1
|
Thiemann M, Zimmermann M, Diederich C, Zhan H, Lebedev M, Pletz J, Baumgarten J, Handke M, Müsken M, Breinbauer R, Krasteva-Christ G, Zanin E, Empting M, Schiedel M, Kunick C, Blankenfeldt W. From Bones to Bugs: Structure-Based Development of Raloxifene-Derived Pathoblockers That Inhibit Pyocyanin Production in Pseudomonas aeruginosa. J Med Chem 2025; 68:7390-7420. [PMID: 40156840 PMCID: PMC11998002 DOI: 10.1021/acs.jmedchem.4c03065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025]
Abstract
The human pathogen Pseudomonas aeruginosa is particularly notorious for its multiple resistance mechanisms. A new concept for anti-infectives is the "pathoblocker" approach, which targets virulence factors to disarm rather than kill pathogens and thus attenuates the development of resistance. Based on the estrogen receptor modulator raloxifene, which had previously been identified as a potential biosynthesis inhibitor of the virulence factor pyocyanin via in silico screening, analogues have been developed as pathoblockers against P. aeruginosa. These compounds reduce the production of pyocyanin by binding to the phenazine biosynthesis enzyme PhzB. Structure-activity relationships (SAR) were explored using nano differential scanning fluorimetry, isothermal titration calorimetry, and 12 X-ray cocrystal structures. Compared to raloxifene, congener 20c shows a 60-fold lower affinity for the human estrogen receptor with a 15-fold increase in pyocyanin inhibitory activity. The comprehensive structural information gathered in this study paves the way for the development of improved pathoblockers with increased potency and selectivity.
Collapse
Affiliation(s)
- Marie Thiemann
- Institute
of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany
- Helmholtz
Centre for Infection Research (HZI), Inhoffenstraße 7, 38124 Braunschweig, Germany
- Center
of Pharmaceutical Engineering (PVZ), Technische
Universität Braunschweig, Franz-Liszt-Straße 35a, 38106 Braunschweig, Germany
| | - Moritz Zimmermann
- Institute
of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany
- Helmholtz
Centre for Infection Research (HZI), Inhoffenstraße 7, 38124 Braunschweig, Germany
- Center
of Pharmaceutical Engineering (PVZ), Technische
Universität Braunschweig, Franz-Liszt-Straße 35a, 38106 Braunschweig, Germany
| | - Christina Diederich
- Helmholtz
Centre for Infection Research (HZI), Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Huilin Zhan
- Helmholtz-Institute
for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for
Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Department
of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
- Faculty of
Medicine, Institute for Anatomy and Cell Biology & Center for
Gender-specific Biology and Medicine (CGBM), Saarland University, Kirrbergerstr. 100, 66424 Homburg, Saar, Germany
- PharmaScienceHub
(PSH), 66123 Saarbrücken, Germany
| | - Mikhail Lebedev
- Department
Biologie, Friedrich-Alexander-Universität
Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Jakob Pletz
- Institute
of Organic Chemistry, Graz University of
Technology, Stremayrgasse 9, 8010 Graz, Austria
| | - Janosch Baumgarten
- Institute
of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany
- Helmholtz
Centre for Infection Research (HZI), Inhoffenstraße 7, 38124 Braunschweig, Germany
- Center
of Pharmaceutical Engineering (PVZ), Technische
Universität Braunschweig, Franz-Liszt-Straße 35a, 38106 Braunschweig, Germany
| | - Maria Handke
- Institute
of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany
- Helmholtz
Centre for Infection Research (HZI), Inhoffenstraße 7, 38124 Braunschweig, Germany
- Center
of Pharmaceutical Engineering (PVZ), Technische
Universität Braunschweig, Franz-Liszt-Straße 35a, 38106 Braunschweig, Germany
| | - Mathias Müsken
- Helmholtz
Centre for Infection Research (HZI), Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Rolf Breinbauer
- Institute
of Organic Chemistry, Graz University of
Technology, Stremayrgasse 9, 8010 Graz, Austria
| | - Gabriela Krasteva-Christ
- Faculty of
Medicine, Institute for Anatomy and Cell Biology & Center for
Gender-specific Biology and Medicine (CGBM), Saarland University, Kirrbergerstr. 100, 66424 Homburg, Saar, Germany
- PharmaScienceHub
(PSH), 66123 Saarbrücken, Germany
| | - Esther Zanin
- Department
Biologie, Friedrich-Alexander-Universität
Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Martin Empting
- Helmholtz-Institute
for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for
Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Department
of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
- PharmaScienceHub
(PSH), 66123 Saarbrücken, Germany
- Partner
Site Hannover-Braunschweig, German Centre
for Infection Research (DZIF), 38124 Braunschweig, Germany
| | - Matthias Schiedel
- Institute
of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany
- Center
of Pharmaceutical Engineering (PVZ), Technische
Universität Braunschweig, Franz-Liszt-Straße 35a, 38106 Braunschweig, Germany
| | - Conrad Kunick
- Institute
of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany
- Center
of Pharmaceutical Engineering (PVZ), Technische
Universität Braunschweig, Franz-Liszt-Straße 35a, 38106 Braunschweig, Germany
| | - Wulf Blankenfeldt
- Helmholtz
Centre for Infection Research (HZI), Inhoffenstraße 7, 38124 Braunschweig, Germany
- Institute
of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany
| |
Collapse
|
2
|
Williams DR, Taylor L, Miter GA, Sheiman JL, Wallace JM, Allen MR, Kohler R, Medeiros C. Synthesis Studies and the Evaluation of C 6 Raloxifene Derivatives. ACS Med Chem Lett 2024; 15:879-884. [PMID: 38894928 PMCID: PMC11181480 DOI: 10.1021/acsmedchemlett.4c00078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Methodology is described for the synthesis of C6 derivatives of raloxifene, a prescribed drug for the treatment and prevention of osteoporosis. Studies have explored the incorporation of electron-withdrawing substituents at C6 of the benzothiophene core. Efficient processes are also examined to introduce hydrogen bond donor and acceptor functionality. Raloxifene derivatives are evaluated with in vitro testing to determine estrogen receptor (ER) binding affinity and gene expression in MC3T3 cells.
Collapse
Affiliation(s)
- David R. Williams
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Levin Taylor
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Gabriel A. Miter
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Johnathan L. Sheiman
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Joseph M. Wallace
- Department
of Biomedical Engineering, Indiana University-Purdue
University, Indianapolis, Indiana 46202, United States
| | - Matthew R. Allen
- Department
of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202 United States
| | - Rachel Kohler
- Department
of Biomedical Engineering, Indiana University-Purdue
University, Indianapolis, Indiana 46202, United States
| | - Claudia Medeiros
- Department
of Biomedical Engineering, Indiana University-Purdue
University, Indianapolis, Indiana 46202, United States
| |
Collapse
|
3
|
Lunghini F, Marcou G, Azam P, Bonachera F, Enrici MH, Van Miert E, Varnek A. Endocrine disruption: the noise in available data adversely impacts the models' performance. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2021; 32:111-131. [PMID: 33461329 DOI: 10.1080/1062936x.2020.1864468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/11/2020] [Indexed: 06/12/2023]
Abstract
This paper is devoted to the analysis of available experimental data and preparation of predictive models for binding affinity of molecules with respect to two nuclear receptors involved in endocrine disruption (ED): the oestrogen (ER) and the androgen (AR) receptors. The ED-relevant data were retrieved from multiple sources, including the CERAPP, CoMPARA, and the Tox21 projects as well as ChEMBL and PubChem databases. Data analysis performed with the help of generative topographic mapping revealed the problem of low agreement between experimental values from different sources. Collected data were used to train both classification models for ER and AR binding activities and regression models for relative binding affinity (RBA) and median inhibition concentration (IC50). These models displayed relatively poor performance in classification (sensitivities ER = 0.34, AR = 0.49) and in regression (determination coefficient r 2 for the RBA and IC50 models in external validation varied from 0.44 to 0.76). Our analysis demonstrates that low models' performance resulted from misinterpreted experimental endpoints or wrongly reported values, thus confirming the observations reported in CERAPP and CoMPARA studies. Developed models and collected data sets included of 6215 (ER) and 3789 (AR) unique compounds, which are freely available.
