Katzenellenbogen JA, and Katzenellenbogen BS
- Abstract
Metastasis-related complications account for the overwhelming majority of breast cancer mortalities. Triple negative breast cancer (TNBC), the most aggressive breast cancer subtype, has a high propensity to metastasize to distant organs, leading to poor patient survival. The forkhead transcription factor, FOXM1, is especially upregulated and overexpressed in TNBC and is known to regulate multiple signaling pathways that control many key cancer properties, including proliferation, invasiveness, stem cell renewal, and therapy resistance, making FOXM1 a critical therapeutic target for TNBC. In this study, we test the effectiveness of a novel class of 1,1-diarylethylene FOXM1 inhibitory compounds in suppressing TNBC cell migration, invasion, and metastasis using in vitro cell culture and in vivo tumor models. We show that these compounds inhibit the motility and invasiveness of TNBC MDA-MB-231 and DT28 cells, along with reducing the expression of important epithelial to mesenchymal transition (EMT) associated genes. Further, orthotopic tumor studies in NOD-SCID-gamma (NSG) mice demonstrate that these compounds reduce FOXM1 expression and suppress TNBC tumor growth as well as distant metastasis. Gene expression and protein analyses confirm the decreased levels of EMT factors and FOXM1-regulated target genes in tumors and metastatic lesions in the inhibitor-treated animals. The findings suggest that these FOXM1 suppressive compounds may have therapeutic potential in treating triple negative breast cancer, with the aim of reducing tumor progression and metastatic outgrowth.
- Published
- 2020
- Full Text
- View/download PDF
37. PET Imaging Agents (FES, FFNP, and FDHT) for Estrogen, Androgen, and Progesterone Receptors to Improve Management of Breast and Prostate Cancers by Functional Imaging.
- Author
-
Katzenellenbogen JA
- Abstract
Many breast and prostate cancers are driven by the action of steroid hormones on their cognate receptors in primary tumors and in metastases, and endocrine therapies that inhibit hormone production or block the action of these receptors provide clinical benefit to many but not all of these cancer patients. Because it is difficult to predict which individuals will be helped by endocrine therapies and which will not, positron emission tomography (PET) imaging of estrogen receptor (ER) and progesterone receptor (PgR) in breast cancer, and androgen receptor (AR) in prostate cancer can provide useful, often functional, information on the likelihood of endocrine therapy response in individual patients. This review covers our development of three PET imaging agents, 16α-[
18 F]fluoroestradiol (FES) for ER, 21-[18 F]fluoro-furanyl-nor-progesterone (FFNP) for PgR, and 16β-[18 F]fluoro-5α-dihydrotestosterone (FDHT) for AR, and the evolution of their clinical use. For these agents, the pathway from concept through development tracks with an emerging understanding of critical performance criteria that is needed for successful PET imaging of these low-abundance receptor targets. Progress in the ongoing evaluation of what they can add to the clinical management of breast and prostate cancers reflects our increased understanding of these diseases and of optimal strategies for predicting the success of clinical endocrine therapies.- Published
- 2020
- Full Text
- View/download PDF
38. Suppression of breast cancer metastasis and extension of survival by a new antiestrogen in a preclinical model driven by mutant estrogen receptors.
- Author
-
Laws MJ, Ziegler Y, Shahoei SH, Dey P, Kim SH, Yasuda M, Park BH, Nettles KW, Katzenellenbogen JA, Nelson ER, and Katzenellenbogen BS
- Subjects
- Animals, Antineoplastic Agents pharmacology, Apoptosis drug effects, Breast Neoplasms genetics, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Proliferation drug effects, Female, Humans, Ketones pharmacology, Liver Neoplasms genetics, Liver Neoplasms metabolism, Liver Neoplasms secondary, MCF-7 Cells, Mice, Mice, Inbred NOD, Mice, SCID, Survival Rate, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Adamantane analogs & derivatives, Adamantane pharmacology, Breast Neoplasms drug therapy, Estrogen Receptor Modulators pharmacology, Liver Neoplasms drug therapy, Mutation, Receptors, Estrogen genetics
- Abstract
Purpose: Many human breast tumors become resistant to endocrine therapies and recur due to estrogen receptor (ERα) mutations that convey constitutive activity and a more aggressive phenotype. Here, we examined the effectiveness of a novel adamantyl antiestrogen, K-07, in suppressing the growth of breast cancer metastases containing the two most frequent ER-activating mutations, Y537S and D538G, and in extending survival in a preclinical metastatic cancer model., Methods: MCF7 breast cancer cells expressing luciferase and Y537S or D538G ER were injected into NOD-SCID-gamma female mice, and animals were treated orally with the antiestrogen K-07 or control vehicle. Comparisons were also made with the antiestrogen Fulvestrant. The development of metastases was monitored by in vivo bioluminescence imaging with phenotypic characterization of the metastases in liver and lung by immunohistochemical and biochemical analyses., Results: These breast cancer cells established metastases in liver and lung, and K-07 treatment reduced the metastatic burden. Mice treated with K-07 also survived much longer. By day 70, only 28% of vehicle-treated mice with mutant ER metastases were alive, whereas all K-07-treated D538G and Y537S mice were still alive. K-07 also markedly reduced the level of metastatic cell ER and the expression of ER-regulated genes., Conclusion: The antiestrogen K-07 can reduce in vivo metastasis of breast cancers and extend host survival in this preclinical model driven by constitutively active mutant ERs, suggesting that this compound may be suitable for further translational examination of its efficacy in suppression of metastasis in breast cancers containing constitutively active mutant ERs.
