Discovery of a Potent Dual Inhibitor of Aromatase and Aldosterone SynthaseClick to copy article linkArticle link copied!
- Annachiara TinivellaAnnachiara TinivellaDepartment of Life Sciences, University of Modena and Reggio Emilia, Via G. Campi, Modena 41125, ItalyMore by Annachiara Tinivella
- Marta BanchiMarta BanchiDepartment of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, Pisa 56126, ItalyMore by Marta Banchi
- Guido GambacortaGuido GambacortaDepartment of Chemistry, University of Durham, Lower Mount Joy, South Rd, Durham DH1 3LE, U.K.More by Guido Gambacorta
- Federica BorghiFederica BorghiDepartment of Life Sciences, University of Modena and Reggio Emilia, Via G. Campi, Modena 41125, ItalyMore by Federica Borghi
- Paola OrlandiPaola OrlandiDepartment of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, Pisa 56126, ItalyMore by Paola Orlandi
- Ian R. BaxendaleIan R. BaxendaleDepartment of Chemistry, University of Durham, Lower Mount Joy, South Rd, Durham DH1 3LE, U.K.More by Ian R. Baxendale
- Antonello Di PaoloAntonello Di PaoloDepartment of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, Pisa 56126, ItalyMore by Antonello Di Paolo
- Guido BocciGuido BocciDepartment of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, Pisa 56126, ItalyMore by Guido Bocci
- Luca Pinzi*Luca Pinzi*Email: [email protected]. Tel.: +39 059 2058625.Department of Life Sciences, University of Modena and Reggio Emilia, Via G. Campi, Modena 41125, ItalyMore by Luca Pinzi
- Giulio Rastelli*Giulio Rastelli*Email: [email protected]. Tel.: +39 059 2058564.Department of Life Sciences, University of Modena and Reggio Emilia, Via G. Campi, Modena 41125, ItalyMore by Giulio Rastelli
Abstract
Estrogen deficiency derived from inhibition of estrogen biosynthesis is a typical condition of postmenopausal women and breast cancer (BCs) patients undergoing antihormone therapy. The ensuing increase in aldosterone levels is considered to be the major cause for cardiovascular diseases (CVDs) affecting these patients. Since estrogen biosynthesis is regulated by aromatase (CYP19A1), and aldosterone biosynthesis is modulated by aldosterone synthase (CYP11B2), a dual inhibitor would allow the treatment of BC while reducing the cardiovascular risks typical of these patients. Moreover, this strategy would help overcome some of the disadvantages often observed in single-target or combination therapies. Following an in-depth analysis of a library of synthesized benzylimidazole derivatives, compound X21 was found to be a potent and selective dual inhibitor of aromatase and aldosterone synthase, with IC50 values of 2.3 and 29 nM, respectively. Remarkably, the compound showed high selectivity with respect to 11β-hydroxylase (CYP11B1), as well as CYP3A4 and CYP1A2. When tested in cells, X21 showed potent antiproliferative activity against BC cell lines, particularly against the ER+ MCF-7 cells (IC50 of 0.26 ± 0.03 μM at 72 h), and a remarkable pro-apoptotic effect. In addition, the compound significantly inhibited mTOR phosphorylation at its IC50 concentration, thereby negatively modulating the PI3K/Akt/mTOR axis, which represents an escape for the dependency from ER signaling in BC cells. The compound was further investigated for cytotoxicity on normal cells and potential cardiotoxicity against hERG and Nav1.5 ion channels, demonstrating a safe biological profile. Overall, these assays demonstrated that the compound is potent and safe, thus constituting an excellent candidate for further evaluation.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Results and Discussion
Chemistry
Computational Analyses
compound ID | CYP19A | CYP11B1 | CYP11B2 | |||
---|---|---|---|---|---|---|
N similar compounds based on ECFP4fpb | N similar compounds based on TanimotoComboc | N similar compounds based on ECFP4fpb | N similar compounds based on TanimotoComboc | N similar compounds based on ECFP4fpb | N similar compounds based on TanimotoComboc | |
X1 | 0, 2 | 0, 0, 0 | 0, 2 | 0, 0, 4 | 1, 1 | 0, 0, 4 |
X2 | 0, 3 | 0, 0, 0 | 0, 3 | 0, 0, 15 | 3, 2 | 0, 0, 14 |
X3 | 0, 3 | 0, 0, 5 | 0, 3 | 0, 0, 24 | 3, 2 | 0, 0, 21 |
X4 | 0, 1 | 0, 0, 0 | 0, 1 | 0, 0, 0 | 0, 0 | 0, 0, 0 |
X5 | 0, 0 | 0, 0, 0 | 0, 1 | 0, 0, 4 | 0, 0 | 0, 0, 4 |
X6 | 0, 3 | 0, 0, 0 | 0, 3 | 0, 0, 17 | 3, 2 | 0, 0, 16 |
X7 | 0, 1 | 0, 0, 0 | 0, 1 | 0, 0, 3 | 0, 0 | 0, 0, 2 |
X8 | 0, 3 | 0, 0, 0 | 0, 2 | 0, 0, 7 | 1, 1 | 0, 0, 6 |
X9 | 0, 1 | 0, 0, 0 | 0, 0 | 0, 0, 0 | 0, 0 | 0, 0, 0 |
X10 | 0, 1 | 0, 0, 0 | 0, 0 | 0, 0, 1 | 0, 0 | 0, 0, 1 |
X11 | 0, 1 | 0, 0, 0 | 0, 1 | 0, 0, 0 | 0, 0 | 0, 0, 0 |
X12 | 0, 1 | 0, 0, 0 | 0, 1 | 0, 0, 0 | 0, 0 | 0, 0, 0 |
X13 | 0, 2 | 0, 0, 0 | 0, 1 | 0, 0, 0 | 2, 0 | 0, 0, 0 |
X14 | 0, 2 | 0, 0, 0 | 0, 1 | 0, 0, 0 | 0, 1 | 0, 0, 0 |
X15 | 0, 1 | 0, 0, 0 | 0, 1 | 0, 0, 0 | 0, 0 | 0, 0, 0 |
X16 | 0, 1 | 0, 0, 0 | 0, 0 | 0, 0, 0 | 0, 0 | 0, 0, 0 |
X17 | 0, 1 | 0, 0, 0 | 0, 1 | 0, 0, 0 | 0, 0 | 0, 0, 0 |
X18 | 0, 1 | 0, 0, 0 | 0, 1 | 0, 0, 3 | 0, 0 | 0, 0, 3 |
X19 | 0, 1 | 0, 0, 0 | 0, 1 | 0, 0, 3 | 0, 0 | 0, 0, 3 |
X20 | 0, 1 | 0, 0, 3 | 1, 1 | 0, 0, 20 | 2, 0 | 0, 0, 18 |
X21 | 0, 2 | 0, 0, 7 | 1, 1 | 1, 1, 15 | 3, 1 | 1, 1, 15 |
X22 | 0, 2 | 0, 0, 0 | 1, 2 | 0, 0, 0 | 5, 2 | 0, 0, 0 |
X23 | 0, 2 | 0, 0, 0 | 0, 1 | 0, 0, 3 | 0, 0 | 0, 0, 2 |
X24 | 0, 1 | 0, 0, 2 | 1, 1 | 0, 0, 9 | 2, 0 | 0, 0, 5 |
ECFP4fp-based similarity estimations were performed with the LigAdvisor Web server, (25) while 3D similarity estimations were made with the ROCS software. (53)
For each synthesized compound, the number of “PDB ligands, DrugBank ligands” that showed Tanimoto index above 0.3 is reported.
For each synthesized compound, the number of “PDB ligands, DrugBank ligands, ChEMBL ligands” that showed TanimotoCombo index above 1.5 is reported (only ChEMBL ligands with reported IC50, Ki, Kd, EC50, and potency below 1 μM were taken into consideration).
Biological Evaluation
In Vitro Inhibitory Activity
Conclusions
Experimental Section
Chemistry
General Information
Experimental Preparation for Compounds X4, X11–12, and X14–18
(1-(Benzo[d][1,3]dioxol-5-ylmethyl)-2-((2-nitro-5-(trifluoromethyl)phenyl)thio)-1H-imidazol-5-yl)methanol, Compound X17
(2-((5-Chloro-2-nitrophenyl)thio)-1-(2-(thiophen-2-yl)ethyl)-1H-imidazol-5-yl)methanol, Compound X14
(1-Benzyl-2-((2-nitro-4-(trifluoromethyl)phenyl)thio)-1H-imidazol-5-yl)methanol, Compound X12
(1-Benzyl-2-((2-nitro-5-(trifluoromethyl)phenyl)thio)-1H-imidazol-5-yl)methanol, Compound X11
(1-Benzyl-2-(methylthio)-1H-imidazol-5-yl)methanol, Compound X18
(1-Benzyl-2-((5-chloro-2-nitrophenyl)thio)-1H-imidazol-5-yl)methanol, Compound X4
(1-Benzyl-2-((3,5-dichloro-2,6-difluoropyridin-4-yl)thio)-1H-imidazol-5-yl)methanol, Compound X15
1-Benzyl-2-((3,5-dichloro-2,6-difluoropyridin-4-yl)thio)-1H-imidazole-5-carbaldehyde, Compound X16
4-((5-(Hydroxymethyl)-1H-imidazol-1-yl)methyl)benzonitrile, Compound X21
4-((5-((4-(3-Chlorophenyl)-3-oxopiperazin-1-yl)methyl)-1H-imidazol-1-yl)methyl)benzonitrile, Compound X22
Computational Methods
Computational Investigations Made with the LigAdvisor Web Server
CYP19A1, CYP11B1, and CYP11B2 Data Set Generation
ChEMBL Data Set
Target type equal to “Single Protein”;
Standard type expressed as Ki, Kd, IC50, EC50, potency;
Standard relation equal to “>” or “=”.
DrugBank Data Set
PDB Data Set
3D Ligand-Based Analyses
In Silico Tautomer Stability Assessment for Compound X1
Structure-Based Analyses
Biological Assays
In Vitro Assays on Recombinant CYP19A1, CYP1A2, and CYP3A4 Enzymes
Pharmacological Assays
Materials, Drugs, and Cells Lines
Cell Proliferation and Apoptosis Assay
Luminex Analysis
Statistical Data Analysis
Cardiac Safety Assessment
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsptsci.3c00183.