Collapse
Affiliation(s)
- F Lunghini
- Laboratory of Chemoinformatics, University of Strasbourg , Strasbourg, France
- Toxicological and Environmental Risk Assessment Unit, Solvay S.A ., St. Fons, France
| | - G Marcou
- Laboratory of Chemoinformatics, University of Strasbourg , Strasbourg, France
| | - P Azam
- Toxicological and Environmental Risk Assessment Unit, Solvay S.A ., St. Fons, France
| | - F Bonachera
- Laboratory of Chemoinformatics, University of Strasbourg , Strasbourg, France
| | - M H Enrici
- Toxicological and Environmental Risk Assessment Unit, Solvay S.A ., St. Fons, France
| | - E Van Miert
- Toxicological and Environmental Risk Assessment Unit, Solvay S.A ., St. Fons, France
| | - A Varnek
- Laboratory of Chemoinformatics, University of Strasbourg , Strasbourg, France
| |
Collapse
|
4
|
Popova EA, Kornev AA, Shmakov SV, Nepochatyi GD, Kotyunina OA, Petrov ML, Boitsov VM, Stepakov AV. In Vitro Activity of Organochalcogen Compounds: II. Cytotoxic Effect of 2-Aminobenzo[b]thiophenes Against K562 and HeLa Tumor Cell Lines. RUSS J GEN CHEM+ 2020. [DOI: 10.1134/s107036322011033x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
5
|
Shagufta, Ahmad I, Mathew S, Rahman S. Recent progress in selective estrogen receptor downregulators (SERDs) for the treatment of breast cancer. RSC Med Chem 2020; 11:438-454. [PMID: 33479648 PMCID: PMC7580774 DOI: 10.1039/c9md00570f] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 02/11/2020] [Indexed: 12/11/2022] Open
Abstract
Selective estrogen receptor downregulators (SERDs) are a novel class of compounds capable of reducing the ERα protein level and blocking ER activity. Therefore, SERDs are considered as a significant therapeutic approach to treat ER+ breast cancer in both early stage and more advanced drug-resistant cases. After the FDA approval of a steroidal drug, fulvestrant, as a SERD for the treatment of breast cancer in patients who have progressed on antihormonal agents, several molecules with diverse chemical structures have been rapidly developed, studied and evaluated for selective estrogen receptor downregulation activity. Here we compile the promising SERDs reported in recent years and discuss the chemical structure and pharmacological profile of the most potent compound of the considered series. Because of the availability of only a limited number of effective drugs for the treatment of breast cancer, the quest for a potent SERD with respectable activity and bioavailability is still ongoing. The goal of this article is to make available to the reader an overview of the current progress in SERDs and provide clues for the future discovery and development of novel pharmacological potent SERDs for the treatment of breast cancer.
Collapse
Affiliation(s)
- Shagufta
- Department of Mathematics and Natural Sciences , School of Arts and Sciences , American University of Ras Al Khaimah , P. O. Box 10021 , Ras Al Khaimah , United Arab Emirates . ;
| | - Irshad Ahmad
- Department of Mathematics and Natural Sciences , School of Arts and Sciences , American University of Ras Al Khaimah , P. O. Box 10021 , Ras Al Khaimah , United Arab Emirates . ;
| | - Shimy Mathew
- Department of Biotechnology , School of Arts and Sciences , American University of Ras Al Khaimah , P. O. Box 10021 , Ras Al Khaimah , United Arab Emirates
| | - Sofia Rahman
- Department of Biotechnology , School of Arts and Sciences , American University of Ras Al Khaimah , P. O. Box 10021 , Ras Al Khaimah , United Arab Emirates
| |
Collapse
|
6
|
Tsuchiya K, Umeno T, Tsuji G, Yokoo H, Tanaka M, Fukuhara K, Demizu Y, Misawa T. Development of Photoswitchable Estrogen Receptor Ligands. Chem Pharm Bull (Tokyo) 2020; 68:398-402. [DOI: 10.1248/cpb.c19-01108] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Keisuke Tsuchiya
- Graduate School of Pharmacy, Showa University
- National Institute of Health Sciences
| | - Tomohiro Umeno
- Graduate School of Biomedical Sciences, Nagasaki University
| | | | - Hidetomo Yokoo
- National Institute of Health Sciences
- Graduate School of Medical Life Science, Yokohama City University
| | | | | | - Yosuke Demizu
- National Institute of Health Sciences
- Graduate School of Medical Life Science, Yokohama City University
| | | |
Collapse
|
7
|
Paul S, Das KK, Manna S, Panda S. Transition‐Metal‐Free Synthesis of Heterobiaryls through 1,2‐Migration of Boronate Complex. Chemistry 2020; 26:1922-1927. [DOI: 10.1002/chem.201904761] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 11/16/2019] [Indexed: 11/09/2022]
Affiliation(s)
- Swagata Paul
- Indian Institute of Technology Kharagpur 721302 India
| | | | - Samir Manna
- Indian Institute of Technology Kharagpur 721302 India
| | - Santanu Panda
- Indian Institute of Technology Kharagpur 721302 India
| |
Collapse
|
8
|
6'-Methoxy Raloxifene-analog enhances mouse bone properties with reduced estrogen receptor binding. Bone Rep 2020; 12:100246. [PMID: 32016137 PMCID: PMC6992940 DOI: 10.1016/j.bonr.2020.100246] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 01/15/2020] [Indexed: 02/08/2023] Open
Abstract
Raloxifene (RAL) is an FDA-approved drug used to treat osteoporosis in postmenopausal women. RAL suppresses bone loss primarily through its role as a selective estrogen receptor modulator (SERM). This hormonal estrogen therapy promotes unintended side effects, such as hot flashes and increased thrombosis risk, and prevents the drug from being used in some patient populations at-risk for fracture, including children with bone disorders. It has recently been demonstrated that RAL can have significant positive effects on overall bone mechanical properties by binding to collagen and increasing bone tissue hydration in a cell-independent manner. A Raloxifene-Analog (RAL-A) was synthesized by replacing the 6-hydroxyl substituent with 6-methoxy in effort to reduce the compound's binding affinity for estrogen receptors (ER) while maintaining its collagen-binding ability. It was hypothesized that RAL-A would improve the mechanical integrity of bone in a manner similar to RAL, but with reduced estrogen receptor binding. Molecular assessment showed that while RAL-A did reduce ER binding, downstream ER signaling was not completely abolished. In-vitro, RAL-A performed similarly to RAL and had an identical concentration threshold on osteocyte cell proliferation, differentiation, and function. To assess treatment effect in-vivo, wildtype (WT) and heterozygous (OIM+/−) female mice from the Osteogenesis Imperfecta (OI) murine model were treated with either RAL or RAL-A from 8 weeks to 16 weeks of age. There was an untreated control group for each genotype as well. Bone microarchitecture was assessed using microCT, and mechanical behavior was assessed using 3-point bending. Results indicate that both compounds produced analogous gains in tibial trabecular and cortical microarchitecture. While WT mechanical properties were not drastically altered with either treatment, OIM+/− mechanical properties were significantly enhanced, most notably, in post-yield properties including bone toughness. This proof-of-concept study shows promising results and warrants the exploration of additional analog iterations to further reduce ER binding and improve fracture resistance.
Collapse
|
9
|
Patel HK, Bihani T. Selective estrogen receptor modulators (SERMs) and selective estrogen receptor degraders (SERDs) in cancer treatment. Pharmacol Ther 2018; 186:1-24. [DOI: 10.1016/j.pharmthera.2017.12.012] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
10
|
Stefan SM, Wiese M. Small-molecule inhibitors of multidrug resistance-associated protein 1 and related processes: A historic approach and recent advances. Med Res Rev 2018; 39:176-264. [DOI: 10.1002/med.21510] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/05/2018] [Accepted: 04/28/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Sven Marcel Stefan
- Pharmaceutical Institute; Rheinische Friedrich-Wilhelms-University; Bonn Germany
| | - Michael Wiese
- Pharmaceutical Institute; Rheinische Friedrich-Wilhelms-University; Bonn Germany
| |
Collapse
|
11
|
Wen SM, Lin CH, Chen CC, Wu MJ. Efficient synthesis of 3-benzoyl Benzo[b]thiophenes and raloxifene via Mercury(II)-Catalyzed cyclization of 2-alkynylphenyl alkyl sulfoxides. Tetrahedron 2018. [DOI: 10.1016/j.tet.2018.03.067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
12
|
Popova EA, Lyapunova AG, Petrov ML, Panikorovskii TL, Androsov DA. A Convenient Approach to 2-Aminobenzo[b]chalcogenophenes Based on Copper-Catalyzed Transformation of 4-(2-Bromophenyl)-1,2,3-chalcogenodiazoles in the Presence of a Base and Amines. RUSS J GEN CHEM+ 2018. [DOI: 10.1134/s1070363218040126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
13
|
Tria GS, Abrams T, Baird J, Burks HE, Firestone B, Gaither LA, Hamann LG, He G, Kirby CA, Kim S, Lombardo F, Macchi KJ, McDonnell DP, Mishina Y, Norris JD, Nunez J, Springer C, Sun Y, Thomsen NM, Wang C, Wang J, Yu B, Tiong-Yip CL, Peukert S. Discovery of LSZ102, a Potent, Orally Bioavailable Selective Estrogen Receptor Degrader (SERD) for the Treatment of Estrogen Receptor Positive Breast Cancer. J Med Chem 2018; 61:2837-2864. [PMID: 29562737 DOI: 10.1021/acs.jmedchem.7b01682] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In breast cancer, estrogen receptor alpha (ERα) positive cancer accounts for approximately 74% of all diagnoses, and in these settings, it is a primary driver of cell proliferation. Treatment of ERα positive breast cancer has long relied on endocrine therapies such as selective estrogen receptor modulators, aromatase inhibitors, and selective estrogen receptor degraders (SERDs). The steroid-based anti-estrogen fulvestrant (5), the only approved SERD, is effective in patients who have not previously been treated with endocrine therapy as well as in patients who have progressed after receiving other endocrine therapies. Its efficacy, however, may be limited due to its poor physicochemical properties. We describe the design and synthesis of a series of potent benzothiophene-containing compounds that exhibit oral bioavailability and preclinical activity as SERDs. This article culminates in the identification of LSZ102 (10), a compound in clinical development for the treatment of ERα positive breast cancer.