- Published
- 2020
- Full Text
- View/download PDF
39. Long-Term Follow-Up and Treatment of a Female With Complete Estrogen Insensitivity.
- Author
-
Brakta S, Chorich LP, Kim HG, Coons LA, Katzenellenbogen JA, Hall JE, Korach KS, and Layman LC
- Subjects
- Adolescent, Adult, Amenorrhea complications, Animals, COS Cells, Chlorocebus aethiops, Female, Follow-Up Studies, Hep G2 Cells, Humans, Ovarian Cysts complications, Ovarian Cysts genetics, Ovarian Cysts therapy, Puberty, Delayed complications, Puberty, Delayed genetics, Puberty, Delayed therapy, Young Adult, Amenorrhea genetics, Amenorrhea therapy, Drug Resistance genetics, Estrogen Receptor alpha genetics
- Abstract
Context: We previously reported the first female with a causative ESR1 gene variant, who exhibited absent puberty and high estrogens. At age 15 years, she presented with lower abdominal pain, absent breast development, primary amenorrhea, and multicystic ovaries. The natural history of complete estrogen insensitivity (CEI) in women is unknown., Objective: The purpose of this report is to present the neuroendocrine phenotype of CEI, identify potential ligands, and determine the effect of targeted treatment., Design: We have characterized gonadotropin pulsatility and followed this patient's endocrine profile and bone density over 8 years. Seventy-five different compounds were tested for transactivation of the variant receptor. A personalized medicine approach was tailored to our patient., Setting: Academic medical center., Patient or Other Participants: A 24-year-old adopted white female with CEI., Intervention(s): The patient was treated with diethylstilbestrol (DES) for approximately 2.5 years., Main Outcome Measure(s): Induction of secondary sexual characteristics., Results: Luteinizing hormone (LH) pulse studies demonstrated normal pulsatile LH secretion, elevated mean LH, and mildly elevated mean follicle-stimulating hormone (FSH) in the presence of markedly increased estrogens. DES transactivated the variant ESR1 in vitro. However, DES treatment did not induce secondary sexual characteristics in our patient., Conclusions: Treatment with DES was not successful in our patient. She remains hypoestrogenic despite the presence of ovarian cysts with a hypoestrogenic vaginal smear, absent breast development, and low bone mineral mass. Findings suggest additional receptor mechanistic actions are required to elicit clinical hormone responses., (© Endocrine Society 2020. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
40. The tissue-specific effects of different 17β-estradiol doses reveal the key sensitizing role of AF1 domain in ERα activity.