Results of the similarity estimations performed with the LigAdvisor Web server; results of the similarity estimations performed with respect to compounds with activity annotation on CYP19A1, CYP11B2, and CYP11B1, reported in the DrugBank, PDB, and ChEMBL databases; docking scores of the investigated compounds into the PDB crystal structures 3EQM (CYP19A1), 6M7X (CYP11B1), and 4FDH (CYP11B2); binding mode predicted for (S)- and (R)-fadrozole into the CYP19A1 binding site (PDB ID: 3EQM); titration curves of compound X21 against CYP19A1, CYP1A2, and CYP3A4, with their respective controls; antiproliferative in vitro activity of fadrozole on human MCF-7 (Estrogen Receptor+) at 24, 48, and 72 h; and titration curves of compound X21 and the reference compounds E-4031 and tetrodoxin, against hERG and Nav1.5 (manual patch clamp assays), respectively (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
We thank OpenEye Scientific Software, Inc. for a free academic license. We wish to thank Dr. Arianna Bandini for technical assistance.
ACE | angiotensin-converting enzyme |
AI | aromatase inhibitor |
AKT | protein kinase B |
Ang II | angiotensin II |
AT1R | angiotensin type 1 receptor |
ATCC | American Type Culture Collection |
BC | breast cancer |
bFGF | basic fibroblast growth factor |
C | vehicle-treated control |
CVD | cardiovascular disease |
CYP11B1 | cytochrome P450 family 11 subfamily B member 1 |
CYP11B2 | aldosterone synthase |
CYP19A1 | aromatase |
CYP1A2 | cytochromes P450 1A2 |
CYP3A4 | cytochromes P450 3A4 |
EGF | epidermal growth factor |
ERBB2/HER2+ | human epidermal growth factor receptor-2 positive |
ER | estrogen receptor |
FBS | foetal bovine serum |
GSK | glycogen synthase kinase 3 |
HNDF | human normal dermal human fibroblast cell line |
IGF1R | insulin-like growth factor 1 (IGF-1) receptor |
IR | insulin receptor |
IRS1 | insulin receptor substrate 1 |
mTOR | mammalian target of rapamycin |
MW | molecular weight |
p70S6K | ribosomal protein S6 kinase beta-1 |
PDB | protein data bank |
PR | progesterone receptor |
PTEN | phosphatase and tensin homologue |
RAAS | renin–angiotensin–aldosterone system |
RMSD | root-mean-square deviation |
RP6S | ribosomal protein S6 |
SERD | selective estrogen receptors degrader |
SERM | selective estrogen receptors modulator |
TSC2 | tuberous sclerosis complex 2 |
References
This article references 72 other publications.
- 1Nardin, S.; Mora, E.; Varughese, F. M.; D’Avanzo, F.; Vachanaram, A. R.; Rossi, V.; Saggia, C.; Rubinelli, S.; Gennari, A. Breast Cancer Survivorship, Quality of Life, and Late Toxicities. Front. Oncol. 2020, 10, 864, DOI: 10.3389/fonc.2020.00864Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB38nntlaqsw%253D%253D&md5=bface92499349059a608f9c51d724b5eBreast Cancer Survivorship, Quality of Life, and Late ToxicitiesNardin Simone; Mora Edoardo; Varughese Feba Mariam; Vachanaram Ajay Ram; Gennari Alessandra; Nardin Simone; Mora Edoardo; Varughese Feba Mariam; Vachanaram Ajay Ram; Gennari Alessandra; D'Avanzo Francesca; Rossi Valentina; Saggia Chiara; Rubinelli Sara; Gennari AlessandraFrontiers in oncology (2020), 10 (), 864 ISSN:2234-943X.Breast cancer is the most frequent cancer in women: in 2018, almost two million cases have been diagnosed all over the world and it represents the principal cause of death from a neoplastic disease in women. In the past years, breast cancer prognosis has significantly improved over time: currently 5-year survival rates are in the range of 90%, and 10-year survival is about 80%. This improvement has been mostly observed in western countries, due to high coverage and compliance with screening programs, leading to early diagnosis, i.e., when the disease is at a subclinical level, and to an improvement in tumor molecular characterization and innovative systemic treatments. Yet the identification of different biological breast cancer subtypes prompted the development of innovative targeted agents and improved treatment personalization. On the other hand, longer survival rates and increasing proportions of cured patients require dedicated strategies to manage long-term sequelae of breast cancer treatments, with particular attention to quality of life. This review analyzes the most important issues, potentially occurring with cancer treatments, concerning long-term sequelae and quality of life, to define a global approach to breast cancer survivorship.
- 2International Agency for Research on Cancer, Lyon, France. Global Cancer Observatory. https://gco.iarc.fr/.Google ScholarThere is no corresponding record for this reference.
- 3Smolarz, B.; Nowak, A. Z.; Romanowicz, H. Breast Cancer─Epidemiology, Classification, Pathogenesis and Treatment (Review of Literature). Cancers 2022, 14 (10), 2569, DOI: 10.3390/cancers14102569Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhsV2ht7%252FF&md5=022928457eab2d2e1b9419b2b659bce9Breast Cancer-Epidemiology, Classification, Pathogenesis and Treatment (Review of Literature)Smolarz, Beata; Nowak, Anna Zadrozna; Romanowicz, HannaCancers (2022), 14 (10), 2569CODEN: CANCCT; ISSN:2072-6694. (MDPI AG)Simple Summary: Breast cancer is the most-commonly diagnosed malignant tumor in women in the world, as well as the first cause of death from malignant tumors. The incidence of breast cancer is constantly increasing in all regions of the world. For this reason, despite the progress in its detection and treatment, which translates into improved mortality rates, it seems necessary to look for new therapeutic methods, predictive and prognostic factors. The article presents a review of the literature on breast carcinoma - a disease affecting women in the world. Abstr.: Breast cancer is the most-commonly diagnosed malignant tumor in women in the world, as well as the first cause of death from malignant tumors. The incidence of breast cancer is constantly increasing in all regions of the world. For this reason, despite the progress in its detection and treatment, which translates into improved mortality rates, it seems necessary to look for new therapeutic methods, and predictive and prognostic factors. Treatment strategies vary depending on the mol. subtype. Breast cancer treatment is multidisciplinary; it includes approaches to locoregional therapy (surgery and radiation therapy) and systemic therapy. Systemic therapies include hormone therapy for hormone-pos. disease, chemotherapy, anti-HER2 therapy for HER2-pos. disease, and quite recently, immunotherapy. Triple neg. breast cancer is responsible for more than 15-20% of all breast cancers. It is of particular research interest as it presents a therapeutic challenge, mainly due to its low response to treatment and its highly invasive nature. Future therapeutic concepts for breast cancer aim to individualize therapy and de-escalate and escalate treatment based on cancer biol. and early response to therapy. The article presents a review of the literature on breast carcinoma-a disease affecting women in the world.
- 4Haque, Md. M.; Desai, K. V. Pathways to Endocrine Therapy Resistance in Breast Cancer. Front. Endocrinol. 2019, 10, 573, DOI: 10.3389/fendo.2019.00573Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MrmtlOgtw%253D%253D&md5=f90b9a06e6fb87d34ab346490bb105e5Pathways to Endocrine Therapy Resistance in Breast CancerHaque Md Moquitul; Desai Kartiki VFrontiers in endocrinology (2019), 10 (), 573 ISSN:1664-2392.Breast cancers with positive expression of Estrogen Receptor (ER+) are treated with anti-hormone/endocrine therapy which targets the activity of the receptor, the half-life of the receptor or the availability of estrogen. This has significantly decreased mortality in women with ER+ breast cancer, however, about 25-30% of treated women run the risk or recurrence due to either intrinsic or acquired resistance to endocrine therapies. While ER itself is a predictor of response to such therapies, there exists a need to find more biomarkers and novel targets to treat resistant tumors. In this review, we summarize the known mechanisms and describe the ability of genomics in unraveling rare mutations and gene rearrangements that may impact the development of resistance and therefore treatment of ER+ breast cancer in the near future.
- 5Sørlie, T.; Perou, C. M.; Tibshirani, R.; Aas, T.; Geisler, S.; Johnsen, H.; Hastie, T.; Eisen, M. B.; Van De Rijn, M.; Jeffrey, S. S.; Thorsen, T.; Quist, H.; Matese, J. C.; Brown, P. O.; Botstein, D.; Lønning, P. E.; Børresen-Dale, A.-L. Gene Expression Patterns of Breast Carcinomas Distinguish Tumor Subclasses with Clinical Implications. Proc. Natl. Acad. Sci. U. S. A. 2001, 98 (19), 10869– 10874, DOI: 10.1073/pnas.191367098Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXntVGmsbo%253D&md5=5a54045e69bbc92a5294eb5e73934bd7Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implicationsSorlie, Therese; Perou, Charles M.; Tibshirani, Robert; Aas, Turid; Geisler, Stephanie; Johnsen, Hilde; Hastie, Trevor; Eisen, Michael B.; Van de Rijn, Matt; Jeffrey, Stefanie S.; Thorsen, Thor; Quist, Hanne; Matese, John C.; Brown, Patrick O.; Botstein, David; Lonning, Per Eystein; Borresen-Dale, Anne-LiseProceedings of the National Academy of Sciences of the United States of America (2001), 98 (19), 10869-10874CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The purpose of this study was to classify breast carcinomas based on variations in gene expression patterns derived from cDNA microarrays and to correlate tumor characteristics to clin. outcome. A total of 85 cDNA microarray expts. representing 78 cancers, three fibroadenomas, and four normal breast tissues were analyzed by hierarchical clustering. As reported previously, the cancers could be classified into a basal epithelial-like group, an ERBB2-overexpressing group and a normal breast-like group based on variations in gene expression. A novel finding was that the previously characterized luminal epithelial/estrogen receptor-pos. group could be divided into at least two subgroups, each with a distinctive expression profile. These subtypes proved to be reasonably robust by clustering using two different gene sets: first, a set of 456 cDNA clones previously selected to reflect intrinsic properties of the tumors and, second, a gene set that highly correlated with patient outcome. Survival analyses on a subcohort of patients with locally advanced breast cancer uniformly treated in a prospective study showed significantly different outcomes for the patients belonging to the various groups, including a poor prognosis for the basal-like subtype and a significant difference in outcome for the two estrogen receptor-pos. groups.
- 6Musgrove, E. A.; Sutherland, R. L. Biological Determinants of Endocrine Resistance in Breast Cancer. Nat. Rev. Cancer 2009, 9 (9), 631– 643, DOI: 10.1038/nrc2713Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtVegtrzL&md5=e57a6a082e0f41d26422350657201de5Biological determinants of endocrine resistance in breast cancerMusgrove, Elizabeth A.; Sutherland, Robert L.Nature Reviews Cancer (2009), 9 (9), 631-643CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)A review. Endocrine therapies targeting estrogen action (anti-estrogens, such as tamoxifen, and aromatase inhibitors) decrease mortality from breast cancer, but their efficacy is limited by intrinsic and acquired therapeutic resistance. Candidate mol. biomarkers and gene expression signatures of tamoxifen response emphasize the importance of deregulation of proliferation and survival signaling in endocrine resistance. However, definition of the specific genetic lesions and mol. processes that det. clin. endocrine resistance is incomplete. The development of large-scale computational and genetic approaches offers the promise of identifying the mediators of endocrine resistance that may be exploited as potential therapeutic targets and biomarkers of response in the clinic.