Collapse
Affiliation(s)
- George S Tria
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Tinya Abrams
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Jason Baird
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Heather E Burks
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Brant Firestone
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - L Alex Gaither
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Lawrence G Hamann
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Guo He
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Christina A Kirby
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Sunkyu Kim
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Franco Lombardo
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Kaitlin J Macchi
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology , Duke University School of Medicine , Durham , North Carolina 27710 , United States
| | - Yuji Mishina
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - John D Norris
- Department of Pharmacology and Cancer Biology , Duke University School of Medicine , Durham , North Carolina 27710 , United States
| | - Jill Nunez
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Clayton Springer
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Yingchuan Sun
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Noel M Thomsen
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Chunrong Wang
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Jianling Wang
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Bing Yu
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Choi-Lai Tiong-Yip
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Stefan Peukert
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| |
Collapse
|
14
|
Abstract
The efficient production, folding, and secretion of proteins is critical for cancer cell survival. However, cancer cells thrive under stress conditions that damage proteins, so many cancer cells overexpress molecular chaperones that facilitate protein folding and target misfolded proteins for degradation via the ubiquitin-proteasome or autophagy pathway. Stress response pathway induction is also important for cancer cell survival. Indeed, validated targets for anti-cancer treatments include molecular chaperones, components of the unfolded protein response, the ubiquitin-proteasome system, and autophagy. We will focus on links between breast cancer and these processes, as well as the development of drug resistance, relapse, and treatment.
Collapse
Affiliation(s)
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, A320 Langley Hall, 4249 Fifth Ave, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
15
|
Cook I, Wang T, Wang W, Kopp F, Wu P, Leyh TS. Controlling Sulfuryl-Transfer Biology. Cell Chem Biol 2017; 23:579-586. [PMID: 27203377 DOI: 10.1016/j.chembiol.2016.04.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/06/2016] [Accepted: 04/14/2016] [Indexed: 10/21/2022]
Abstract
In humans, the cytosolic sulfotransferases (SULTs) catalyze regiospecific transfer of the sulfuryl moiety (-SO3) from 3'-phosphoadenosine 5'-phosphosulfate to thousands of metabolites, including numerous signaling small molecules, and thus regulates their activities and half-lives. Imbalances in the in vivo set points of these reactions leads to disease. Here, with the goal of controlling sulfonation in vivo, molecular ligand-recognition principles in the SULT and nuclear receptor families are integrated in creating a strategy that can prevent sulfonation of a compound without significantly altering its receptor affinity, or inhibiting SULTS. The strategy is validated by using it to control the sulfonation and estrogen receptor (ER) activating activity of raloxifene (a US Food and Drug Administration-approved selective estrogen receptor modulator) and its derivatives. Preventing sulfonation is shown to enhance ER-activation efficacy 10(4)-fold in studies using Ishikawa cells. The strategy offers the opportunity to control sulfuryl transfer on a compound-by-compound basis, to enhance the efficacy of sulfonated drugs, and to explore the biology of sulfuryl transfer with unprecedented precision.
Collapse
Affiliation(s)
- Ian Cook
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461-1926, USA
| | - Ting Wang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461-1926, USA
| | - Wei Wang
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461-1926, USA
| | - Felix Kopp
- Department of Chemical Biology Core Facility, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461-1926, USA
| | - Peng Wu
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461-1926, USA
| | - Thomas S Leyh
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461-1926, USA.
| |
Collapse
|
16
|
Saraiah B, Gautam V, Acharya A, Pasha MA, Hiriyakkanavar I. One-Pot Synthesis of 2-(Aryl/Alkyl)amino-3-cyanobenzo[b
]thiophenes and Their Hetero-Fused Analogues by Pd-Catalyzed Intramolecular Oxidative C-H Functionalization/Arylthiolation. European J Org Chem 2017. [DOI: 10.1002/ejoc.201700963] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Bonagiri Saraiah
- Department of Studies in Chemistry; Central College Campus; Bangalore University; Palace Road 560001 Bangalore India
| | - Vibha Gautam
- New Chemistry Unit; Central College Campus; Jawaharlal Nehru Centre for Advanced Scientific Research; Jakkur 560064 Bangalore India
| | - Anand Acharya
- New Chemistry Unit; Central College Campus; Jawaharlal Nehru Centre for Advanced Scientific Research; Jakkur 560064 Bangalore India
| | - Mohamed A. Pasha
- Department of Studies in Chemistry; Central College Campus; Bangalore University; Palace Road 560001 Bangalore India
| | - Ila Hiriyakkanavar
- New Chemistry Unit; Central College Campus; Jawaharlal Nehru Centre for Advanced Scientific Research; Jakkur 560064 Bangalore India
| |
Collapse
|
17
|
Keri RS, Chand K, Budagumpi S, Balappa Somappa S, Patil SA, Nagaraja BM. An overview of benzo[b]thiophene-based medicinal chemistry. Eur J Med Chem 2017; 138:1002-1033. [PMID: 28759875 DOI: 10.1016/j.ejmech.2017.07.038] [Citation(s) in RCA: 191] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 05/15/2017] [Accepted: 07/20/2017] [Indexed: 01/16/2023]
Abstract
Among sulfur containing heterocycles, benzothiophene and its derivatives are at the focus as these candidates have structural similarities with active compounds to develop new potent lead molecules in drug design. Benzo[b]thiophene scaffold is one of the privileged structures in drug discovery as this core exhibits various biological activities allowing them to act as anti-microbial, anti-cancer, anti-inflammatory, anti-oxidant, anti-tubercular, anti-diabetic, anti-convulsant agents and many more. Further, numerous benzothiophene-based compounds as clinical drugs have been extensively used to treat various types of diseases with high therapeutic potency, which has led to their extensive developments. Due to the wide range of biological activities of benzothiophene, their structure activity relationships (SAR) have generated interest among medicinal chemists, and this has culminated in the discovery of several lead molecules against numerous diseases. The present review is endeavoring to highlight the progress in the various pharmacological activities of benzo[b]thiophene derivatives. It is hoped that this review will be helpful for new thoughts in the quest for rational designs of more active and less toxic benzothiophene-based medicinal drugs, as well as more effective diagnostic agents and pathologic probes. Also, SAR studies that highlight the chemical groups responsible for evoking the potential activities of benzothiophene derivatives are studied and compared.
Collapse
Affiliation(s)
- Rangappa S Keri
- Centre for Nano and Material Sciences, Jain University, Jain Global Campus, Kanakapura, Ramanagaram, Bangalore 562112, India.
| | - Karam Chand
- Department of Chemistry, Umeå University, 90187 Umeå, Sweden
| | - Srinivasa Budagumpi
- Centre for Nano and Material Sciences, Jain University, Jain Global Campus, Kanakapura, Ramanagaram, Bangalore 562112, India
| | - Sasidhar Balappa Somappa
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India; Organic Chemistry Section, Chemical Sciences and Technology Division, Council of Scientific and Industrial Research (CSIR)-National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, India
| | - Siddappa A Patil
- Centre for Nano and Material Sciences, Jain University, Jain Global Campus, Kanakapura, Ramanagaram, Bangalore 562112, India
| | - Bhari Mallanna Nagaraja
- Centre for Nano and Material Sciences, Jain University, Jain Global Campus, Kanakapura, Ramanagaram, Bangalore 562112, India
| |
Collapse
|
18
|
Traboulsi T, El Ezzy M, Gleason JL, Mader S. Antiestrogens: structure-activity relationships and use in breast cancer treatment. J Mol Endocrinol 2017; 58:R15-R31. [PMID: 27729460 PMCID: PMC5148801 DOI: 10.1530/jme-16-0024] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 10/11/2016] [Indexed: 12/11/2022]
Abstract
About 70% of breast tumors express estrogen receptor alpha (ERα), which mediates the proliferative effects of estrogens on breast epithelial cells, and are candidates for treatment with antiestrogens, steroidal or non-steroidal molecules designed to compete with estrogens and antagonize ERs. The variable patterns of activity of antiestrogens (AEs) in estrogen target tissues and the lack of systematic cross-resistance between different types of molecules have provided evidence for different mechanisms of action. AEs are typically classified as selective estrogen receptor modulators (SERMs), which display tissue-specific partial agonist activity (e.g. tamoxifen and raloxifene), or as pure AEs (e.g. fulvestrant), which enhance ERα post-translational modification by ubiquitin-like molecules and accelerate its proteasomal degradation. Characterization of second- and third-generation AEs, however, suggests the induction of diverse ERα structural conformations, resulting in variable degrees of receptor downregulation and different patterns of systemic properties in animal models and in the clinic.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Hormonal/chemistry
- Antineoplastic Agents, Hormonal/pharmacology
- Antineoplastic Agents, Hormonal/therapeutic use
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Clinical Trials as Topic
- Drug Evaluation, Preclinical
- Drug Resistance, Neoplasm
- Estrogen Antagonists/chemistry
- Estrogen Antagonists/pharmacology
- Estrogen Antagonists/therapeutic use
- Estrogen Receptor alpha/antagonists & inhibitors
- Estrogen Receptor alpha/chemistry
- Estrogen Receptor alpha/metabolism
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Models, Molecular
- Molecular Conformation
- Molecular Structure
- Mutation
- Protein Binding
- Protein Processing, Post-Translational
- Receptors, Estrogen/antagonists & inhibitors
- Receptors, Estrogen/chemistry
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Selective Estrogen Receptor Modulators/chemistry
- Selective Estrogen Receptor Modulators/pharmacology
- Selective Estrogen Receptor Modulators/therapeutic use
- Structure-Activity Relationship
- Treatment Outcome
Collapse
Affiliation(s)
- T Traboulsi
- Institute for Research in Immunology and CancerUniversité de Montréal, Montréal, Québec, Canada
- Department of Biochemistry and Molecular MedicineUniversité de Montréal, Montréal, Québec, Canada
| | - M El Ezzy
- Institute for Research in Immunology and CancerUniversité de Montréal, Montréal, Québec, Canada
| | - J L Gleason
- Department of ChemistryMcGill University, Montréal, Québec, Canada
| | - S Mader
- Institute for Research in Immunology and CancerUniversité de Montréal, Montréal, Québec, Canada
- Department of Biochemistry and Molecular MedicineUniversité de Montréal, Montréal, Québec, Canada
| |
Collapse
|
19
|
Misawa T, Fujisato T, Kanda Y, Ohoka N, Shoda T, Yorioka M, Makishima M, Sekino Y, Naito M, Demizu Y, Kurihara M. Design and synthesis of novel selective estrogen receptor degradation inducers based on the diphenylheptane skeleton. MEDCHEMCOMM 2017; 8:239-246. [PMID: 30108709 PMCID: PMC6072319 DOI: 10.1039/c6md00553e] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 11/17/2016] [Indexed: 12/31/2022]
Abstract
Estrogen receptors (ERs) are a family of nuclear receptors (NRs) that regulate physiological effects such as reproduction and bone homeostasis. It has been reported that approximately 70% of human breast cancers are hormone-dependent and ERα-positive. Recently, novel anti-breast cancer drugs based on different mechanisms of action have received significant attention. In this article, we have designed and synthesized a selective ER degradation inducer based on the diphenylheptane skeleton. Western blotting analysis revealed that PBP-NC10 degraded ERα through the ubiquitin-proteasome system. We also performed computational docking analysis to predict the binding mode of PBP-NC10 to ERα.