- Author
-
Fontaine C, Buscato M, Vinel A, Giton F, Raymond-Letron I, Kim SH, Katzenellenbogen BS, Katzenellenbogen JA, Gourdy P, Milon A, Flouriot G, Ohlsson C, Lenfant F, and Arnal JF
- Subjects
- Animals, Bone and Bones drug effects, Cell Line, Tumor, Cholesterol blood, Estradiol blood, Female, Mice, Inbred C57BL, Protein Domains, Structure-Activity Relationship, Uterus drug effects, Vagina drug effects, Estradiol pharmacology, Estrogen Receptor alpha chemistry, Estrogen Receptor alpha metabolism, Organ Specificity
- Abstract
17β-Estradiol (E2) action can be mediated by the full-length estrogen receptor alpha (ERα66), and also by the AF1 domain-deficient ERα (ERα46) isoform, but their respective sensitivity to E2 is essentially unknown. We first performed a dose response study using subcutaneous home-made pellets mimicking either metestrus, proestrus or a pharmacological doses of E2, which resulted in plasma concentrations around 3, 30 and 600 pM, respectively. Analysis of the uterus, vagina and bone after chronic exposure to E2 demonstrated dose-dependent effects, with a maximal response reached at the proestrus-dose in wild type mice expressing mainly ERα66. In contrast, in transgenic mice harbouring only an ERα deleted in AF1, these effects of E2 were either strongly shifted rightward (10-100-fold) and/or attenuated, depending on the tissue studied. Finally, experiments in different cell lines transfected with ERα66 or ERα46 also delineated varying profiles of ERα AF1 sensitivity to E2. Altogether, this work emphasizes the importance of dose in the tissue-specific actions of E2 and demonstrates the key sensitizing role of AF1 in ERα activity., Competing Interests: Declaration of competing interest The authors have nothing to disclose., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
41. Suppression of FOXM1 activities and breast cancer growth in vitro and in vivo by a new class of compounds.
- Author
-
Ziegler Y, Laws MJ, Sanabria Guillen V, Kim SH, Dey P, Smith BP, Gong P, Bindman N, Zhao Y, Carlson K, Yasuda MA, Singh D, Li Z, El-Ashry D, Madak-Erdogan Z, Katzenellenbogen JA, and Katzenellenbogen BS
- Abstract
The transcription factor FOXM1 is upregulated and overexpressed in aggressive, therapy-resistant forms of hormone receptor-positive and triple negative breast cancers, and is associated with less good patient survival. FOXM1 signaling is also a key driver in many other cancers. Here, we identify a new class of compounds effective in suppressing FOXM1 activity in breast cancers, and displaying good potency for antitumor efficacy. The compounds bind directly to FOXM1 and alter its proteolytic sensitivity, reduce the cellular level of FOXM1 protein by a proteasome- dependent process, and suppress breast cancer cell proliferation and cell cycle progression and increase apoptosis. RNA-seq and gene set enrichment analyses indicate that the compounds decrease expression of FOXM1-regulated genes and suppress gene ontologies under FOXM1 regulation. Several compounds have favorable pharmacokinetic properties and show good tumor suppression in preclinical breast tumor models. These compounds may be suitable for further clinical evaluation in targeting aggressive breast cancers driven by FOXM1., Competing Interests: Competing interestsJ.A.K., B.S.K., and S.H.K. are coinventors on a Provisional Application filed by the University of Illinois to cover the compounds described in this paper. The other authors declare no competing interests., (© The Author(s) 2019.)
- Published
- 2019
- Full Text
- View/download PDF
42. Differential Actions of Estrogen Receptor α and β via Nongenomic Signaling in Human Prostate Stem and Progenitor Cells.
- Author
-
Majumdar S, Rinaldi JC, Malhotra NR, Xie L, Hu DP, Gauntner TD, Grewal HS, Hu WY, Kim SH, Katzenellenbogen JA, Kasper S, and Prins GS
- Subjects
- Caveolin 1 metabolism, Cells, Cultured, Histone Methyltransferases metabolism, Humans, MAP Kinase Signaling System, Male, Phosphorylation, Phosphotransferases metabolism, Prostate cytology, Estrogen Receptor alpha metabolism, Estrogen Receptor beta metabolism, Neoplastic Stem Cells metabolism, Prostate metabolism
- Abstract
Human prostate stem and progenitor cells express estrogen receptor (ER)α and ERβ and exhibit proliferative responses to estrogens. In this study, membrane-initiated estrogen signaling was interrogated in human prostate stem/progenitor cells enriched from primary epithelial cultures and stem-like cell lines from benign and cancerous prostates. Subcellular fractionation and proximity ligation assays localized ERα and ERβ to the cell membrane with caveolin-1 interactions. Exposure to 17β-estradiol (E2) for 15 to 60 minutes led to sequential phosphorylation of signaling molecules in MAPK and AKT pathways, IGF1 receptor, epidermal growth factor receptor, and ERα, thus documenting an intact membrane signalosome that activates diverse downstream cascades. Treatment with an E2-dendrimer conjugate or ICI 182,870 validated E2-mediated actions through membrane ERs. Overexpression and knockdown of ERα or ERβ in stem/progenitor cells identified pathway selectivity; ERα preferentially activated AKT, whereas ERβ selectively activated MAPK cascades. Furthermore, prostate cancer stem-like cells expressed only ERβ, and brief E2 exposure activated MAPK but not AKT cascades. A gene subset selectively regulated by nongenomic E2 signaling was identified in normal prostate progenitor cells that includes BGN, FOSB, FOXQ1, and MAF. Membrane-initiated E2 signaling rapidly modified histone methyltransferases, with MLL1 cleavage observed downstream of phosphorylated AKT and EZH2 phosphorylation downstream of MAPK signaling, which may jointly modify histones to permit rapid gene transcription. Taken together, the present findings document ERα and ERβ membrane-initiated signaling in normal and cancerous human prostate stem/progenitor cells with differential engagement of downstream effectors. These signaling pathways influence normal prostate stem/progenitor cell homeostasis and provide novel therapeutic sites to target the elusive prostate cancer stem cell population., (Copyright © 2019 Endocrine Society.)