- 7Liu, C.-Y.; Wu, C.-Y.; Petrossian, K.; Huang, T.-T.; Tseng, L.-M.; Chen, S. Treatment for the Endocrine Resistant Breast Cancer: Current Options and Future Perspectives. J. Steroid Biochem. Mol. Biol. 2017, 172, 166– 175, DOI: 10.1016/j.jsbmb.2017.07.001Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFGmtLjK&md5=93d1de1dcc2f66ae08f3f95887c38adaTreatment for the endocrine resistant breast cancer: Current options and future perspectivesLiu, Chun-Yu; Wu, Chia-Yun; Petrossian, Karineh; Huang, Tzu-Ting; Tseng, Ling-Ming; Chen, ShiuanJournal of Steroid Biochemistry and Molecular Biology (2017), 172 (), 166-175CODEN: JSBBEZ; ISSN:0960-0760. (Elsevier Ltd.)Endocrine resistance remains a challenge and an unmet need for managing hormone receptor-pos. breast cancer. The mechanisms of endocrine resistance are multifaceted and are likely to evolve over time following various single or combination therapies. The purpose of this review article is to provide general understanding of mol. basis of endocrine resistance of breast cancer and to offer comprehensive review on current treatment options and potential new treatment strategies for endocrine resistant breast cancers. Last but not the least, we discuss current challenges and future directions for management of endocrine resistant breast cancers.
- 8Hu, Q.; Yin, L.; Hartmann, R. W. Selective Dual Inhibitors of CYP19 and CYP11B2: Targeting Cardiovascular Diseases Hiding in the Shadow of Breast Cancer. J. Med. Chem. 2012, 55 (16), 7080– 7089, DOI: 10.1021/jm3004637Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtFClurjO&md5=67a25d86529e58c06f10a4baf5463542Selective Dual Inhibitors of CYP19 and CYP11B2: Targeting Cardiovascular Diseases Hiding in the Shadow of Breast CancerHu, Qingzhong; Yin, Lina; Hartmann, Rolf W.Journal of Medicinal Chemistry (2012), 55 (16), 7080-7089CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Postmenopausal women are at high risk for cardiovascular diseases because of the estrogen deficiency. As for postmenopausal breast cancer patients, this risk is even higher due to inhibition of estrogens biosyntheses in peripheral tissue by the aromatase (CYP19) inhibitors applied. Because estrogen deficiency results in significantly elevated aldosterone levels, which are a major cause of cardiovascular diseases, dual inhibition of CYP19 and CYP11B2 (aldosterone synthase) is a promising treatment for breast cancer and the coinstantaneous cardiovascular diseases. By combination of important structural features of known CYP19 and CYP11B2 inhibitors, we succeeded in obtaining compds. 3 and 5 as selective dual inhibitors with IC50 values around 50 and 20 nM toward CYP19 and CYP11B2, resp. These compds. showed also good selectivity toward CYP11B1 (selectivity factors (IC50 CYP11B1/IC50 CYP11B2) around 50) and CYP17 (no inhibition).
- 9Perez, E. A. Safety Profiles of Tamoxifen and the Aromatase Inhibitors in Adjuvant Therapy of Hormone-Responsive Early Breast Cancer. Ann. Oncol. 2007, 18, viii26– viii35, DOI: 10.1093/annonc/mdm263Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2sjht1ahsQ%253D%253D&md5=4e51288fe254120a955a47510271d03bSafety profiles of tamoxifen and the aromatase inhibitors in adjuvant therapy of hormone-responsive early breast cancerPerez E AAnnals of oncology : official journal of the European Society for Medical Oncology / ESMO (2007), 18 Suppl 8 (), viii26-35 ISSN:.Adjuvant endocrine therapy plays an important role in the management of hormone-receptor-positive early breast cancer, and has increased life expectancy for millions of women. Many patients receive adjuvant treatment for at least 5 years following tumor resection, hence good long-term safety is important for endocrine agents to gain widespread acceptance. Tamoxifen has been used as adjuvant therapy for early breast cancer for many years, and safety data have been well documented, but a poor risk:benefit profile limits treatment duration to 5 years. Increased efficacy over tamoxifen and good tolerability have recently made the third-generation aromatase inhibitors (AIs) the first-choice agents for adjuvant endocrine therapy; however, it is currently not known whether AI therapy, like tamoxifen, will be limited to 5 years. Many side effects of endocrine therapy, such as hot flushes and mood disturbances, are related to estrogen deprivation and are common to tamoxifen and AIs, reflecting the mechanism of action of these drugs. In addition, tamoxifen has estrogenic effects that are beneficial in some tissues: tamoxifen lowers serum cholesterol levels and protects against bone loss and cardiovascular disease, but is also associated with potentially life-threatening side effects, such as endometrial cancer and thromboembolic disease. As AIs lack estrogenic activity, they are not associated with these serious adverse events. Clinical trials comparing AIs with tamoxifen in the adjuvant setting have shown that AIs are well tolerated and are associated with a lower incidence of gynecological symptoms and hot flushes than tamoxifen. However, AIs are associated with musculoskeletal side effects, such as arthralgia, myalgia and bone loss, but these events are preventable or manageable. The effects of AIs on lipid metabolism and the cardiovascular system are still debatable, but placebo-controlled trials provide no evidence to suggest that AIs adversely affect these systems. Furthermore, the AIs allow women to maintain a good quality of life, comparable with women receiving tamoxifen or placebo, and are a cost-effective therapeutic option. Ongoing trials will provide more information regarding the long-term effects of AI therapy and will provide comparative data on the efficacy and safety of the different AIs, thereby helping to determine the optimal treatment strategy for these highly effective and well-tolerated drugs.
- 10Chapman, J.-A. W.; Meng, D.; Shepherd, L.; Parulekar, W.; Ingle, J. N.; Muss, H. B.; Palmer, M.; Yu, C.; Goss, P. E. Competing Causes of Death From a Randomized Trial of Extended Adjuvant Endocrine Therapy for Breast Cancer. JNCI J. Natl. Cancer Inst. 2008, 100 (4), 252– 260, DOI: 10.1093/jnci/djn014Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1c7isFSltA%253D%253D&md5=afc9fa23e8500381ad33cfabbd5783c8Competing causes of death from a randomized trial of extended adjuvant endocrine therapy for breast cancerChapman Judith-Anne W; Meng Daniel; Shepherd Lois; Parulekar Wendy; Ingle James N; Muss Hyman B; Palmer Michael; Yu Changhong; Goss Paul EJournal of the National Cancer Institute (2008), 100 (4), 252-60 ISSN:.BACKGROUND: Older women with early-stage breast cancer experience higher rates of non-breast cancer-related death. We examined factors associated with cause-specific death in a large cohort of breast cancer patients treated with extended adjuvant endocrine therapy. METHODS: In the MA.17 trial, conducted by the National Cancer Institute of Canada Clinical Trials Group, 5170 breast cancer patients (median age = 62 years; range = 32-94 years) who were disease free after approximately 5 years of adjuvant tamoxifen treatment were randomly assigned to treatment with letrozole (2583 women) or placebo (2587 women). The median follow-up was 3.9 years (range 0-7 years). We investigated the association of 11 baseline factors with the competing risks of death from breast cancer, other malignancies, and other causes. All statistical tests were two-sided likelihood ratio criterion tests. RESULTS: During follow-up, 256 deaths were reported (102 from breast cancer, 50 from other malignancies, 100 from other causes, and four from an unknown cause). Non-breast cancer deaths accounted for 60% of the 252 known deaths (72% for those > or = 70 years and 48% for those < 70 years). Two baseline factors were differentially associated with type of death: cardiovascular disease was associated with a statistically significant increased risk of death from other causes (P.002), and osteoporosis was associated with a statistically significant increased risk of death from other malignancies (P.05). An increased risk of breast cancer-specific death was associated with lymph node involvement (P < .001). Increased risk of death from all three causes was associated with older age (P < .001). CONCLUSIONS: Non-breast cancer-related deaths were more common than breast cancer-specific deaths in this cohort of 5-year breast cancer survivors, especially among older women.
- 11Wang, Y.; Wang, Q.; Zhao, Y.; Gong, D.; Wang, D.; Li, C.; Zhao, H. Protective Effects of Estrogen Against Reperfusion Arrhythmias Following Severe Myocardial Ischemia in Rats. Circ. J. 2010, 74 (4), 634– 643, DOI: 10.1253/circj.CJ-09-0223Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXls1Sksbk%253D&md5=82f084f57eb2caeb3d27f0caad1dbdfdProtective effects of estrogen against reperfusion arrhythmias following severe myocardial ischemia in ratsWang, Yan; Wang, Qi; Zhao, Yingnan; Gong, Dezheng; Wang, Dongmei; Li, Cong; Zhao, HenanCirculation Journal (2010), 74 (4), 634-643CODEN: CJIOBY; ISSN:1346-9843. (Japanese Circulation Society)Background: Female sex hormones may have protective effects against arrhythmias, including reperfusion arrhythmias (RAs), but the mechanisms are still not completely known. Methods and Results: Serial changes in rat hearts (rhythm, apoptosis and the its influencing factors; cardiac vinculin mRNA expression and connexin43 (Cx43) dephosphorylation) were examd. during periods of ischemia-reperfusion with and without estrogen treatment. After reperfusion, although the incidence of arrhythmias became higher in both the vehicle-group and estrogen-group, compared with the ischemia period, estrogen prevented reperfusion-induced upregulation of the incidence of arrhythmias, esp. ventricular premature beats (VPB) and ventricular tachycardia (VT). The duration of VT and fibrillation, and the no. of VPB and VT, were all significantly decreased in the estrogen-group. The expression of cardiac vinculin mRNA decreased significantly in the vehicle-group but not in the estrogen-group. Cx43 dephosphorylation and myocyte apoptosis increased in both groups, but the values for the estrogen-group were all markedly lower than those for the vehicle-group. A selective estrogen receptor (ER) β agonist prevented reperfusion-induced upregulation of the incidence of both VPB and VT significantly; a selective ERα agonist had no significant influence. Conclusions: Estrogen can protect the heart against RAs, at least in part, mediated through gap junctions. Upregulation of ERβ but not ERα mediated most of the estrogen-induced cardioprotection against RA.