Collapse
Affiliation(s)
- Takashi Misawa
- National Institute of Health Sciences Setagaya , Tokyo , 158-8501 , Japan . ;
| | - Takuma Fujisato
- National Institute of Health Sciences Setagaya , Tokyo , 158-8501 , Japan . ;
| | - Yasunari Kanda
- National Institute of Health Sciences Setagaya , Tokyo , 158-8501 , Japan . ;
| | - Nobumichi Ohoka
- National Institute of Health Sciences Setagaya , Tokyo , 158-8501 , Japan . ;
| | - Takuji Shoda
- National Institute of Health Sciences Setagaya , Tokyo , 158-8501 , Japan . ;
| | - Momoko Yorioka
- National Institute of Health Sciences Setagaya , Tokyo , 158-8501 , Japan . ;
| | | | - Yuko Sekino
- National Institute of Health Sciences Setagaya , Tokyo , 158-8501 , Japan . ;
| | - Mikihiko Naito
- National Institute of Health Sciences Setagaya , Tokyo , 158-8501 , Japan . ;
| | - Yosuke Demizu
- National Institute of Health Sciences Setagaya , Tokyo , 158-8501 , Japan . ;
| | - Masaaki Kurihara
- National Institute of Health Sciences Setagaya , Tokyo , 158-8501 , Japan . ;
| |
Collapse
|
20
|
Xie X, Li P, Shi Q, Wang L. Visible-light-induced tandem cyclization of 2-alkynylanilines with disulfides: a convenient method for accessing benzothiophenes under transition-metal-free and photocatalyst-free conditions. Org Biomol Chem 2017; 15:7678-7684. [DOI: 10.1039/c7ob01747b] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
An efficient route to benzothiophenes via visible-light induced cyclization of 2-alkynylanilines with disulfides under transition-metal-free and photocatalyst-free conditions was developed.
Collapse
Affiliation(s)
- Xiaoyu Xie
- Department of Chemistry
- Huaibei Normal University
- Huaibei
- P. R. China
| | - Pinhua Li
- Department of Chemistry
- Huaibei Normal University
- Huaibei
- P. R. China
| | - Qing Shi
- Department of Chemistry
- Huaibei Normal University
- Huaibei
- P. R. China
| | - Lei Wang
- Department of Chemistry
- Huaibei Normal University
- Huaibei
- P. R. China
- State Key Laboratory of Organometallic Chemistry
| |
Collapse
|
21
|
Ye LM, Qian L, Chen YY, Zhang XJ, Yan M. A practical synthesis of benzothiophenes via visible-light-promoted cyclization of disulfides and alkynes. Org Biomol Chem 2017; 15:550-554. [DOI: 10.1039/c6ob02461k] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Visible-light-promoted radical cyclization of disulfides and alkynes provided benzothiophenes in good yields.
Collapse
Affiliation(s)
- Lin-Miao Ye
- Institute of Drug Synthesis and Pharmaceutical Process
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou 510006
- China
| | - Lu Qian
- Institute of Drug Synthesis and Pharmaceutical Process
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou 510006
- China
| | - Yan-Yan Chen
- College of Pharmacy
- Guilin Medical University
- Guilin 541004
- China
| | - Xue-Jing Zhang
- Institute of Drug Synthesis and Pharmaceutical Process
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou 510006
- China
| | - Ming Yan
- Institute of Drug Synthesis and Pharmaceutical Process
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou 510006
- China
| |
Collapse
|
22
|
Kathiravan PR, Venugopal M, Muthukumaran S. Synthesis and biological evaluation of novel active arylidene derivatives of 5,6-dihydro-4H-cyclopenta[b]- and 4,5,6,7-tetrahydrobenzo[b]-thiophene-2-carboxylic acid. RESEARCH ON CHEMICAL INTERMEDIATES 2016. [DOI: 10.1007/s11164-016-2763-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
23
|
Vogel VG. Raloxifene: A Selective Estrogen Receptor Modulator for Reducing the Risk of Invasive Breast Cancer in Postmenopausal Women. WOMENS HEALTH 2016; 3:139-53. [DOI: 10.2217/17455057.3.2.139] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Raloxifene hydrochloride is a selective estrogen receptor modulator that has antiestrogenic effects on breast and endometrial tissue and estrogenic effects on bone, lipid metabolism and blood clotting. Tamoxifen is the prototypical selective estrogen receptor modulator and reduces the risk of both in situ and invasive breast cancers by half when compared with placebo. The limitations on the use of tamoxifen for breast cancer risk reduction relate to its well-known, but rare, side effects. A number of clinical trials have established the benefit of raloxifene on osteoporosis and fracture. Raloxifene significantly improves serum lipids and serum markers of cardiovascular disease risk, but has no significant effect on the risk of primary coronary events. In several osteoporosis trials and the Raloxifene Use for The Heart (RUTH) trial, raloxifene decreased the risk of estrogen receptor-positive breast cancer by 44–90%. In the Study of Tamoxifen And Raloxifene (STAR) trial, the effect of raloxifene on invasive breast cancer was equivalent to that of tamoxifen, with more favorable effects on uterine malignancy and clotting events. Symptomatic side effects are acceptable. In total, the available data indicate that raloxifene represents an acceptable alternative to tamoxifen for the reduction of the risk of postmenopausal breast cancer in high-risk women. The potential market for a compound shown to reduce the risk of breast cancer in postmenopausal women who are at increased risk for breast cancer is more than 10 million women in the USA alone.
Collapse
Affiliation(s)
- Victor G Vogel
- UPMC Cancer Center Magee-Women's Hospital, 300 Halket Street, Room 3524, Pittsburgh, PA 15213, USA, Tel.: +1 412 641 6500; Fax: +1 412 641 6461
| |
Collapse
|
24
|
Synthesis and evaluation of raloxifene derivatives as a selective estrogen receptor down-regulator. Bioorg Med Chem 2016; 24:2914-2919. [PMID: 27185013 DOI: 10.1016/j.bmc.2016.04.068] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 04/28/2016] [Accepted: 04/28/2016] [Indexed: 11/22/2022]
Abstract
Estrogen receptors (ERs) play a major role in the growth of human breast cancer cells. A selective estrogen receptor down-regulator (SERD) that acts as not only an inhibitor of ligand binding, but also induces the down-regulation of ER, would be useful for the treatment for ER-positive breast cancer. We previously reported that tamoxifen derivatives, which have a long alkyl chain, had the ability to down-regulate ERα. With the aim of expanding range of the currently available SERDs, we designed and synthesized raloxifene derivatives, which had various lengths of the long alkyl chains, and evaluated their SERD activities. All compounds were able to bind ERα, and RC10, which has a decyl group on the amine moiety of raloxifene, was shown to be the most potent compound. Our findings suggest that the ligand core was replaceable, and that the alkyl length was important for controlling SERD activity. Moreover, RC10 showed antagonistic activity and its potency was superior to that of 4,4'-(heptane-4,4-diyl)bis(2-methylphenol) (18), a competitive antagonist of ER without SERD activity. These results provide information that will be useful for the development of promising SERDs candidates.
Collapse
|
25
|
Design and synthesis of novel flexible ester-containing analogs of tamoxifen and their evaluation as anticancer agents. Future Med Chem 2016; 8:249-56. [DOI: 10.4155/fmc.15.181] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: Tamoxifen (TAM) is metabolized to the more active 4-hydroxytamoxifen by CYP2D6 enzyme. Due to the genetic polymorphisms in CYP2D6, clinical outcomes of TAM treatment vary. Novel flexible TAM analogs with altered activation pathway were synthesized and were tested for their antiproliferative action on MCF-7 cell lines and their binding affinity for ERα and ERβ. Results: All compounds showed better antiproliferative activity than TAM. Compound 3 showed 80-times more ERα binding than TAM, 900-times more selectivity toward ERα. Compound 3 was tested on the entire National Cancer Institute cancerous cell lines; results indicated a broad spectrum anticancer activity. Conclusion: The novel analogs were more potent than TAM with higher selectivity toward ERα and with potential metabolic stability toward CYP2D6.