- Published
- 2019
- Full Text
- View/download PDF
43. Diffusion tensor imaging identifies aspects of therapeutic estrogen receptor β ligand-induced remyelination in a mouse model of multiple sclerosis.
- Author
-
Atkinson KC, Lee JB, Hasselmann JPC, Kim SH, Drew A, Soto J, Katzenellenbogen JA, Harris NG, Obenaus A, and Tiwari-Woodruff SK
- Subjects
- Animals, Corpus Callosum drug effects, Cuprizone, Demyelinating Diseases chemically induced, Demyelinating Diseases drug therapy, Diffusion Tensor Imaging, Disease Models, Animal, Female, Indazoles pharmacology, Magnetic Resonance Imaging, Male, Mice, Multiple Sclerosis chemically induced, Multiple Sclerosis drug therapy, Neuroprotective Agents pharmacology, Corpus Callosum diagnostic imaging, Demyelinating Diseases diagnostic imaging, Estrogen Receptor beta agonists, Indazoles therapeutic use, Multiple Sclerosis diagnostic imaging, Neuroprotective Agents therapeutic use, Remyelination drug effects
- Abstract
Diffusion tensor imaging (DTI) has been shown to detect white matter degeneration in multiple sclerosis (MS), a neurodegenerative autoimmune disease that presents with diffuse demyelination of the central nervous system. However, the utility of DTI in evaluating therapeutic remyelination has not yet been well-established. Here, we assessed the ability of DTI to distinguish between remyelination and neuroprotection following estrogen receptor β ligand (Indazole chloride, IndCl) treatment, which has been previously shown to stimulate functional remyelination, in the cuprizone (CPZ) diet mouse model of MS. Adult C57BL/6 J male and female mice received a normal diet (control), demyelination-inducing CPZ diet (9wkDM), or CPZ diet followed by two weeks of a normal diet (i.e., remyelination period) with either IndCl (RM + IndCl) or vehicle (RM + Veh) injections. We evaluated tissue microstructure of the corpus callosum utilizing in vivo and ex vivo DTI and immunohistochemistry (IHC) for validation. Compared to control mice, the 9wkDM group showed decreased fractional anisotropy (FA), increased radial diffusivity (RD), and no changes in axial diffusivity (AD) both in vivo and ex vivo. Meanwhile, RM + IndCl groups showed increased FA and decreased RD ex vivo compared to the RM + Veh group, in accordance with the evidence of remyelination by IHC. In conclusion, the DTI technology used in the present study can identify some changes in myelination and is a valuable translational tool for evaluating MS pathophysiology and therapeutic efficacy., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
44. Free Fatty Acids Rewire Cancer Metabolism in Obesity-Associated Breast Cancer via Estrogen Receptor and mTOR Signaling.