- 12Beer, S.; Reincke, M.; Kral, M.; Callies, F.; Strömer, H.; Dienesch, C.; Steinhauer, S.; Ertl, G.; Allolio, B.; Neubauer, S. High-Dose 17β–Estradiol Treatment Prevents Development of Heart Failure Post–Myocardial Infarction in the Rat. Basic Res. Cardiol. 2007, 102 (1), 9– 18, DOI: 10.1007/s00395-006-0608-1Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhvFymu78%253D&md5=2cb6046ecf83dade6c6e81832925ccb9High-dose 17β-estradiol treatment prevents development of heart failure post-myocardial infarction in the ratBeer, Stephanie; Reincke, Martin; Kral, Maike; Callies, Frank; Stroemer, Hinrik; Dienesch, Charlotte; Steinhauer, Sonja; Ertl, Georg; Allolio, Bruno; Neubauer, StefanBasic Research in Cardiology (2007), 102 (1), 9-18CODEN: BRCAB7; ISSN:0300-8428. (Steinkopff Verlag)Prognosis of heart failure remains poor despite therapeutic advances, such as angiotensin converting enzyme inhibition or β-receptor blockade. Thus, more effective forms of treatment are urgently needed. Since estrogens have been shown to modulate migration and proliferation of cardiac fibroblasts and to modulate the expression of estrogen receptors of cardiomyocytes we examd. whether high-dose estrogen treatment can affect post-myocardial infarction left ventricular remodeling. Female rats were treated with 17β-estradiol (7.5 mg/90 d) or placebo for ten weeks, starting two weeks prior to exptl. myocardial infarction. Eight weeks after infarction, in vivo echocardiog. and hemodynamic measurements as well as isolated heart perfusion were performed. In vivo, chronic estrogen treatment almost completely prevented the development of all signs of heart failure that occur in untreated infarcted hearts, such as increased left ventricular diams. (dilatation), reduced fractional shortening (systolic dysfunction) or increased left ventricular end-diastolic pressure (diastolic dysfunction). In vitro, the right- (indicating structural dilatation) and downward (indicating left ventricular dysfunction) shift of left ventricular pressure-vol. curves occurring in untreated infarcted hearts was completely prevented by estrogen. High dose estradiol treatment prevented development of post-MI remodeling, as assessed by in vivo and in vitro parameters of LV dysfunction. Estrogen may hold the potential of becoming a new form of heart failure treatment. However, the mechanisms responsible for this striking and unexpected beneficial action of estrogen in heart failure remain to be elucidated.
- 13Gardner, J. D.; Murray, D. B.; Voloshenyuk, T. G.; Brower, G. L.; Bradley, J. M.; Janicki, J. S. Estrogen Attenuates Chronic Volume Overload Induced Structural and Functional Remodeling in Male Rat Hearts. Am. J. Physiol.-Heart Circ. Physiol. 2010, 298 (2), H497– H504, DOI: 10.1152/ajpheart.00336.2009Google ScholarThere is no corresponding record for this reference.
- 14Donaldson, C.; Eder, S.; Baker, C.; Aronovitz, M. J.; Weiss, A. D.; Hall-Porter, M.; Wang, F.; Ackerman, A.; Karas, R. H.; Molkentin, J. D.; Patten, R. D. Estrogen Attenuates Left Ventricular and Cardiomyocyte Hypertrophy by an Estrogen Receptor–Dependent Pathway That Increases Calcineurin Degradation. Circ. Res. 2009, 104 (2), 265– 275, DOI: 10.1161/CIRCRESAHA.108.190397Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXntFahsw%253D%253D&md5=c59457053ff82df0c1197e958017a27eEstrogen attenuates left ventricular and cardiomyocyte hypertrophy by an estrogen receptor-dependent pathway That Increases Calcineurin DegradationDonaldson, Cameron; Eder, Sarah; Baker, Corey; Aronovitz, Mark J.; Weiss, Alexandra Dabreo; Hall-Porter, Monica; Wang, Feng; Ackerman, Adam; Karas, Richard H.; Molkentin, Jeffery D.; Patten, Richard D.Circulation Research (2009), 104 (2), 265-275CODEN: CIRUAL; ISSN:0009-7330. (Lippincott Williams & Wilkins)Left ventricular (LV) hypertrophy commonly develops in response to chronic hypertension and is a significant risk factor for heart failure and death. The serine-threonine phosphatase calcineurin (Cn)A plays a crit. role in the development of pathol. hypertrophy. Previous exptl. studies in murine models show that estrogen limits pressure overload-induced hypertrophy; our purpose was to explore further the mechanisms underlying this estrogen effect. Wild-type, ovariectomized female mice were treated with placebo or 17β-estradiol (E2), followed by transverse aortic constriction (TAC), to induce pressure overload. At 2 wk, mice underwent physiol. evaluation, immediate tissue harvest, or dispersion of cardiomyocytes. E2 replacement limited TAC-induced LV and cardiomyocyte hypertrophy while attenuating deterioration in LV systolic function and contractility. These E2 effects were assocd. with reduced abundance of CnA. The primary downstream targets of CnA are the nuclear factor of activated T-cell (NFAT) family of transcription factors. In transgenic mice expressing a NFAT-activated promoter/luciferase reporter gene, E2 limited TAC-induced activation of NFAT. Moreover, the inhibitory effects of E2 on LV hypertrophy were absent in CnA knockout mice, supporting the notion that CnA is an important target of E2-mediated inhibition. In cultured rat cardiac myocytes, E2 inhibited agonist-induced hypertrophy while also decreasing CnA abundance and NFAT activation. Agonist stimulation also reduced CnA ubiquitination and degrdn. that was prevented by E2; all in vitro effects of estrogen were reversed by an estrogen receptor (ER) antagonist. These data support that E2 reduces pressure overload induced hypertrophy by an ER-dependent mechanism that increases CnA degrdn., unveiling a novel mechanism by which E2 and ERs regulate pathol. LV and cardiomyocyte growth.
- 15Arias-Loza, P.-A.; Muehlfelder, M.; Elmore, S. A.; Maronpot, R.; Hu, K.; Blode, H.; Hegele-Hartung, C.; Fritzemeier, K. H.; Ertl, G.; Pelzer, T. Differential Effects of 17β-Estradiol and of Synthetic Progestins on Aldosterone-Salt–Induced Kidney Disease. Toxicol. Pathol. 2009, 37 (7), 969– 982, DOI: 10.1177/0192623309350475Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXmvVKrsQ%253D%253D&md5=b57c3c86b1e1591cb504a0513f7c60f4Differential effects of 17β-estradiol and of synthetic progestins on aldosterone-salt-induced kidney diseaseArias-Loza, Paula-Anahi; Muehlfelder, Melanie; Elmore, Susan A.; Maronpot, Robert; Hu, Kai; Blode, Hartmut; Hegele-Hartung, Christa; Fritzemeier, Karl Heinrich; Ertl, Georg; Pelzer, TheoToxicologic Pathology (2009), 37 (7), 969-982CODEN: TOPADD; ISSN:0192-6233. (Sage Publications)Elevated mineralocorticoid levels and female sex hormones have been shown to confer opposing effects on renal injury, but their combined effects are still unknown. Identify the function of estrogens and of different synthetic progestins on aldosterone salt-mediated renal disease. The role of 17β-estradiol, medroxyprogesterone acetate (MPA), and drospirenone during renal injury was studied in Wistar rats subjected to uni-nephrectomy plus aldosterone salt treatment. Aldo-salt treatment of intact, ovariectomized, and estradiol-treated female rats resulted in remnant kidney hypertrophy without structural damage. Co-treatment with MPA, but not with drospirenone, increased kidney hypertrophy, fluid turnover, sodium retention, and potassium excretion. Medroxyprogesterone acetate also caused glomerular, vascular, tubular, and interstitial lesions that were accompanied by increased blood pressure and enhanced NADPH oxidase (p67phox) and sodium channel (α-ENaC) expression. Drospirenone, a progestin with anti-mineralocorticoid function, and spironolactone prevented kidney hypertrophy, hypertension, and sodium retention. Drospirenone and spironolactone also increased renal angiotensin II type 2 receptor expression and relieved aldosterone-induced suppression of serum angiotensin II levels. The choice of specific synthetic progestins has profound implications on the development of kidney injury and renal gene expression under conditions of elevated aldosterone serum levels and salt intake.
- 16Kwan, M. L.; Yao, S.; Laurent, C. A.; Roh, J. M.; Quesenberry, C. P.; Kushi, L. H.; Lo, J. C. Changes in Bone Mineral Density in Women with Breast Cancer Receiving Aromatase Inhibitor Therapy. Breast Cancer Res. Treat. 2018, 168 (2), 523– 530, DOI: 10.1007/s10549-017-4626-5Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhvF2gt7jJ&md5=8f674ba15febd9a0f96926c4942a75f3Changes in bone mineral density in women with breast cancer receiving aromatase inhibitor therapyKwan, Marilyn L.; Yao, Song; Laurent, Cecile A.; Roh, Janise M.; Quesenberry, Charles P., Jr.; Kushi, Lawrence H.; Lo, Joan C.Breast Cancer Research and Treatment (2018), 168 (2), 523-530CODEN: BCTRD6; ISSN:0167-6806. (Springer)Purpose: We assessed bone mineral d. (BMD) change with aromatase inhibitor (AI) treatment in a contemporary cohort of women with breast cancer treated in Kaiser Permanente Northern California. Methods: Percent and estd. annual percent changes in BMD at the total hip and lumbar spine were examd. in 676 women receiving AI therapy who had two serial BMD reports available (at least 1 yr apart) before and after AI initiation (N = 317) or during continued AI therapy (N = 359). BMD changes were examd. at the total hip and lumbar spine and compared by age and clin. subgroups. Results: Women experienced BMD declines after AI initiation or continued therapy, with median annual percent change - 1.2% (interquartile range, IQR - 2.4 to - 0.1%) at the hip and - 1.0% (IQR - 2.3 to 0.1%) at the spine after AI initiation, and - 1.1% (IQR - 2.4 to 0.1%) at the hip and - 0.9% (IQR - 2.4 to 0.5%) at the spine during continued therapy. Higher levels of bone loss were obsd. among younger (< 55 years) compared with older (≥ 75 years) women at the hip (- 1.6% vs. - 0.8%) and at the spine (- 1.5% vs. - 0.5%) after AI initiation, and at the hip (- 1.4% vs. - 1.2%) and at the spine (- 2.4% vs. - 0.001%) during continued therapy. Conclusions: Small but consistent declines in total hip and lumbar spine BMD were present in breast cancer patients following AI therapy initiation or continued AI therapy. Although the overall rates of osteoporosis were low, greater estd. levels of annual bone loss were evident among women < 55 years.