Collapse
|
26
|
Ahmed NS, Elghazawy NH, ElHady AK, Engel M, Hartmann RW, Abadi AH. Design and synthesis of novel tamoxifen analogues that avoid CYP2D6 metabolism. Eur J Med Chem 2016; 112:171-179. [PMID: 26896706 DOI: 10.1016/j.ejmech.2016.02.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 02/07/2016] [Accepted: 02/08/2016] [Indexed: 11/28/2022]
Abstract
Tamoxifen (TAM) is a widely used drug in the prophylaxis and treatment of breast cancer. TAM is metabolized to the more active 4-hydroxytamoxifen (4-OH-TAM) and endoxifen by cytochrome P450 (CYP) mainly CYP2D6 and CYP3A4 enzymes. Due to the genetic polymorphisms in CYP2D6 genes, high variation in the clinical outcomes of TAM treatment is observed among women of different populations. To address this issue, novel TAM analogues with possible altered activation pathways were synthesized. These analogues were tested for their antiproliferative action on MCF-7 breast cancer cell lines as well as their binding affinity for estrogen receptor (ER) ER-α and ER-β receptors. These entire novel compounds showed better antiproliferative activity than did TAM on the MCF-7 cells. Moreover, compound 10 exhibited a half maximal growth inhibition (GI50) that was 1000 times more potent than that of TAM (GI50 < 0.005 μM vs 1.58 μM, respectively). Along with a broad spectrum activity on various cancer cell lines, all the TAM analogues showed considerable activity on the ER-negative breast cancer cell line. For further study, compound 10 was incubated in human liver microsomes (HLM), human hepatocytes (hHEP) and CYP2D6 supersomes. The active hydroxyl metabolite was detected after incubation in HLM and hHEP, implicating the involvement of other enzymes in its metabolism. These results prove that this novel series of TAM analogues might provide improved clinical outcomes for poor 2D6 metabolizers.
Collapse
Affiliation(s)
- Nermin S Ahmed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835 Cairo, Egypt
| | - Nehal H Elghazawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835 Cairo, Egypt
| | - Ahmed K ElHady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835 Cairo, Egypt
| | - Matthias Engel
- Helmholtz Institute for Pharmaceutical Research Saarland and Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, 66123 Saarbrücken, Germany
| | - Rolf W Hartmann
- Helmholtz Institute for Pharmaceutical Research Saarland and Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, 66123 Saarbrücken, Germany
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835 Cairo, Egypt.
| |
Collapse
|
27
|
Xiong R, Patel HK, Gutgesell LM, Zhao J, Delgado-Rivera L, Pham TND, Zhao H, Carlson K, Martin T, Katzenellenbogen JA, Moore TW, Tonetti DA, Thatcher GRJ. Selective Human Estrogen Receptor Partial Agonists (ShERPAs) for Tamoxifen-Resistant Breast Cancer. J Med Chem 2015; 59:219-237. [PMID: 26681208 DOI: 10.1021/acs.jmedchem.5b01276] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Almost 70% of breast cancers are estrogen receptor α (ERα) positive. Tamoxifen, a selective estrogen receptor modulator (SERM), represents the standard of care for many patients; however, 30-50% develop resistance, underlining the need for alternative therapeutics. Paradoxically, agonists at ERα such as estradiol (E2) have demonstrated clinical efficacy in patients with heavily treated breast cancer, although side effects in gynecological tissues are unacceptable. A drug that selectively mimics the actions of E2 in breast cancer therapy but minimizes estrogenic effects in other tissues is a novel, therapeutic alternative. We hypothesized that a selective human estrogen receptor partial agonist (ShERPA) at ERα would provide such an agent. Novel benzothiophene derivatives with nanomolar potency in breast cancer cell cultures were designed. Several showed partial agonist activity, with potency of 0.8-76 nM, mimicking E2 in inhibiting growth of tamoxifen-resistant breast cancer cell lines. Three ShERPAs were tested and validated in xenograft models of endocrine-independent and tamoxifen-resistant breast cancer, and in contrast to E2, ShERPAs did not cause significant uterine growth.
Collapse
Affiliation(s)
- Rui Xiong
- Department of Medicinal Chemistry & Pharmacognosy, University of Illinois at Chicago, 833 S Wood St, Chicago, Illinois 60612
| | - Hitisha K Patel
- Department of Medicinal Chemistry & Pharmacognosy, University of Illinois at Chicago, 833 S Wood St, Chicago, Illinois 60612
| | - Lauren M Gutgesell
- Department of Medicinal Chemistry & Pharmacognosy, University of Illinois at Chicago, 833 S Wood St, Chicago, Illinois 60612
| | - Jiong Zhao
- Department of Medicinal Chemistry & Pharmacognosy, University of Illinois at Chicago, 833 S Wood St, Chicago, Illinois 60612
| | - Loruhama Delgado-Rivera
- Department of Medicinal Chemistry & Pharmacognosy, University of Illinois at Chicago, 833 S Wood St, Chicago, Illinois 60612
| | - Thao N D Pham
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, 833 S Wood St, Chicago, Illinois 60612
| | - Huiping Zhao
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, 833 S Wood St, Chicago, Illinois 60612
| | - Kathryn Carlson
- Department of Chemistry, University of Illinois, Urbana Champaign, 600 South Mathews Avenue, Urbana, IL 61801
| | - Teresa Martin
- Department of Chemistry, University of Illinois, Urbana Champaign, 600 South Mathews Avenue, Urbana, IL 61801
| | - John A Katzenellenbogen
- Department of Chemistry, University of Illinois, Urbana Champaign, 600 South Mathews Avenue, Urbana, IL 61801
| | - Terry W Moore
- Department of Medicinal Chemistry & Pharmacognosy, University of Illinois at Chicago, 833 S Wood St, Chicago, Illinois 60612
| | - Debra A Tonetti
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, 833 S Wood St, Chicago, Illinois 60612
| | - Gregory R J Thatcher
- Department of Medicinal Chemistry & Pharmacognosy, University of Illinois at Chicago, 833 S Wood St, Chicago, Illinois 60612
| |
Collapse
|
28
|
Bivi N, Hu H, Chavali B, Chalmers MJ, Reutter CT, Durst GL, Riley A, Sato M, Allen MR, Burr DB, Dodge JA. Structural features underlying raloxifene's biophysical interaction with bone matrix. Bioorg Med Chem 2015; 24:759-67. [PMID: 26795112 DOI: 10.1016/j.bmc.2015.12.045] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 12/18/2015] [Accepted: 12/28/2015] [Indexed: 12/29/2022]
Abstract
Raloxifene, a selective estrogen receptor modulator (SERM), reduces fracture risk at least in part by improving the mechanical properties of bone in a cell- and estrogen receptor-independent manner. In this study, we determined that raloxifene directly interacts with the bone tissue. Through the use of multiple and complementary biophysical techniques including nuclear magnetic resonance (NMR) and Fourier transform infrared spectroscopy (FTIR), we show that raloxifene interacts specifically with the organic component or the organic/mineral composite, and not with hydroxyapatite. Structure-activity studies reveal that the basic side chain of raloxifene is an instrumental determinant in the interaction with bone. Thus, truncation of portions of the side chain reduces bone binding and also diminishes the increase in mechanical properties. Our results support a model wherein the piperidine interacts with bone matrix through electrostatic interactions with the piperidine nitrogen and through hydrophobic interactions (van der Waals) with the aliphatic groups in the side chain and the benzothiophene core. Furthermore, in silico prediction of the potential binding sites on the surface of collagen revealed the presence of a groove with sufficient space to accommodate raloxifene analogs. The hydroxyl groups on the benzothiophene nucleus, which are necessary for binding of SERMs to the estrogen receptor, are not required for binding to the bone surface, but mediate a more robust binding of the compound to the bone powder. In conclusion, we report herein a novel property of raloxifene analogs that allows them to interact with the bone tissue through potential contacts with the organic matrix and in particular collagen.