- Author
-
Madak-Erdogan Z, Band S, Zhao YC, Smith BP, Kulkoyluoglu-Cotul E, Zuo Q, Santaliz Casiano A, Wrobel K, Rossi G, Smith RL, Kim SH, Katzenellenbogen JA, Johnson ML, Patel M, Marino N, Storniolo AMV, and Flaws JA
- Subjects
- Breast Neoplasms blood, Breast Neoplasms complications, CD36 Antigens blood, Cell Line, Tumor, Chromatin metabolism, Female, Gene Expression Regulation, Neoplastic, Humans, Obesity blood, Obesity complications, Phosphatidylinositol 3-Kinases metabolism, Postmenopause, Breast Neoplasms metabolism, Estrogen Receptor alpha metabolism, Fatty Acids, Nonesterified metabolism, Obesity metabolism, Signal Transduction, TOR Serine-Threonine Kinases metabolism
- Abstract
Obesity is a risk factor for postmenopausal estrogen receptor alpha (ERα)-positive (ER
+ ) breast cancer. Molecular mechanisms underlying factors from plasma that contribute to this risk and how these mechanisms affect ERα signaling have yet to be elucidated. To identify such mechanisms, we performed whole metabolite and protein profiling in plasma samples from women at high risk for breast cancer, which led us to focus on factors that were differentially present in plasma of obese versus nonobese postmenopausal women. These studies, combined with in vitro assays, identified free fatty acids (FFA) as circulating plasma factors that correlated with increased proliferation and aggressiveness in ER+ breast cancer cells. FFAs activated both the ERα and mTOR pathways and rewired metabolism in breast cancer cells. Pathway preferential estrogen-1 (PaPE-1), which targets ERα and mTOR signaling, was able to block changes induced by FFA and was more effective in the presence of FFA. Collectively, these data suggest a role for obesity-associated gene and metabolic rewiring in providing new targetable vulnerabilities for ER+ breast cancer in postmenopausal women. Furthermore, they provide a basis for preclinical and clinical trials where the impact of agents that target ERα and mTOR signaling cross-talk would be tested to prevent ER+ breast cancers in obese postmenopausal women. SIGNIFICANCE: These findings show that obesity-associated changes in certain blood metabolites rewire metabolic programs in cancer cells, influence mammary epithelial cell tumorigenicity and aggressiveness, and increase breast cancer risk., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
45. Pocket similarity identifies selective estrogen receptor modulators as microtubule modulators at the taxane site.
- Author
-
Lo YC, Cormier O, Liu T, Nettles KW, Katzenellenbogen JA, Stearns T, and Altman RB
- Subjects
- Antineoplastic Agents pharmacology, Binding Sites, Cell Death drug effects, Cell Line, Tumor drug effects, Cell Proliferation drug effects, Humans, Ligands, Microtubule Proteins drug effects, Models, Molecular, Paclitaxel pharmacology, Tubulin drug effects, Tumor Microenvironment, Antineoplastic Agents chemistry, Bridged-Ring Compounds chemistry, Bridged-Ring Compounds pharmacology, Selective Estrogen Receptor Modulators chemistry, Selective Estrogen Receptor Modulators metabolism, Taxoids chemistry, Taxoids pharmacology, Tubulin chemistry, Tubulin Modulators chemistry, Tubulin Modulators metabolism
- Abstract
Taxanes are a family of natural products with a broad spectrum of anticancer activity. This activity is mediated by interaction with the taxane site of beta-tubulin, leading to microtubule stabilization and cell death. Although widely used in the treatment of breast cancer and other malignancies, existing taxane-based therapies including paclitaxel and the second-generation docetaxel are currently limited by severe adverse effects and dose-limiting toxicity. To discover taxane site modulators, we employ a computational binding site similarity screen of > 14,000 drug-like pockets from PDB, revealing an unexpected similarity between the estrogen receptor and the beta-tubulin taxane binding pocket. Evaluation of nine selective estrogen receptor modulators (SERMs) via cellular and biochemical assays confirms taxane site interaction, microtubule stabilization, and cell proliferation inhibition. Our study demonstrates that SERMs can modulate microtubule assembly and raises the possibility of an estrogen receptor-independent mechanism for inhibiting cell proliferation.
- Published
- 2019
- Full Text
- View/download PDF
46. Analogues of ERβ ligand chloroindazole exert immunomodulatory and remyelinating effects in a mouse model of multiple sclerosis.