- 17Tian, W.; Wu, M.; Deng, Y. Comparison of Changes in the Lipid Profiles of Eastern Chinese Postmenopausal Women With Early-Stage Breast Cancer Treated With Different Aromatase Inhibitors: A Retrospective Study. Clin. Pharmacol. Drug Dev. 2018, 7 (8), 837– 843, DOI: 10.1002/cpdd.420Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFGqurfJ&md5=4aeb5a224fb921e1a678a9f84efbccf5Comparison of Changes in the Lipid Profiles of Eastern Chinese Postmenopausal Women With Early-Stage Breast Cancer Treated With Different Aromatase Inhibitors: A Retrospective StudyTian, Wei; Wu, Miaowei; Deng, YongchuanClinical Pharmacology in Drug Development (2018), 7 (8), 837-843CODEN: CPDDAH; ISSN:2160-7648. (John Wiley & Sons, Inc.)Cardiovascular morbidity is closely assocd. with serum lipid level. We aimed to investigate the effects of different aromatase inhibitors, including letrozole, anastrozole, and exemestane, on the lipid profile of eastern Chinese breast cancer patients. We evaluated a retrospective cohort of eastern Chinese postmenopausal women with early-stage breast cancer who received aromatase inhibitors. A total of 116 postmenopausal women with early-stage breast cancer without prior cardiovascular disease were included. Lipid changes at 3, 6, 12, and 24 mo were compared across the endocrine therapy categories. Our data demonstrated that exemestane treatment significantly decreased triglyceride level compared with letrozole after 24 mo. However, the aromatase inhibitors had almost equiv. impacts on high-d. liportein cholesterol, low-d. lipoprotein cholesterol, and triglyceride after long-term aromatase inhibitor treatment. As a small-size retrospective study, our data do not support a judgment about whether one AI or another carries more or less risk in terms of lipid disorders in eastern Chinese breast cancer patients. The exact effects need further randomized, controlled trials to investigate.
- 18Castelli, W. P. Cardiovascular Disease in Women. Am. J. Obstet. Gynecol. 1988, 158 (6), 1553– 1560, DOI: 10.1016/0002-9378(88)90189-5Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaL1c3jvVKqtA%253D%253D&md5=7e37bdc4dfdc154b9e8f1ed6bfe3f223Cardiovascular disease in womenCastelli W PAmerican journal of obstetrics and gynecology (1988), 158 (6 Pt 2), 1553-60, 1566-7 ISSN:0002-9378.Cardiovascular disease is the leading cause of death in American women. Atherosclerotic diseases, primarily myocardial infarction and stroke, are important causes of morbidity and mortality among women. Contrary to popular belief, the actual number of myocardial infarctions in men and women is similar, although women develop cardiovascular disease an average of 10 years later than do men. Total serum cholesterol level is a major indicator of risk of coronary heart disease; for every 1% increase in the total serum cholesterol level, a 2% increase in incidence of coronary heart disease is found. A high level of low-density lipoproteins is an independent risk factor for coronary heart disease in both mean and women, but high triglyceride level is an independent risk factor only in women. Conversely, the higher the level of high-density lipoprotein, the lower the risk of coronary heart disease. To reduce the risk of coronary heart disease, target levels of total serum cholesterol must be lowered from 300 to 200 mg/dl. If the total cholesterol/high-density lipoprotein ratio is greater than or equal to 4.5 of if the low-density lipoprotein concentration is greater than 150 mg/dl, the patient is at high risk for coronary heart disease. Clinical trials that use diet or drugs to lower serum cholesterol levels have consistently shown a 2% reduction in the incidence of coronary heart disease for every 1% reduction in total serum cholesterol level.
- 19Fischer, M. Renin Angiotensin System and Gender Differences in the Cardiovascular System. Cardiovasc. Res. 2002, 53 (3), 672– 677, DOI: 10.1016/S0008-6363(01)00479-5Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XhtleqsLk%253D&md5=b8d7cee6b950772eff8fd78a49878084Renin angiotensin system and gender differences in the cardiovascular systemFischer, Marcus; Baessler, Andrea; Schunkert, HeribertCardiovascular Research (2002), 53 (3), 672-677CODEN: CVREAU; ISSN:0008-6363. (Elsevier Science B.V.)A review. To explain gender-related differences of the cardiovascular system, the renin-angiotensin system experienced intensive exploration. Indeed, the development of hypertension as well as the progression of coronary artery disease and heart failure have 2 factors in common: (1) display distinct gender specific characteristics and (2) are enhanced by the renin-angiotensin system. It is therefore interesting to note that data from exptl. animals, epidemiol. surveys, and clin. investigations suggest that the components of the circulating as well as tissue-based renin-angiotensin system are markedly affected by gender. However, the issue is complicated by counter-regulatory effects of estrogen on the system with the substrate, on one hand, and the processing enzymes as well as the chief receptor, on the other hand. In fact, angiotensinogen is up-regulated particularly by oral administration of estrogen, whereas renin, angiotensin-converting enzyme (ACE), and AT-1 receptor are down-regulated by the hormone. While under well-defined exptl. conditions the net effect of estrogen appears to result in suppression of the renin-angiotensin system, the clin. situation may be more complex. The judgment is further complicated by the difficulty in precisely measuring the activity of the system at the tissue level. Moreover, clin. relevant read-outs for the activity of the renin-angiotensin system may be regulated multifactorially or only indirectly affected by the system. Nevertheless, the undisputable, profound biochem. changes in the renin-angiotensin system related to the estrogen status allow speculation that such interaction explains some of the differences in the cardiovascular system of men and women.
- 20Roesch, D. M.; Tian, Y.; Zheng, W.; Shi, M.; Verbalis, J. G.; Sandberg, K. Estradiol Attenuates Angiotensin-Induced Aldosterone Secretion in Ovariectomized Rats. Endocrinology 2000, 141 (12), 4629– 4636, DOI: 10.1210/endo.141.12.7822Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXosVSqsbY%253D&md5=bd78f0ea5d5b9cf3f78dda265373702cEstradiol attenuates angiotensin-induced aldosterone secretion in ovariectomized ratsRoesch, Darren M.; Tian, Ying; Zheng, Wei; Shi, Min; Verbalis, Joseph G.; Sandberg, KathrynEndocrinology (2000), 141 (12), 4629-4636CODEN: ENDOAO; ISSN:0013-7227. (Endocrine Society)Estrogen replacement therapy significantly reduces the risk of cardiovascular disease in postmenopausal women. Previous studies indicate that estradiol (E2) decreases angiotensin II (AT) receptor d. in the adrenal and pituitary in NaCl-loaded rats. We used an in vivo model that eliminates the potentially confounding influence of ACTH to det. whether the E2-induced decrease in adrenal AT receptor expression affects aldosterone responses to angiotensin II (Ang II). Female rats were ovariectomized, treated with oil (OVX) or E2(OVX+E2; 10 μg, s.c.) for 14 days, and fed a NaCl-deficient diet for the last 7 days to maximize adrenal AT receptor expression and responsiveness. On days 12-14 rats were treated with dexamethasone (DEX; 25 μg, i.p., every 12 h) to suppress plasma ACTH. On day 14 aldosterone secretion was measured after a 30-min infusion of Ang II (330 ng/min). Ang II infusion increased the peak plasma aldosterone levels to a lesser degree in the OVX+E2 than in the OVX rats (OVX, 1870 pg/mL; OVX+E2, 1010 pg/mL). Ang II-induced ACTH and aldosterone secretion was also studied in rats that were not treated with DEX. In the absence of DEX, the peak plasma aldosterone response was also significantly decreased (OVX, 5360 pg/mL; OVX+E2, 2960 pg/mL). However, E2 also reduced the plasma ACTH response to Ang II (OVX, 220 pg/mL; OVX+E2, 160 pg/mL), suggesting that reduced pituitary ACTH responsiveness to Ang II contributes to the effect of E2 on Ang II-induced aldosterone secretion. Adrenal AT1 binding studies confirmed that E2 significantly reduces adrenal AT1 receptor expression in both the presence and absence of DEX in NaCl-deprived rats. These results indicate that E2-induced decreases in pituitary and adrenal AT1 receptor expression are assocd. with attenuated pituitary ACTH and adrenal aldosterone responses to Ang II and suggest that estrogen replacement therapy may modulate Ang II-stimulated aldosterone secretion as part of its well known cardioprotective actions.
- 21Chappell, M. C.; Gallagher, P. E.; Averill, D. B.; Ferrario, C. M.; Brosnihan, K. B. Estrogen or the AT1 Antagonist Olmesartan Reverses the Development of Profound Hypertension in the Congenic mRen2.Lewis Rat. Hypertension 2003, 42 (4), 781– 786, DOI: 10.1161/01.HYP.0000085210.66399.A3Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXosF2rtb4%253D&md5=7901d1ae6145064874ee2b2793ce3b7aEstrogen or the AT1 antagonist olmesartan reverses the development of profound hypertension in the congenic mRen2.Lewis ratChappell, Mark C.; Gallagher, Patricia E.; Averill, David B.; Ferrario, Carlos M.; Brosnihan, K. BridgetHypertension (2003), 42 (4, Pt. 2), 781-786CODEN: HPRTDN; ISSN:0194-911X. (Lippincott Williams & Wilkins)The influence of estrogen on the regulation of cardiovascular function remains a controversial and complex area of investigation. We assessed the effects of estrogen depletion in the congenic mRen(2).Lewis rat, established from the back-cross of the original (mRen2)-27 transgenic onto the Lewis inbred strain. Ovariectomy of heterozygous mRen(2).Lewis at 4 to 5 wk resulted in a progressive increase in blood pressure compared with the sham surgery congenics at weeks 6 to 11. At 11 wk, the ovariectomized mRen(2).Lewis (OVX) systolic blood pressure averaged 195±3.7 mm Hg vs. 141±4.0 mm Hg for sham. Plasma Angiotensin (Ang) II, serum ACE activity, plasma renin concn., as well as urinary excretion of Ang II, 8-isoprostane F2α, and endothelin-1 were elevated; however, renal mRNA levels of eNOS were suppressed after ovariectomy. Estrogen replacement reduced blood pressure below both the sham and OVX by 11 wk (125±2.9 mm Hg, n=7, P<0.01 vs. OVX and sham). Moreover, the AT1 receptor antagonist olmesartan (CS866; week 12 to 16) essentially normalized blood pressure to 113±5.4 mm Hg (n=6, P<0.01 vs. OVX and sham). The attenuation of the hypertension was still evident 7 wk after complete withdrawal of treatment (124±4.1 mm Hg at week 23). In summary, the OVX mRen.2.Lewis exhibited a rapid and sustained increase in blood pressure. Estrogen or olmesartan lowered pressure by a similar extent. We conclude that the ovary exerts considerable influence on the regulation of the blood pressure in the mRen2.Lewis strain, possibly by limiting activation of the renin-angiotensin system.