Collapse
Affiliation(s)
- Nicoletta Bivi
- Lilly Research Laboratories, Indianapolis, IN, United States
| | - Haitao Hu
- Lilly Research Laboratories, Indianapolis, IN, United States
| | | | | | | | - Gregory L Durst
- Lilly Research Laboratories, Indianapolis, IN, United States
| | - Anna Riley
- Lilly Research Laboratories, Indianapolis, IN, United States
| | - Masahiko Sato
- Indiana University Purdue University Indianapolis, Indianapolis, IN, United States
| | - Matthew R Allen
- Indiana University Purdue University Indianapolis, Indianapolis, IN, United States
| | - David B Burr
- Indiana University Purdue University Indianapolis, Indianapolis, IN, United States
| | - Jeffrey A Dodge
- Lilly Research Laboratories, Indianapolis, IN, United States
| |
Collapse
|
29
|
Lai A, Kahraman M, Govek S, Nagasawa J, Bonnefous C, Julien J, Douglas K, Sensintaffar J, Lu N, Lee KJ, Aparicio A, Kaufman J, Qian J, Shao G, Prudente R, Moon MJ, Joseph JD, Darimont B, Brigham D, Grillot K, Heyman R, Rix PJ, Hager JH, Smith ND. Identification of GDC-0810 (ARN-810), an Orally Bioavailable Selective Estrogen Receptor Degrader (SERD) that Demonstrates Robust Activity in Tamoxifen-Resistant Breast Cancer Xenografts. J Med Chem 2015; 58:4888-904. [PMID: 25879485 DOI: 10.1021/acs.jmedchem.5b00054] [Citation(s) in RCA: 184] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Approximately 80% of breast cancers are estrogen receptor alpha (ER-α) positive, and although women typically initially respond well to antihormonal therapies such as tamoxifen and aromatase inhibitors, resistance often emerges. Although a variety of resistance mechanism may be at play in this state, there is evidence that in many cases the ER still plays a central role, including mutations in the ER leading to constitutively active receptor. Fulvestrant is a steroid-based, selective estrogen receptor degrader (SERD) that both antagonizes and degrades ER-α and is active in patients who have progressed on antihormonal agents. However, fulvestrant suffers from poor pharmaceutical properties and must be administered by intramuscular injections that limit the total amount of drug that can be administered and hence lead to the potential for incomplete receptor blockade. We describe the identification and characterization of a series of small-molecule, orally bioavailable SERDs which are potent antagonists and degraders of ER-α and in which the ER-α degrading properties were prospectively optimized. The lead compound 11l (GDC-0810 or ARN-810) demonstrates robust activity in models of tamoxifen-sensitive and tamoxifen-resistant breast cancer, and is currently in clinical trials in women with locally advanced or metastatic estrogen receptor-positive breast cancer.
Collapse
Affiliation(s)
- Andiliy Lai
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Mehmet Kahraman
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Steven Govek
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Johnny Nagasawa
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Celine Bonnefous
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Jackie Julien
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Karensa Douglas
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - John Sensintaffar
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Nhin Lu
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Kyoung-Jin Lee
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Anna Aparicio
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Josh Kaufman
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Jing Qian
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Gang Shao
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Rene Prudente
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Michael J Moon
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - James D Joseph
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Beatrice Darimont
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Daniel Brigham
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Kate Grillot
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Richard Heyman
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Peter J Rix
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Jeffrey H Hager
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Nicholas D Smith
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| |
Collapse
|
30
|
Chen CT, Hsu JTA, Lin WH, Lu CT, Yen SC, Hsu T, Huang YL, Song JS, Chen CH, Chou LH, Yen KJ, Chen CP, Kuo PC, Huang CL, Liu HE, Chao YS, Yeh TK, Jiaang WT. Identification of a potent 5-phenyl-thiazol-2-ylamine-based inhibitor of FLT3 with activity against drug resistance-conferring point mutations. Eur J Med Chem 2015; 100:151-61. [PMID: 26081023 DOI: 10.1016/j.ejmech.2015.05.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 04/30/2015] [Accepted: 05/05/2015] [Indexed: 12/19/2022]
Abstract
Numerous FLT3 inhibitors have been explored as a viable therapy for the treatment of acute myeloid leukemia (AML). However, clinical data have been underwhelming due to incomplete inhibition of FLT3 or the emergence of resistant mutations treated with these older agents. We previously developed a series of 3-phenyl-1H-5-pyrazolylamine derivatives as highly potent and selective FLT3 inhibitors with good in vivo efficacy using an intravenous (IV) route. However, the poor bioavailability of these pyrazole compounds limits the development of these promising antileukemic compounds for clinical use. Herein, we describe a novel class of 5-phenyl-thiazol-2-ylamine compounds that are multi-targeted FLT3 inhibitors. From this class of compounds, compound 7h was very potent against AML cell lines and exhibited excellent oral efficacy in AML xenograft models. In addition, further studies demonstrated that compound 7h exhibited potent in vitro and in vivo activities against clinically relevant AC220 (3)-resistant kinase domain mutants of FLT3-ITD.
Collapse
Affiliation(s)
- Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - John T-A Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - Wen-Hsing Lin
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - Cheng-Tai Lu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - Shih-Chieh Yen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - Tsu Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - Yu-Ling Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - Jen-Shin Song
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - Chun-Hwa Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - Ling-Hui Chou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - Kuei-Jung Yen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - Ching-Ping Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - Po-Chu Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - Chen-Lung Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - H Eugene Liu
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan, ROC; Division of Hematology and Oncology, Department of Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan, ROC
| | - Yu-Sheng Chao
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli County 350, Taiwan, ROC
| | - Weir-Torn Jiaang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli County 350, Taiwan, ROC.
| |
Collapse
|
31
|
Sandhya NC, Nandeesh KN, Rangappa KS, Ananda S. One-pot synthesis of 2,3-substituted benzo[b]thiophenes via Cu(i) catalysed intramolecular cyclisation from dithioesters. RSC Adv 2015. [DOI: 10.1039/c5ra02114f] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The novel route involves CuI catalyzed C–S bond formation using dithioesters. This approach is valuable for synthesis of rolaxifene analogues.
Collapse
Affiliation(s)
| | | | | | - Sannaiah. Ananda
- Department of Studies in Chemistry
- University of Mysore
- Mysuru-570006
- India
| |
Collapse
|
32
|
Hou C, He Q, Yang C. Direct Synthesis of Diverse 2-Aminobenzo[b]thiophenes via Palladium-Catalyzed Carbon–Sulfur Bond Formation Using Na2S2O3 as the Sulfur Source. Org Lett 2014; 16:5040-3. [DOI: 10.1021/ol502381e] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Chuanwei Hou
- State Key Laboratory of Drug
Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Qian He
- State Key Laboratory of Drug
Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Chunhao Yang
- State Key Laboratory of Drug
Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| |
Collapse
|
33
|
Molloy ME, White BEP, Gherezghiher T, Michalsen BT, Xiong R, Patel H, Zhao H, Maximov PY, Jordan VC, Thatcher GRJ, Tonetti DA. Novel selective estrogen mimics for the treatment of tamoxifen-resistant breast cancer. Mol Cancer Ther 2014; 13:2515-26. [PMID: 25205655 DOI: 10.1158/1535-7163.mct-14-0319] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Endocrine-resistant breast cancer is a major clinical obstacle. The use of 17β-estradiol (E2) has reemerged as a potential treatment option following exhaustive use of tamoxifen or aromatase inhibitors, although side effects have hindered its clinical usage. Protein kinase C alpha (PKCα) expression was shown to be a predictor of disease outcome for patients receiving endocrine therapy and may predict a positive response to an estrogenic treatment. Here, we have investigated the use of novel benzothiophene selective estrogen mimics (SEM) as an alternative to E2 for the treatment of tamoxifen-resistant breast cancer. Following in vitro characterization of SEMs, a panel of clinically relevant PKCα-expressing, tamoxifen-resistant models were used to investigate the antitumor effects of these compounds. SEM treatment resulted in growth inhibition and apoptosis of tamoxifen-resistant cell lines in vitro. In vivo SEM treatment induced tumor regression of tamoxifen-resistant T47D:A18/PKCα and T47D:A18-TAM1 tumor models. T47D:A18/PKCα tumor regression was accompanied by translocation of estrogen receptor (ER) α to extranuclear sites, possibly defining a mechanism through which these SEMs initiate tumor regression. SEM treatment did not stimulate growth of E2-dependent T47D:A18/neo tumors. In addition, unlike E2 or tamoxifen, treatment with SEMs did not stimulate uterine weight gain. These findings suggest the further development of SEMs as a feasible therapeutic strategy for the treatment of endocrine-resistant breast cancer without the side effects associated with E2.
Collapse
Affiliation(s)
- Mary Ellen Molloy
- Department of Biopharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Bethany E Perez White
- Department of Biopharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Teshome Gherezghiher
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Bradley T Michalsen
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Rui Xiong
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Hitisha Patel
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Huiping Zhao
- Department of Biopharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Philipp Y Maximov
- Department of Oncology, Georgetown University, Lombardi Comprehensive Cancer Center, Washington, District of Columbia
| | - V Craig Jordan
- Department of Oncology, Georgetown University, Lombardi Comprehensive Cancer Center, Washington, District of Columbia
| | - Gregory R J Thatcher
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Debra A Tonetti
- Department of Biopharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
34
|
Yang Y, Zhang T, Huang W, Shen Z. Piperidine Nucleophilic Substitution Without Solvent: An Efficient Synthesis of Raloxifene. SYNTHETIC COMMUN 2014. [DOI: 10.1080/00397911.2014.943348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
35
|
Gallant MA, Brown DM, Hammond M, Wallace JM, Du J, Deymier-Black AC, Almer JD, Stock SR, Allen MR, Burr DB. Bone cell-independent benefits of raloxifene on the skeleton: a novel mechanism for improving bone material properties. Bone 2014; 61:191-200. [PMID: 24468719 PMCID: PMC3955028 DOI: 10.1016/j.bone.2014.01.009] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 01/04/2014] [Accepted: 01/14/2014] [Indexed: 12/29/2022]
Abstract
Raloxifene is an FDA approved agent used to treat bone loss and decrease fracture risk. In clinical trials and animal studies, raloxifene reduces fracture risk and improves bone mechanical properties, but the mechanisms of action remain unclear because these benefits occur largely independent of changes to bone mass. Using a novel experimental approach, machined bone beams, both from mature male canine and human male donors, were depleted of living cells and then exposed to raloxifene ex vivo. Our data show that ex vivo exposure of non-viable bone to raloxifene improves intrinsic toughness, both in canine and human cortical bone beams tested by 4-point bending. These effects are cell-independent and appear to be mediated by an increase in matrix bound water, assessed using basic gravimetric weighing and sophisticated ultrashort echo time magnetic resonance imaging. The hydroxyl groups (OH) on raloxifene were shown to be important in both the water and toughness increases. Wide and small angle X-ray scattering patterns during 4-pt bending show that raloxifene alters the transfer of load between the collagen matrix and the mineral crystals, placing lower strains on the mineral, and allowing greater overall deformation prior to failure. Collectively, these findings provide a possible mechanistic explanation for the therapeutic effect of raloxifene and more importantly identify a cell-independent mechanism that can be utilized for novel pharmacological approaches for enhancing bone strength.