- Author
-
Karim H, Kim SH, Lauderdale K, Lapato AS, Atkinson K, Yasui N, Yamate-Morgan H, Sekyi M, Katzenellenbogen JA, and Tiwari-Woodruff SK
- Subjects
- Animals, Cell Proliferation drug effects, Cell Survival drug effects, Cytokines metabolism, Estrogen Receptor beta metabolism, Female, Ligands, Male, Mice, Oligodendroglia metabolism, Oligodendroglia pathology, Corpus Callosum metabolism, Corpus Callosum pathology, Estrogen Receptor beta agonists, Immunologic Factors pharmacology, Indazoles pharmacology, Multiple Sclerosis drug therapy, Multiple Sclerosis metabolism, Multiple Sclerosis pathology, Remyelination drug effects
- Abstract
Pharmaceutical agents currently approved for the treatment of multiple sclerosis reduce relapse rates, but do not reverse or prevent neurodegeneration nor initiate myelin repair. The highly selective estrogen receptor (ER) β ligand chloroindazole (IndCl) shows particular promise promoting both remyelination while reducing inflammatory cytokines in the central nervous system of mice with experimental autoimmune encephalomyelitis. To optimize these benefits, we developed and screened seven novel IndCl analogues for their efficacy in promoting primary oligodendrocyte (OL) progenitor cell survival, proliferation, and differentiation in vitro by immunohistochemistry. Two analogues, IndCl-o-chloro and IndCl-o-methyl, induced proliferation and differentiation equivalent to IndCl and were selected for subsequent in vivo evaluation for their impact on clinical disease course, white matter pathology, and inflammation. Both compounds ameliorated disease severity, increased mature OLs, and improved overall myelination in the corpus callosum and white matter tracts of the spinal cord. These effects were accompanied by reduced production of the OL toxic molecules interferon-γ and chemokine (C-X-C motif) ligand, CXCL10 by splenocytes with no discernable effect on central nervous system-infiltrating leukocyte numbers, while IndCl-o-methyl also reduced peripheral interleukin (IL)-17. In addition, expression of the chemokine CXCL1, which is associated with developmental oligodendrogenesis, was upregulated by IndCl and both analogues. Furthermore, callosal compound action potential recordings from analogue-treated mice demonstrated a larger N1 component amplitude compared to vehicle, suggesting more functionally myelinated fibers. Thus, the o-Methyl and o-Chloro IndCl analogues represent a class of ERβ ligands that offer significant remyelination and neuroprotection as well as modulation of the immune system; hence, they appear appropriate to consider further for therapeutic development in multiple sclerosis and other demyelinating diseases.
- Published
- 2019
- Full Text
- View/download PDF
47. Antagonists for Constitutively Active Mutant Estrogen Receptors: Insights into the Roles of Antiestrogen-Core and Side-Chain.
- Author
-
Sharma A, Toy W, Guillen VS, Sharma N, Min J, Carlson KE, Mayne CG, Lin S, Sabio M, Greene G, Katzenellenbogen BS, Chandarlapaty S, and Katzenellenbogen JA
- Subjects
- Binding Sites, Cell Proliferation drug effects, Down-Regulation, Estradiol analogs & derivatives, Estradiol chemistry, Estrogen Antagonists chemical synthesis, Estrogen Antagonists chemistry, Estrogen Receptor Modulators chemical synthesis, Estrogen Receptor Modulators chemistry, Estrogen Receptor alpha genetics, Humans, Ligands, MCF-7 Cells, Molecular Docking Simulation, Molecular Structure, Mutation, Phenols chemical synthesis, Phenols chemistry, Estrogen Antagonists pharmacology, Estrogen Receptor Modulators pharmacology, Estrogen Receptor alpha antagonists & inhibitors, Phenols pharmacology
- Abstract
A major risk for patients having estrogen receptor α (ERα)-positive breast cancer is the recurrence of drug-resistant metastases after initial successful treatment with endocrine therapies. Recent studies have implicated a number of activating mutations in the ligand-binding domain of ERα that stabilize the agonist conformation as a prominent mechanism for this acquired resistance. There are several critical gaps in our knowledge regarding the specific pharmacophore requirements of an antagonist that could effectively inhibit all or most of the different mutant ERs. To address this, we screened various chemotypes for blocking mutant ER-mediated transcriptional signaling and identified RU58668 as a model compound that contains structural elements that support potent ligand-induced inhibition of mutant ERs. We designed and synthesized a focused library of novel antagonists and probed how small and large perturbations in different ligand structural regions influenced inhibitory activity on individual mutant ERs in breast cancer cells. Effective inhibition derives from both nonpolar and moderately polar motifs in a multifunctional side chain of the antagonists, with the nature of the ligand core making important contributions by increasing the potency of ligands possessing similar types of side chains. Some of our new antagonists potently blocked the transcriptional activity of the three most common mutant ERs (L536R, Y537S, D538G) and inhibited mutant ER-mediated cell proliferation. Supported by our molecular modeling, these studies provide new insights into the role of specific components, involving both the ligand core and multifunctional side chain, in suppressing wild-type and mutant ER-mediated transcription and breast cancer cell proliferation.