- 22Harrison-Bernard, L. M.; Schulman, I. H.; Raij, L. Postovariectomy Hypertension Is Linked to Increased Renal AT1 Receptor and Salt Sensitivity. Hypertension 2003, 42 (6), 1157– 1163, DOI: 10.1161/01.HYP.0000102180.13341.50Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXpsVGis78%253D&md5=75c66cb3b3beeb4857adbb5aa243bd1dPostovariectomy hypertension is linked to increased renal AT1 receptor and salt sensitivityHarrison-Bernard, Lisa M.; Schulman, Ivonne Hernandez; Raij, LeopoldoHypertension (2003), 42 (6), 1157-1163CODEN: HPRTDN; ISSN:0194-911X. (Lippincott Williams & Wilkins)The functional balance between angiotensin II (Ang II) and nitric oxide (NO) plays a key role in modulating salt sensitivity. Estrogen has been shown to downregulate angiotensin type 1 (AT1) receptor expression and to increase the bioavailability of endothelium-derived NO, which decreases AT1 receptor expression. The present study tests the hypothesis that in the presence of genetic salt sensitivity, deficiency of endogenous estrogens after ovariectomy (OVX) fosters an upregulation of Ang II. Female Dahl salt-resistant (DR), Dahl salt-sensitive (DS), Wistar-Kyoto (WKY), and spontaneously hypertensive (SHR) rats underwent bilateral OVX or sham surgery (SHX) and were fed a normal salt diet (0.5% NaCl) for 14 wk. Systolic blood pressures were measured every 2 wk and were not significantly different between OVX and SHX for DR, WKY, and SHR groups. However, at the end of 14 wk of normal salt diet, hypertension developed in DS OVX but not SHX rats (160±3 vs. 136±3 mm Hg; P<0.05). Hypertension also developed in DS OVX rats pair-fed a normal salt diet (166±7 mm Hg). Development of hypertension in DS OVX rats was prevented by estrogen replacement (132±3 mm Hg), AT1 receptor blockade (119±3 mm Hg), or feeding a very low salt diet (0.1% NaCl; 129±4 mm Hg). Renal AT1 receptor protein expression was significantly elevated 2-fold in DS OVX relative to SHX rats and was prevented by estrogen replacement. These data strongly suggest that after OVX in salt-sensitive rats there is a lower threshold for the hypertensinogenic effect of salt that is linked to an activation of Ang II.
- 23Krishnamurthi, K.; Verbalis, J. G.; Zheng, W.; Wu, Z.; Clerch, L. B.; Sandberg, K. Estrogen Regulates Angiotensin AT1 Receptor Expression via Cytosolic Proteins That Bind to the 52 Leader Sequence of the Receptor mRNA. Endocrinology 1999, 140 (11), 5435– 5438, DOI: 10.1210/endo.140.11.7242Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXmvFemuro%253D&md5=553e5501aa97bd6969ba135d482c7ef2Estrogen regulates angiotensin AT1 receptor expression via cytosolic proteins that bind to the 5' leader sequence of the receptor mRNAKrishnamurthi, Kamakshi; Verbalis, Joseph G.; Zheng, Wei; Wu, Zheng; Clerch, Linda B.; Sandberg, KathrynEndocrinology (1999), 140 (11), 5435-5438CODEN: ENDOAO; ISSN:0013-7227. (Endocrine Society)Two of the most highly recognized factors implicated in the pathogenesis of hypertension, atherosclerosis, congestive heart failure and assocd. cardiovascular disease are the renin-angiotensin system (RAS) and estrogen. A major effect of estrogen results from its influence on the RAS. β-Estradiol (E2) replacement in ovariectomized (OVX) rats significantly decreased type 1 angiotensin (AT1) receptor expression in the pituitary and adrenal, whereas it significantly increased receptor expression in the uterus when compared to OVX controls. Addnl. evidence demonstrated an important influence of estrogen on a recently discovered post-transcriptional mechanism for regulating expression of the AT1 receptor. This mechanism consists of cytosolic RNA binding proteins (BPs) that recognize the 5' leader sequence (5'LS) of the receptor mRNA. The activities of these 5'LS BPs were modulated by estrogen in an inverse manner to AT1 receptor regulation. Moreover, in vitro translation assays in wheat germ lysates suggested that the 5'LS BPs inhibited AT1 receptor translation. Our data therefore indicate that hormonal regulation of AT1 receptors involves modulation of 5'LS BPs by estrogen. These findings may in part account for the obsd. protective effects of estrogen on cardiovascular disease.
- 24Ries, C.; Lucas, S.; Heim, R.; Birk, B.; Hartmann, R. W. Selective Aldosterone Synthase Inhibitors Reduce Aldosterone Formation in Vitro and in Vivo. J. Steroid Biochem. Mol. Biol. 2009, 116 (3–5), 121– 126, DOI: 10.1016/j.jsbmb.2009.04.013Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXptVGmt70%253D&md5=a03f287e26dc3bfff893dd9f3e5d0541Selective aldosterone synthase inhibitors reduce aldosterone formation in vitro and in vivoRies, Christina; Lucas, Simon; Heim, Ralf; Birk, Barbara; Hartmann, Rolf W.Journal of Steroid Biochemistry and Molecular Biology (2009), 116 (3-5), 121-126CODEN: JSBBEZ; ISSN:0960-0760. (Elsevier Ltd.)Aldosterone plays a crucial role in salt and water homeostasis but in case of pathol. increased plasma aldosterone levels it is also involved in the development and the progression of severe cardiovascular diseases like heart failure and myocardial fibrosis. For the treatment of these diseases we propose inhibition of the aldosterone forming enzyme CYP11B2 as a new pharmacol. strategy. We recently developed in vitro highly potent and selective inhibitors of human CYP11B2, but the evidence of their in vivo activity is still missing. For this purpose, rat aldosterone synthase gene was cloned and expressed in V79MZ cells to establish a new screening assay for the identification of "rat-active" substances. Compds. from the class of heteroaryl substituted 3,4-dihydro-1H-quinolin-2-ones showed a moderate inhibitory effect (65% at 2 μM) on rat CYP11B2 in vitro. Furthermore, it diminished the conversion of deoxycorticosterone to aldosterone in rat adrenals and significantly reduced plasma aldosterone levels in vivo.
- 25Anighoro, A.; Bajorath, J.; Rastelli, G. Polypharmacology: Challenges and Opportunities in Drug Discovery. J. Med. Chem. 2014, 57 (19), 7874– 7887, DOI: 10.1021/jm5006463Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtVShsL3I&md5=5ab36c961a84462d90bc25130d90a281Polypharmacology: Challenges and Opportunities in Drug DiscoveryAnighoro, Andrew; Bajorath, Jurgen; Rastelli, GiulioJournal of Medicinal Chemistry (2014), 57 (19), 7874-7887CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. At present, the legendary magic bullet, i.e., a drug with high potency and selectivity toward a specific biol. target, shares the spotlight with an emerging and alternative polypharmacol. approach. Polypharmacol. suggests that more effective drugs can be developed by specifically modulating multiple targets. It is generally thought that complex diseases such as cancer and central nervous system diseases may require complex therapeutic approaches. In this respect, a drug that "hits" multiple sensitive nodes belonging to a network of interacting targets offers the potential for higher efficacy and may limit drawbacks generally arising from the use of a single-target drug or a combination of multiple drugs. In this review, we will compare advantages and disadvantages of multitarget vs. combination therapies, discuss potential drug promiscuity arising from off-target effects, comment on drug repurposing, and introduce approaches to the computational design of multitarget drugs.
- 26Pinzi, L.; Tinivella, A.; Gagliardelli, L.; Beneventano, D.; Rastelli, G. LigAdvisor: A Versatile and User-Friendly Web-Platform for Drug Design. Nucleic Acids Res. 2021, 49 (W1), W326– W335, DOI: 10.1093/nar/gkab385Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvFWgtbrO&md5=eee342dee01ed98315cd33d2f25a288dLigAdvisor: a versatile and user-friendly web-platform for drug designPinzi, Luca; Tinivella, Annachiara; Gagliardelli, Luca; Beneventano, Domenico; Rastelli, GiulioNucleic Acids Research (2021), 49 (W1), W326-W335CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)Although several tools facilitating in silico drug design are available, their results are usually difficult to integrate with publicly available information or require further processing to be fully exploited. The rational design of multi-target ligands (polypharmacol.) and the repositioning of known drugs towards unmet therapeutic needs (drug repurposing) have raised increasing attention in drug discovery, although they usually require careful planning of tailored drug design strategies. Computational tools and data-driven approaches can help to reveal novel valuable opportunities in these contexts, as they enable to efficiently mine publicly available chem., biol., clin., and disease-related data. Based on these premises, we developed LigAdvisor, a data-driven webserver which integrates information reported in DrugBank, Protein Data Bank, UniProt, Clin. Trials and Therapeutic Target Database into an intuitive platform, to facilitate drug discovery tasks as drug repurposing, polypharmacol., target fishing and profiling. As designed, LigAdvisor enables easy integration of similarity estn. results with clin. data, thereby allowing a more efficient exploitation of information in different drug discovery contexts. Users can also develop customizable drug design tasks on their own mols., by means of ligand- and target-based search modes, and download their results.