Collapse
Affiliation(s)
- Maxime A Gallant
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, 635 Barnhill Dr, MS-5035, Indianapolis, IN 46202, USA
| | - Drew M Brown
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, 635 Barnhill Dr, MS-5035, Indianapolis, IN 46202, USA
| | - Max Hammond
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN 47907, USA
| | - Joseph M Wallace
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, 723 West Michigan Street, SL 220, Indianapolis, IN 46202, USA
| | - Jiang Du
- Department of Radiology, University of California, 200 West Arbor Drive, MC 0834 San Diego, CA 92103, USA
| | - Alix C Deymier-Black
- Department of Materials Science and Engineering, Northwestern University, 2220 Campus Drive, Cook Hall Room 2036, Evanston, IL 60208, USA
| | - Jonathan D Almer
- Advanced Photon Source, Argonne National Laboratory, Building 401, 9700 S. Cass Avenue, Argonne, IL 60439, USA
| | - Stuart R Stock
- Department of Molecular Pharmacology and Biological Chemistry, Feinberg School of Medicine, Northwestern University, Abbott Hall Suite 810, 710 N Lake Shore Drive, Chicago, IL 60611, USA
| | - Matthew R Allen
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, 635 Barnhill Dr, MS-5035, Indianapolis, IN 46202, USA
| | - David B Burr
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, 635 Barnhill Dr, MS-5035, Indianapolis, IN 46202, USA; Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, 723 West Michigan Street, SL 220, Indianapolis, IN 46202, USA.
| |
Collapse
|
36
|
Bonano VI, Yokoyama-Yasunaka JKU, Miguel DC, Jones SA, Dodge JA, Uliana SRB. Discovery of SyntheticLeishmaniaInhibitors by Screening of a 2-Arylbenzothiophene Library. Chem Biol Drug Des 2013; 83:289-96. [DOI: 10.1111/cbdd.12239] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 09/18/2013] [Accepted: 09/27/2013] [Indexed: 02/03/2023]
Affiliation(s)
- Vivian I. Bonano
- Departamento de Parasitologia; Instituto de Ciências Biomédicas; Universidade de São Paulo; São Paulo SP 05508-900 Brazil
| | | | - Danilo C. Miguel
- Departamento de Parasitologia; Instituto de Ciências Biomédicas; Universidade de São Paulo; São Paulo SP 05508-900 Brazil
| | - Scott A. Jones
- Lilly Research Laboratories; Eli Lilly and Company; Lilly Corporate Center; Indianapolis IN 46285 USA
| | - Jeffrey A. Dodge
- Lilly Research Laboratories; Eli Lilly and Company; Lilly Corporate Center; Indianapolis IN 46285 USA
| | - Silvia R. B. Uliana
- Departamento de Parasitologia; Instituto de Ciências Biomédicas; Universidade de São Paulo; São Paulo SP 05508-900 Brazil
| |
Collapse
|
37
|
Brogi S, Papazafiri P, Roussis V, Tafi A. 3D-QSAR using pharmacophore-based alignment and virtual screening for discovery of novel MCF-7 cell line inhibitors. Eur J Med Chem 2013; 67:344-51. [DOI: 10.1016/j.ejmech.2013.06.048] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 05/10/2013] [Accepted: 06/19/2013] [Indexed: 02/06/2023]
|
38
|
|
39
|
Computational study of estrogen receptor-alpha antagonist with three-dimensional quantitative structure-activity relationship, support vector regression, and linear regression methods. INTERNATIONAL JOURNAL OF MEDICINAL CHEMISTRY 2013; 2013:743139. [PMID: 25505989 PMCID: PMC4245501 DOI: 10.1155/2013/743139] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 03/29/2013] [Indexed: 02/07/2023]
Abstract
Human estrogen receptor (ER) isoforms, ERα and ERβ, have long been an important focus in the field of biology. To better understand the structural features associated with the binding of ERα ligands to ERα and modulate their function, several QSAR models, including CoMFA, CoMSIA, SVR, and LR methods, have been employed to predict the inhibitory activity of 68 raloxifene derivatives. In the SVR and LR modeling, 11 descriptors were selected through feature ranking and sequential feature addition/deletion to generate equations to predict the inhibitory activity toward ERα. Among four descriptors that constantly appear in various generated equations, two agree with CoMFA and CoMSIA steric fields and another two can be correlated to a calculated electrostatic potential of ERα.
Collapse
|
40
|
Maximov PY, Lee TM, Jordan VC. The discovery and development of selective estrogen receptor modulators (SERMs) for clinical practice. CURRENT CLINICAL PHARMACOLOGY 2013; 8:135-55. [PMID: 23062036 PMCID: PMC3624793 DOI: 10.2174/1574884711308020006] [Citation(s) in RCA: 258] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 07/11/2012] [Accepted: 10/03/2012] [Indexed: 01/13/2023]
Abstract
Selective estrogen receptor modulators (SERMs) are structurally different compounds that interact with intracellular estrogen receptors in target organs as estrogen receptor agonists or antagonists. These drugs have been intensively studied over the past decade and have proven to be a highly versatile group for the treatment of different conditions associated with postmenopausal women's health, including hormone responsive cancer and osteoporosis. Tamoxifen, a failed contraceptive is currently used to treat all stages of breast cancer, chemoprevention in women at high risk for breast cancer and also has beneficial effects on bone mineral density and serum lipids in postmenopausal women. Raloxifene, a failed breast cancer drug, is the only SERM approved internationally for the prevention and treatment of postmenopausal osteoporosis and vertebral fractures. However, although these SERMs have many benefits, they also have some potentially serious adverse effects, such as thromboembolic disorders and, in the case of tamoxifen, uterine cancer. These adverse effects represent a major concern given that long-term therapy is required to prevent osteoporosis or prevent and treat breast cancer. The search for the 'ideal' SERM, which would have estrogenic effects on bone and serum lipids, neutral effects on the uterus, and antiestrogenic effects on breast tissue, but none of the adverse effects associated with current therapies, is currently under way. Ospemifene, lasofoxifene, bazedoxifene and arzoxifene, which are new SERM molecules with potentially greater efficacy and potency than previous SERMs, have been investigated for use in the treatment and prevention of osteoporosis. These drugs have been shown to be comparably effective to conventional hormone replacement therapy in animal models, with potential indications for an improved safety profile. Clinical efficacy data from ongoing phase III trials are available or are awaited for each SERM so that a true understanding of the therapeutic potential of these compounds can be obtained. In this article, we describe the discovery and development of the group of medicines called SERMs. The newer SERMs in late development: ospemifene, lasofoxifene, bazedoxifene, are arzoxifene are described in detail.
Collapse
Affiliation(s)
- Philipp Y Maximov
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3970 Reservoir Rd NW, Research Building, Suite E204A, Washington, DC 20057, USA
| | - Theresa M Lee
- Division of Hematology and Oncology, Georgetown University Medical Center, 3970 Reservoir Rd NW, Washington, DC 20057, USA
| | - V. Craig Jordan
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3970 Reservoir Rd NW, Research Building, Suite E204A, Washington, DC 20057, USA
| |
Collapse
|
41
|
Lin WH, Hsu JTA, Hsieh SY, Chen CT, Song JS, Yen SC, Hsu T, Lu CT, Chen CH, Chou LH, Yang YN, Chiu CH, Chen CP, Tseng YJ, Yen KJ, Yeh CF, Chao YS, Yeh TK, Jiaang WT. Discovery of 3-phenyl-1H-5-pyrazolylamine derivatives containing a urea pharmacophore as potent and efficacious inhibitors of FMS-like tyrosine kinase-3 (FLT3). Bioorg Med Chem 2013; 21:2856-67. [PMID: 23618709 DOI: 10.1016/j.bmc.2013.03.083] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 03/28/2013] [Accepted: 03/30/2013] [Indexed: 01/19/2023]
Abstract
Preclinical investigations and early clinical trials suggest that FLT3 inhibitors are a viable therapy for acute myeloid leukemia. However, early clinical data have been underwhelming due to incomplete inhibition of FLT3. We have developed 3-phenyl-1H-5-pyrazolylamine as an efficient template for kinase inhibitors. Structure-activity relationships led to the discovery of sulfonamide, carbamate and urea series of FLT3 inhibitors. Previous studies showed that the sulfonamide 4 and carbamate 5 series were potent and selective FLT3 inhibitors with good in vivo efficacy. Herein, we describe the urea series, which we found to be potent inhibitors of FLT3 and VEGFR2. Some inhibited growth of FLT3-mutated MOLM-13 cells more strongly than the FLT3 inhibitors sorafenib (2) and ABT-869 (3). In preliminary in vivo toxicity studies of the four most active compounds, 10f was found to be the least toxic. A further in vivo efficacy study demonstrated that 10f achieved complete tumor regression in a higher proportion of MOLM-13 xenograft mice than 4 and 5 (70% vs 10% and 40%). These results show that compound 10f possesses improved pharmacologic and selectivity profiles and could be more effective than previously disclosed FLT3 inhibitors in the treatment of acute myeloid leukemia.