- Published
- 2018
- Full Text
- View/download PDF
48. The SERM/SERD bazedoxifene disrupts ESR1 helix 12 to overcome acquired hormone resistance in breast cancer cells.
- Author
-
Fanning SW, Jeselsohn R, Dharmarajan V, Mayne CG, Karimi M, Buchwalter G, Houtman R, Toy W, Fowler CE, Han R, Lainé M, Carlson KE, Martin TA, Nowak J, Nwachukwu JC, Hosfield DJ, Chandarlapaty S, Tajkhorshid E, Nettles KW, Griffin PR, Shen Y, Katzenellenbogen JA, Brown M, and Greene GL
- Subjects
- Estrogen Receptor alpha genetics, Female, Fulvestrant pharmacology, Humans, Indoles chemistry, Ligands, MCF-7 Cells, Mutant Proteins metabolism, Mutation genetics, Piperazines pharmacology, Protein Binding drug effects, Protein Domains, Protein Structure, Secondary, Pyridines pharmacology, Raloxifene Hydrochloride pharmacology, Selective Estrogen Receptor Modulators chemistry, Structure-Activity Relationship, Tamoxifen pharmacology, Breast Neoplasms pathology, Drug Resistance, Neoplasm drug effects, Estrogen Receptor alpha chemistry, Indoles pharmacology, Selective Estrogen Receptor Modulators pharmacology
- Abstract
Acquired resistance to endocrine therapy remains a significant clinical burden for breast cancer patients. Somatic mutations in the ESR1 (estrogen receptor alpha (ERα)) gene ligand-binding domain (LBD) represent a recognized mechanism of acquired resistance. Antiestrogens with improved efficacy versus tamoxifen might overcome the resistant phenotype in ER +breast cancers. Bazedoxifene (BZA) is a potent antiestrogen that is clinically approved for use in hormone replacement therapies. We found that BZA possesses improved inhibitory potency against the Y537S and D538G ERα mutants compared to tamoxifen and has additional inhibitory activity in combination with the CDK4/6 inhibitor palbociclib. In addition, comprehensive biophysical and structural biology studies show BZA's selective estrogen receptor degrading (SERD) properties that override the stabilizing effects of the Y537S and D538G ERα mutations., Competing Interests: SF, RJ, VD, MK, WT, CF, RH, ML, KC, TM, JN, JN, DH, ET, KN, PG, YS, JK, MB No competing interests declared, CM Employee and shareholder of Celgene. GB Employee and shareholder of Celgene, RH Employee of PamGene International, SC Dr. Chandaralapaty does not receive financial benefit from any data derived from this publication. However, he has received research funds from Daiichi Sankyo and ad hoc consulting honoraria from Novartis, Sermonix, Context Therapeutics, and Lilly. GG GLG does not have direct financial competing interests in regards to bazedoxifene. However, has received research funds as well as consulting fees from Pfizer and Sermonix Pharmaceuticals and is a member of the scientific advisory board of Olema Pharmaceuticals., (© 2018, Fanning et al.)
- Published
- 2018
- Full Text
- View/download PDF
49. Selective Nonnuclear Estrogen Receptor Activation Decreases Stroke Severity and Promotes Functional Recovery in Female Mice.