- 27Ankley, G. T.; Kahl, M. D.; Jensen, K. M.; Hornung, M. W.; Korte, J. J.; Makynen, E. A.; Leino, R. L. Evaluation of the Aromatase Inhibitor Fadrozole in a Short-Term Reproduction Assay with the Fathead Minnow (Pimephales Promelas). Toxicol. Sci. 2002, 67 (1), 121– 130, DOI: 10.1093/toxsci/67.1.121Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XjtlGmsLw%253D&md5=02e4e03e90dee1df37ed1bffe02e6188Evaluation of the aromatase inhibitor fadrozole in a short-term reproduction assay with the fathead minnow (Pimephales promelas)Ankley, Gerald T.; Kahl, Michael D.; Jensen, Kathleen M.; Hornung, Michael W.; Korte, Joseph J.; Makynen, Elizabeth A.; Leino, Richard L.Toxicological Sciences (2002), 67 (1), 121-130CODEN: TOSCF2; ISSN:1096-6080. (Oxford University Press)Cytochrome P 450 aromatase (CYP19) is a key enzyme in vertebrate steroidogenesis, catalyzing the conversion of C19 androgens to C18 estrogens such as β-estradiol (E2). The objective of this study was to assess effects of the CYP19 inhibitor fadrozole on fathead minnow (Pimephales promelas) reproductive endocrinol. and physiol. in a short-term reprodn. assay proposed for identifying specific classes of endocrine-disrupting chems. A concn.-dependent redn. in fecundity was obsd. in fish exposed for 21 days to water concns. of fadrozole ranging from 2 to 50 μg/l. Consistent with the expected mechanism of action, there was a significant inhibition of brain aromatase activity in both male and female fathead minnows exposed to fadrozole. In females, this inhibition was accompanied by a concn.-dependent decrease in plasma E2 and vitellogenin concns.; the latter observation is consistent with the fact that activation of the estrogen receptor by E2 initiates hepatic vitellogenin prodn. in oviparous vertebrates. Histol. assessment of ovaries from females exposed to fadrozole indicated a decrease in mature oocytes and an increase in preovulatory atretic follicles. Exposure of male fathead minnows to fadrozole significantly increased plasma concns. of the androgens testosterone (T) and 11-ketotestosterone (KT) and resulted in a marked accumulation of sperm in the tests. Results of this study indicate that the proposed fathead minnow assay should effectively identify test chems. as potential aromatase inhibitors, both in the context of their reproductive toxicity and the specific mechanism of action. These results also should be of utility in assessing the potential ecol. risk of CYP19 inhibitors, in particular in the context of relating alterations in subcellular indicators of endocrine function (changes in steroids, proteins) to adverse consequences in the whole organism.
- 28Browne, L. J.; Gude, C.; Rodriguez, H.; Steele, R. E.; Bhatnager, A. Fadrozole Hydrochloride: A Potent, Selective, Nonsteroidal Inhibitor of Aromatase for the Treatment of Estrogen-Dependent Disease. J. Med. Chem. 1991, 34 (2), 725– 736, DOI: 10.1021/jm00106a038Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXhtVWrsLg%253D&md5=a154e38c3cdb14113a3fefbddf8781eeFadrozole hydrochloride: a potent, selective, nonsteroidal inhibitor of aromatase for the treatment of estrogen-dependent diseaseBrowne, L. J.; Gude, C.; Rodriguez, H.; Steele, R. E.; Bhatnager, A.Journal of Medicinal Chemistry (1991), 34 (2), 725-36CODEN: JMCMAR; ISSN:0022-2623.The prepn. of 5-phenyl-5,6,7,8-tetrahydroimidazopyridine derivs. I (R = cyano, Br, CH2OH, Me, CO2H, CO2Et) and their evaluation as inhibitors for aromatase and estrogen prodn. was described; the most potent aromatase inhibitor was (+)-I (R = cyano), i.e. fadrozole (CGS 16949A). The mol. structure-activity relationship was discussed.
- 29Ménard, J.; Pascoe, L. Can the Dextroenantiomer of the Aromatase Inhibitor Fadrozole Be Useful for Clinical Investigation of Aldosterone-Synthase Inhibition?. J. Hypertens. 2006, 24 (6), 993– 997, DOI: 10.1097/01.hjh.0000226183.98439.b3Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xktlartrk%253D&md5=e56a1f5ec586a4dfa515f6f995f731c9Can the dextroenantiomer of the aromatase inhibitor fadrozole be useful for clinical investigation of aldosterone-synthase inhibition?Menard, Joel; Pascoe, LeighJournal of Hypertension (2006), 24 (6), 993-997CODEN: JOHYD3; ISSN:0263-6352. (Lippincott Williams & Wilkins)A review. The beneficial effects of spironolactone, eplerenone, amiloride, and potassium in preventing cardiovascular damage in various exptl. models of salt-induced hypertension can be dissocd. from blood pressure effects, and have drawn attention to the direct genomic and non-genomic actions of aldosterone at the level of the vessels, the heart, and the kidneys. Exposure to endogenous aldosterone could be decreased by direct and specific aldosterone-synthase inhibition. FAD 286A, the dextroenantiomer of the aromatase inhibitor fadrozole, might be a 1st candidate to investigate in humans, the physiol. impact and therapeutic properties of aldosterone-synthase inhibition, esp. in various forms of primary aldosteronism.
- 30Smith, I. E.; Norton, A. Fadrozole and Letrozole in Advanced Breast Cancer: Clinical and Biochemical Effects. Breast Cancer Res. Treat. 1998, 49 (S1), S67– S71, DOI: 10.1023/A:1006005024377Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXnt1Sktbk%253D&md5=34ab654833d3201e0ce7602e3f51984dFadrozole and letrozole in advanced breast cancer: clinical and biochemical effectsSmith, Ian E.; Norton, AlisonBreast Cancer Research and Treatment (1998), 49 (Suppl. 1), S67-S71CODEN: BCTRD6; ISSN:0167-6806. (Kluwer Academic Publishers)A review with 18 refs.
- 31Lamberts, S. W. J.; Bruining, H. A.; Marzouk, H.; Zuiderwijk, J.; Uitterlinden, P.; Blijd, J. J.; Hackeng, W. H. L.; Jong, F. H. D. The New Aromatase Inhibitor CGS-16949A SuppressesAldosterone and Cortisol Production by Human Adrenal Cells in Vitro. J. Clin. Endocrinol. Metab. 1989, 69 (4), 896– 901, DOI: 10.1210/jcem-69-4-896Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1MXlvVOhur4%253D&md5=9c9846ae461e8242a07d225a8b20ebf6The new aromatase inhibitor CGS-16949A suppresses aldosterone and cortisol production by human adrenal cells in vitroLamberts, S. W. J.; Bruining, H. A.; Marzouk, H.; Zuiderwijk, J.; Uitterlinden, P.; Blijd, J. J.; Hackeng, W. H. L.; De Jong, F. H.Journal of Clinical Endocrinology and Metabolism (1989), 69 (4), 896-901CODEN: JCEMAZ; ISSN:0021-972X.CGS-16949A is a new orally active nonsteroidal aromatase inhibitor which is more than 100-fold more potent than aminoglutethimide. This compd. is an imidazole deriv., and therefore, its possible effect on cytochrome P 450-dependent enzyme activities in the adrenal gland was evaluated. In vitro investigations with dispersed normal and hyperplastic human adrenocortical cells showed that CGS-16949A at 10-7-10-6M is a potent 11β-hydroxylase inhibitor, which inhibits ACTH-stimulated cortisol release to a similar extent as an equimolar concn. of metyrapone (IC50 for both compds., 10-7-5 × 10-7M). Etomidate was a more potent 11 β-hydroxylase inhibitor (IC50, ∼10-8M), while 10-7-10-6M ketoconazole caused (via 17α-hydroxylase inhibition) a similar inhibition of cortisol release as 10-7M CGS-16949A (IC50, 10-7-5 × 10-5M). The 11β-hydroxylase inhibition by CGS-16949A was accompanied by a dose-dependent increase in the release of precursor steroids by the adrenocortical cells in vitro, including deoxycortisol, 17-hydroxyprogesterone, and androstenedione. Aldosterone release was suppressed 50% by 10-8M CGS-16949A, while the IC50 for cortisol in the same cells was 10-7M. Aldosterone release by the dispersed adenoma cells obtained from a patient with primary aldosteronism was also suppressed by CGS-16949A. It is concluded that the new nonsteroidal aromatase inhibitor CGS 16949A is an inhibitor of 11β-hydroxylase which is equipotent to metyrapone. At present it is unclear whether the compd. at the dose that causes complete aromatase inhibition in vivo also affects stress-induced cortisol release in man. Also, CGS-16949A exerts a very potent inhibitory effect on normal aldosterone release (IC50, 10-9M) and on a tumorous aldosterone secretion. CGS-16949A might, therefore, be a drug that can be used in the treatment of primary hyperaldosteronism.
- 32Hu, Q.; Yin, L.; Hartmann, R. W. Aldosterone Synthase Inhibitors as Promising Treatments for Mineralocorticoid Dependent Cardiovascular and Renal Diseases: Miniperspective. J. Med. Chem. 2014, 57 (12), 5011– 5022, DOI: 10.1021/jm401430eGoogle Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXotVelsQ%253D%253D&md5=585f8d88a1df8d22382e6a58ab0283b0Aldosterone Synthase Inhibitors as Promising Treatments for Mineralocorticoid Dependent Cardiovascular and Renal DiseasesHu, Qingzhong; Yin, Lina; Hartmann, Rolf W.Journal of Medicinal Chemistry (2014), 57 (12), 5011-5022CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Besides the well-known roles of aldosterone as a mineralocorticoid in regulating homeostasis of electrolytes and vol., recent studies revealed that it is also a potent proinflammation factor inducing reactive oxygen species and up-regulating a panel of fibrosis related genes. Under pathol. circumstances, excessive aldosterone is involved in a lot of chronic diseases, including hypertension, cardiac fibrosis, congestive heart failure, ventricular remodeling, and diabetic nephropathy. Therefore, the inhibition of aldosterone synthase (CYP11B2), which is the pivotal enzyme in aldosterone biosynthesis, was proposed as a superior approach. Expected pharmacodynamic effects have been demonstrated in both animal models and clin. trials after the application of CYP11B2 inhibitors. The importance of selectivity over other steroidogenic CYP enzymes, in particular 11β-hydroxylase (CYP11B1), was also revealed. Recently, much more selective CYP11B2 inhibitors have been reported, which could be promising drug candidates for the treatment of aldosterone related diseases.