Collapse
Affiliation(s)
- Wen-Hsing Lin
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli Country 350, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Petrov ML, Androsov DA. 4-(2-R-Aryl)-1,2,3-chalcogenadiazoles in the synthesis of fused heterocycles. RUSSIAN JOURNAL OF ORGANIC CHEMISTRY 2013. [DOI: 10.1134/s1070428013040015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
43
|
Li WM, Li XB, Sun SX, Liang J, Wang RL, Wang SQ. Agonist and antagonist recognition studies for oestrogen receptor by molecular dynamics simulation. MOLECULAR SIMULATION 2013. [DOI: 10.1080/08927022.2012.717281] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
44
|
Design and synthesis of condensed thienocoumarins by Suzuki–Miyaura reaction/lactonization tandem protocol. Tetrahedron Lett 2012. [DOI: 10.1016/j.tetlet.2012.10.096] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
45
|
Application of 4D-QSAR studies to a series of raloxifene analogs and design of potential selective estrogen receptor modulators. Molecules 2012; 17:7415-39. [PMID: 22706372 PMCID: PMC6268799 DOI: 10.3390/molecules17067415] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 06/04/2012] [Accepted: 06/05/2012] [Indexed: 12/12/2022] Open
Abstract
Four-dimensional quantitative structure-activity relationship (4D-QSAR) analysis was applied on a series of 54 2-arylbenzothiophene derivatives, synthesized by Grese and coworkers, based on raloxifene (an estrogen receptor-alpha antagonist), and evaluated as ERa ligands and as inhibitors of estrogen-stimulated proliferation of MCF-7 breast cancer cells. The conformations of each analogue, sampled from a molecular dynamics simulation, were placed in a grid cell lattice according to three trial alignments, considering two grid cell sizes (1.0 and 2.0 Å). The QSAR equations, generated by a combined scheme of genetic algorithms (GA) and partial least squares (PLS) regression, were evaluated by "leave-one-out" cross-validation, using a training set of 41 compounds. External validation was performed using a test set of 13 compounds. The obtained 4D-QSAR models are in agreement with the proposed mechanism of action for raloxifene. This study allowed a quantitative prediction of compounds' potency and supported the design of new raloxifene analogs.
Collapse
|
46
|
Hsu JTA, Yeh TK, Yen SC, Chen CT, Hsieh SY, Hsu T, Lu CT, Chen CH, Chou LH, Chiu CH, Chang YI, Tseng YJ, Yen KR, Chao YS, Lin WH, Jiaang WT. 3-Phenyl-1H-5-pyrazolylamine-based derivatives as potent and efficacious inhibitors of FMS-like tyrosine kinase-3 (FLT3). Bioorg Med Chem Lett 2012; 22:4654-9. [PMID: 22726931 DOI: 10.1016/j.bmcl.2012.05.116] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 05/18/2012] [Accepted: 05/24/2012] [Indexed: 10/28/2022]
Abstract
A new class of FLT3 inhibitors has been identified based on the 3-phenyl-1H-5-pyrazolylamine scaffold. The structure-activity relationships led to the discovery of two carbamate series, and some potent compounds within these two series exhibited better growth inhibition of FLT3-mutated MOLM-13 cells than FLT3 inhibitors sorafenib (2) and ABT-869 (3). In particular, compound 8d exhibited the ability to regress tumors in mouse xenograft model using MOLM-13 cells.
Collapse
Affiliation(s)
- John T-A Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No. 35, Keyan Rd., Zhunan Town, Miaoli Country 350, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Dadiboyena S. Recent advances in the synthesis of raloxifene: A selective estrogen receptor modulator. Eur J Med Chem 2012; 51:17-34. [DOI: 10.1016/j.ejmech.2012.02.021] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 11/30/2011] [Accepted: 02/11/2012] [Indexed: 10/28/2022]
|
48
|
Chalmers MJ, Wang Y, Novick S, Sato M, Bryant HU, Montrose-Rafizdeh C, Griffin PR, Dodge JA. Hydrophobic Interactions Improve Selectivity to ERα for Ben-zothiophene SERMs. ACS Med Chem Lett 2012; 3:207-210. [PMID: 22582136 DOI: 10.1021/ml2002532] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The discovery, pharmacology, and biophysical characterization of an ERα selective benzothiophene (BTPα) is described. BTPα (4) is a high affinity ligand with 140-fold greater selectivity for ERα (K(i)=0.25 nM) over ERbeta (K(i)=35 nM). In rodent models of estrogen action, BTPα blocks the effects of estrogen in the uterus but mimics the effects estrogen on bone. The basis of ERα selectivity for BTPα was evaluated by using protein crystallography and hydrogen/deuterium exchange (HDX) mass spectrometry. HDX data supports that the n-butyl chain of BTPα stabilizes helix 7 in ERα relative to that of ERβ which we propose leads to an enhancement of affinity to the alpha receptor sub-type.
Collapse
Affiliation(s)
- Michael J. Chalmers
- Department
of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33458,
United States
| | - Yong Wang
- Lilly Research
Laboratories, Eli Lilly and Company, Indianapolis,
Indiana 46285,
United States
| | - Scott Novick
- Department
of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33458,
United States
| | - Masahiko Sato
- Lilly Research
Laboratories, Eli Lilly and Company, Indianapolis,
Indiana 46285,
United States
| | - Henry U. Bryant
- Lilly Research
Laboratories, Eli Lilly and Company, Indianapolis,
Indiana 46285,
United States
| | | | - Patrick R. Griffin
- Department
of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33458,
United States
| | - Jeffrey A. Dodge
- Lilly Research
Laboratories, Eli Lilly and Company, Indianapolis,
Indiana 46285,
United States
| |
Collapse
|
49
|
Lin YT, Chen GY. A scaffold-independent subcellular event-based analysis: characterization of significant structural modifications. J Chem Inf Model 2012; 52:506-14. [PMID: 22288932 DOI: 10.1021/ci200540y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
General and singular subcellular events within the ligand-dependent receptor-mediated cellular response were separated by using the Jurs and the electrotopological state (ES) descriptors, allowing characterization of the significant structural modifications in a given set of collected peroxisome proliferator-activated receptor γ (PPARγ) agonists. The identified Jurs descriptor is the integrated function of all the general events but is scaffold-dependent. The top captured ES descriptors stand for significant structural modifications, i.e., singular events. To further elucidate the descriptor-event relationship, three biological data sets show that the Jurs descriptor can be further divided into three important descriptors, the log D, polar surface area, and shape-like descriptor. The identification of the essential descriptors for general events is the first regression, and the prioritization of all the possible structural modifications of the 46 collected thiazolidinedione PPARγ agonists is the second regression. As results, the top captured ES symbols can correspond to the singular ligand--receptor interactions as highlighted in the X-ray crystallographic image of rosiglitazone--PPARγ complex.
Collapse
Affiliation(s)
- Ying-Ting Lin
- Department of Biotechnology, College of Life Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | | |
Collapse
|
50
|
Therrien E, Englebienne P, Arrowsmith AG, Mendoza-Sanchez R, Corbeil CR, Weill N, Campagna-Slater V, Moitessier N. Integrating medicinal chemistry, organic/combinatorial chemistry, and computational chemistry for the discovery of selective estrogen receptor modulators with Forecaster, a novel platform for drug discovery. J Chem Inf Model 2011; 52:210-24. [PMID: 22133077 DOI: 10.1021/ci2004779] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
As part of a large medicinal chemistry program, we wish to develop novel selective estrogen receptor modulators (SERMs) as potential breast cancer treatments using a combination of experimental and computational approaches. However, one of the remaining difficulties nowadays is to fully integrate computational (i.e., virtual, theoretical) and medicinal (i.e., experimental, intuitive) chemistry to take advantage of the full potential of both. For this purpose, we have developed a Web-based platform, Forecaster, and a number of programs (e.g., Prepare, React, Select) with the aim of combining computational chemistry and medicinal chemistry expertise to facilitate drug discovery and development and more specifically to integrate synthesis into computer-aided drug design. In our quest for potent SERMs, this platform was used to build virtual combinatorial libraries, filter and extract a highly diverse library from the NCI database, and dock them to the estrogen receptor (ER), with all of these steps being fully automated by computational chemists for use by medicinal chemists. As a result, virtual screening of a diverse library seeded with active compounds followed by a search for analogs yielded an enrichment factor of 129, with 98% of the seeded active compounds recovered, while the screening of a designed virtual combinatorial library including known actives yielded an area under the receiver operating characteristic (AU-ROC) of 0.78. The lead optimization proved less successful, further demonstrating the challenge to simulate structure activity relationship studies.
Collapse
Affiliation(s)
- Eric Therrien
- Department of Chemistry, McGill University, 801 Sherbrooke St W, Montreal, QC, Canada H3A 2K6
| | | | | | | | | | | | | | | |
Collapse
|