- Author
-
Selvaraj UM, Zuurbier KR, Whoolery CW, Plautz EJ, Chambliss KL, Kong X, Zhang S, Kim SH, Katzenellenbogen BS, Katzenellenbogen JA, Mineo C, Shaul PW, and Stowe AM
- Subjects
- Animals, Behavior, Animal drug effects, Female, Infarction, Middle Cerebral Artery metabolism, Infarction, Middle Cerebral Artery pathology, Mice, Neuronal Plasticity, Ovariectomy, Severity of Illness Index, Stroke metabolism, Stroke pathology, Stroke physiopathology, Uterus drug effects, Estradiol pharmacology, Estrogens pharmacology, Infarction, Middle Cerebral Artery physiopathology, Psychomotor Performance drug effects, Receptors, Estrogen metabolism, Recovery of Function
- Abstract
Estrogens provide neuroprotection in animal models of stroke, but uterotrophic effects and cancer risk limit translation. Classic estrogen receptors (ERs) serve as transcription factors, whereas nonnuclear ERs govern numerous cell processes and exert beneficial cardiometabolic effects without uterine or breast cancer growth in mice. Here, we determined how nonnuclear ER stimulation with pathway-preferential estrogen (PaPE)-1 affects stroke outcome in mice. Ovariectomized female mice received vehicle, estradiol (E2), or PaPE-1 before and after transient middle cerebral artery occlusion (tMCAo). Lesion severity was assessed with MRI, and poststroke motor function was evaluated through 2 weeks after tMCAo. Circulating, spleen, and brain leukocyte subpopulations were quantified 3 days after tMCAo by flow cytometry, and neurogenesis and angiogenesis were evaluated histologically 2 weeks after tMCAo. Compared with vehicle, E2 and PaPE-1 reduced infarct volumes at 3 days after tMCAo, though only PaPE-1 reduced leukocyte infiltration into the ischemic brain. Unlike E2, PaPE-1 had no uterotrophic effect. Both interventions had negligible effect on long-term poststroke neuronal or vascular plasticity. All mice displayed a decline in motor performance at 2 days after tMCAo, and vehicle-treated mice did not improve thereafter. In contrast, E2 and PaPE-1 treatment afforded functional recovery at 6 days after tMCAo and beyond. Thus, the selective activation of nonnuclear ER by PaPE-1 decreased stroke severity and improved functional recovery in mice without undesirable uterotrophic effects. The beneficial effects of PaPE-1 are also associated with attenuated neuroinflammation in the brain. PaPE-1 and similar molecules may warrant consideration as efficacious ER modulators providing neuroprotection without detrimental effects on the uterus or cancer risk.
- Published
- 2018
- Full Text
- View/download PDF
50. Preliminary evaluation of a novel 18 F-labeled PARP-1 ligand for PET imaging of PARP-1 expression in prostate cancer.
- Author
-
Zhou D, Xu J, Mpoy C, Chu W, Kim SH, Li H, Rogers BE, and Katzenellenbogen JA
- Subjects
- Animals, Autoradiography, Cell Line, Tumor, Humans, Isotope Labeling, Ligands, Male, Mice, Prostatic Neoplasms pathology, Radiochemistry, Radiopharmaceuticals chemistry, Radiopharmaceuticals pharmacokinetics, Tissue Distribution, Fluorine Radioisotopes, Gene Expression Regulation, Neoplastic, Poly (ADP-Ribose) Polymerase-1 metabolism, Positron-Emission Tomography methods, Prostatic Neoplasms diagnostic imaging, Prostatic Neoplasms metabolism, Radiopharmaceuticals metabolism
- Abstract
Introduction: Poly (ADP-ribose) polymerase-1 (PARP-1) plays many roles in prostate cancer (PC), such as mediating DNA damage repair, transcriptional regulation and nuclear hormone receptor signaling. Because of this, PARP-1 has been targeted for therapy in PC, and non-invasive imaging of PARP-1 could help predict which patients are likely to respond to such therapy. Several PARP-1 positron emission tomography (PET) imaging agents have been developed and show promise for imaging PARP-1 expression in breast, brain, and lung cancer in small animals, but not as yet in prostate cancer. [
18 F]WC-DZ-F is an analogue of [18 F]FluorThanatrace (FTT) and [125 I]KX1, which are well-established PARP-1 ligands for measuring PARP-1 expression. Herein, we evaluated the potential of [18 F]WC-DZ-F for the imaging PARP-1 expression in PC., Methods: [18 F]WC-DZ-F was synthesized by a two-step sequence. [18 F]WC-DZ-F was evaluated by in vitro uptake studies in PC-3 cells and by in vivo biodistribution and microPET imaging using PC-3 tumor xenografts. Ex vivo autoradiography of PC-3 tumors after microPET imaging was also performed., Results: [18 F]WC-DZ-F has high, PARP-1-specific uptake in PC-3 cells. In the microPET imaging study, [18 F]WC-DZ-F accumulated in PC-3 xenograft tumors over 2 h, and the uptake was significantly reduced by blocking with olaparib. PC-3 tumors were clearly visualized in microPET images, and the imaging results were further confirmed by autoradiography of PC-3 tumors ex vivo. In the biodistribution study [18 F]WC-DZ-F washed out quickly from most tissues within 2 h, except for the liver in which the uptake was not blockable by olaparib., Conclusions: We synthesized a novel PARP-1 radioligand, [18 F]WC-DZ-F. The preliminary evaluation of [18 F]WC-DZ-F indicates that it is a suitable PET imaging agent for measuring PARP-1 expression in prostate cancer and should be applicable to other types of cancers., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.