- 33Roumen, L.; Peeters, J. W.; Emmen, J. M. A.; Beugels, I. P. E.; Custers, E. M. G.; De Gooyer, M.; Plate, R.; Pieterse, K.; Hilbers, P. A. J.; Smits, J. F. M.; Vekemans, J. A. J.; Leysen, D.; Ottenheijm, H. C. J.; Janssen, H. M.; Hermans, J. J. R. Synthesis, Biological Evaluation, and Molecular Modeling of 1-Benzyl-1H-Imidazoles as Selective Inhibitors of Aldosterone Synthase (CYP11B2). J. Med. Chem. 2010, 53 (4), 1712– 1725, DOI: 10.1021/jm901356dGoogle Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtlCgtrY%253D&md5=95b05f5ad57464a12715c4cf15502b83Synthesis, Biological Evaluation, and Molecular Modeling of 1-Benzyl-1H-imidazoles as Selective Inhibitors of Aldosterone Synthase (CYP11B2)Roumen, Luc; Peeters, Joris W.; Emmen, Judith M. A.; Beugels, Ilona P. E.; Custers, Erica M. G.; de Gooyer, Marcel; Plate, Ralf; Pieterse, Koen; Hilbers, Peter A. J.; Smits, Jos F. M.; Vekemans, Jef A. J.; Leysen, Dirk; Ottenheijm, Harry C. J.; Janssen, Henk M.; Hermans, J. J. RobJournal of Medicinal Chemistry (2010), 53 (4), 1712-1725CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Reducing aldosterone action is beneficial in various major diseases such as heart failure. Currently, this is achieved with mineralocorticoid receptor antagonists, however, aldosterone synthase (CYP11B2) inhibitors may offer a promising alternative. In this study, three-dimensional modeling of CYP11B2 was used to model the binding modes of the natural substrate 18-hydroxycorticosterone and the recently published CYP11B2 inhibitor R-fadrozole (I) as a rational guide to design 44 structurally simple and achiral 1-benzyl-1H-imidazoles. Their syntheses, in vitro inhibitor potencies, and in silico docking are described. Some promising CYP11B2 inhibitors were identified, with the novel lead MOERAS115 (II) displaying an IC50 for CYP11B2 of 1.7 nM, and a CYP11B2 (vs. CYP11B1) selectivity of 16.5, comparable to R-fadrozole (IC50 for CYP11B2 6.0 nM, selectivity 19.8). Mol. docking of the inhibitors in the models enabled the generation of posthoc hypotheses on their binding modes, providing a valuable basis for future studies and further design of CYP11B2 inhibitors.
- 34Weldon, S. M.; Cerny, M. A.; Gueneva-Boucheva, K.; Cogan, D.; Guo, X.; Moss, N.; Parmentier, J.-H.; Richman, J. R.; Reinhart, G. A.; Brown, N. F. Selectivity of BI 689648, a Novel, Highly Selective Aldosterone Synthase Inhibitor: Comparison with FAD286 and LCI699 in Nonhuman Primates. J. Pharmacol. Exp. Ther. 2016, 359 (1), 142– 150, DOI: 10.1124/jpet.116.236463Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvF2jtLrK&md5=584b931b62922f854df7b68b288d5d19Selectivity of BI 689648, a novel, highly selective aldosterone synthase inhibitor: comparison with FAD286 and LCI699 in nonhuman primatesWeldon, Steven M.; Cerny, Matthew A.; Gueneva-Boucheva, Kristina; Cogan, Derek; Guo, Xin; Moss, Neil; Parmentier, Jean-Hugues; Richman, Jeremy R.; Reinhart, Glenn A.; Brown, Nicholas F.Journal of Pharmacology and Experimental Therapeutics (2016), 359 (1), 142-150CODEN: JPETAB; ISSN:1521-0103. (American Society for Pharmacology and Experimental Therapeutics)The mineralocorticoid aldosterone is an important regulator of blood pressure, vol., and electrolyte balance. However, excess aldosterone can be deleterious as a driver of vascular remodeling and tissue fibrosis assocd. with cardiometabolic diseases. Aldosterone synthase (AS) inhibitors (ASI) attenuate the prodn. of aldosterone directly and have been proposed as an alternative to mineralocorticoid receptor antagonists for blocking the pathol. effects of excess aldosterone. Discovery of selective ASIs has been challenging because of the high sequence identity (93%) AS shares with cortisol synthase (CS), and the low identity of rodent AS compared with human (63%). Using cynomolgus (cyno) monkey-based models, we identified BI 689648 [6-(5-methoxymethyl-pyridin-3-yl)-3,4-dihydro-2H-[1,8]naphthyridine-1-carboxylic acid amide], a novel, highly selective ASI that exhibits an in vitro IC50 of 2 nMagainst AS and 300 nm against CS (150-fold selectivity) compared with the recently described ASIs FAD286 [4-(5,6,7,8-tetrahydroimidazo[1,5-a]pyridin-5-yl)benzonitrile] (3 nM AS; 90 nM CS; 40-fold) and LCI699 (4-[(5R)-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-5-yl]-3-fluorobenzonitrile) (10 nM AS; 80 nM CS; 8-fold). After oral administration in cyno monkeys, BI 689648 (5 mg/kg) exhibits a peak plasma concn. of ∼500 nM. For in vivo profiling we used an ACTH-challenge model in which BI 689648 was.20-fold more selective compared with FAD286 and LCI699. Because both FAD286 and LCI699 failed to provide adequate selectivity for CS when tested in patients, the desire for more selective mols. to test the ASI hypothesis remains high. Therefore, highly selective aldosterone synthase inhibitors such as BI 689648 represent an important step forward toward developing ASIs with greater potential for clin. success in cardiometabolic diseases.
- 35Matore, B. W.; Banjare, P.; Singh, J.; Roy, P. P. In Silico Selectivity Modeling of Pyridine and Pyrimidine Based CYP11B1 and CYP11B2 Inhibitors: A Case Study. J. Mol. Graph. Model. 2022, 116, 108238 DOI: 10.1016/j.jmgm.2022.108238Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhsFKns7rN&md5=6df656ead0f6993c7c6f28f08cbe63caIn silico selectivity modeling of pyridine and pyrimidine based CYP11B1 and CYP11B2 inhibitors: A case studyMatore, Balaji Wamanrao; Banjare, Purusottam; Singh, Jagadish; Roy, Partha PratimJournal of Molecular Graphics & Modelling (2022), 116 (), 108238CODEN: JMGMFI; ISSN:1093-3263. (Elsevier Ltd.)Design of selective drug candidates for highly structural similar targets is a challenging task for researchers. The main objective of this study was to explore the selectivity modeling of pyridine and pyrimidine scaffold towards the highly homologous targets CYP11B1 and CYP11B2 enzymes by in silico (Mol. docking and QSAR) approaches. In this regard, a big dataset (n = 228) of CYP11B1 and CYP11B2 inhibitors were gathered and classified based on heterocyclic ring and the exhaustive anal. was carried out for pyridine and pyrimidinescaffolds. The LibDock algorithm was used to explore the binding pattern, screening, and identify the structural feature responsible for the selectivity of the ligands towards the studied targets. Finally, QSAR anal. was done to explore the correlation between various binding parameters and structural features responsible for the inhibitory activity and selectivity of the ligands in a quant. way. The docking and QSAR anal. clearly revealed and distinguished the importance of structural features, functional groups attached for CYP11B2 and CYP11B1 selectivity for pyridine and pyrimidine analogs. Addnl., the docking anal. highlighted the differentiating amino acids residues for selectivity for ligands for each of the enzymes. The results obtained from this research work will be helpful in designing the selective CYP11B1/CYP11B2 inhibitors.
- 36Baumann, M.; Baxendale, I. R. Sustainable Synthesis of Thioimidazoles via Carbohydrate-Based Multicomponent Reactions. Org. Lett. 2014, 16 (23), 6076– 6079, DOI: 10.1021/ol502845hGoogle Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvFOhsbbO&md5=997439ac86734e67b074f3b686748cefSustainable Synthesis of Thioimidazoles via Carbohydrate-Based Multi-component ReactionsBaumann, Marcus; Baxendale, Ian R.Organic Letters (2014), 16 (23), 6076-6079CODEN: ORLEF7; ISSN:1523-7052. (American Chemical Society)The synthesis of diversely functionalized thioimidazoles through a modern variant of the Marckwald reaction is presented. This new protocol utilizes unprotected carbohydrates as well as simple amine salts as sustainable and biorenewable starting materials. Importantly it was discovered that a bifurcated reaction pathway results from using aldoses and ketoses resp., yielding distinct reaction products in a highly selective manner.
- 37Baumann, M.; Baxendale, I. R. A Continuous-Flow Method for the Desulfurization of Substituted Thioimidazoles Applied to the Synthesis of Etomidate Derivatives: A Continuous-Flow Method for the Desulfurization of Substituted Thioimidazoles Applied to the Synthesis of Etomidate Derivatives. Eur. J. Org. Chem. 2017, 2017 (44), 6518– 6524, DOI: 10.1002/ejoc.201700833Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtlCnsL3I&md5=3de7f2177ab7feedb828041bc89baae0A Continuous-Flow Method for the Desulfurization of Substituted Thioimidazoles Applied to the Synthesis of Etomidate DerivativesBaumann, Marcus; Baxendale, Ian R.European Journal of Organic Chemistry (2017), 2017 (44), 6518-6524CODEN: EJOCFK; ISSN:1099-0690. (Wiley-VCH Verlag GmbH & Co. KGaA)A simple yet robust flow set-up for the efficient desulfurization of a series of thioimidazoles is presented, which generates the corresponding imidazole derivs. I (R = C6H5, 4-BrC6H4, 2,6-F2C6H3,etc.) in high yields. The strategic choice of peristaltic over piston pumps allowed reliable delivery of the heterogeneous stream of the thioimidazole substrate into a T-piece where it reacted with NaNO2 in the presence of acetic acid. This approach enabled the controlled and safe formation of the reactive nitrosonium species without uncontrolled exposure to hazardous nitrous oxide byproducts as obsd. in related batch protocols. The value of the resulting imidazole products was further demonstrated by their conversion into various esters II (R = C6H5, 4-BrC6H4, 2,6-F2C6H3, etc.; R1 = Me, Et, i-Pr, CH2CF3) representing new derivs. of the known analgesic etomidate through an efficient one-pot Corey-Gilman-Ganem oxidn. procedure.
- 38Baumann, M.; Baxendale, I. R. A Continuous Flow Synthesis and Derivatization of 1,2,4-Thiadiazoles. Bioorg. Med. Chem. 2017, 25 (23), 6218– 6223, DOI: 10.1016/j.bmc.2017.01.022Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXitVSqtbY%253D&md5=e6aa8834be2f90a733b7aebefa802e30A continuous flow synthesis and derivatization of 1,2,4-thiadiazolesBaumann, Marcus; Baxendale, Ian R.Bioorganic & Medicinal Chemistry (2017), 25 (23), 6218-6223CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)A continuous flow process is presented that enables the efficient synthesis and derivatization of 1,2,4-thiadiazole heterocycles. Special attention was given to the safe handling of the versatile yet hazardous trichloromethanesulfenyl chloride reagent including its in-line quenching in order to eliminate malodorous and corrosive byproducts. Based on this flow method gram quantities of 5-chloro-3-phenyl-1,2,4-thiadiazole was safely prepd. allowing for further elaboration of this valuable building block by reaction with different nitrogen-, sulfur- and oxygen-based nucleophiles. This synthetic approach was subsequently applied to generate a series of bromophenyl-5-chloro-1,2,4-thiadiazoles providing a valuable entry towards further structural diversification on this important heterocyclic scaffold.
- 39Maligres, P. E.; Waters, M. S.; Weissman, S. A.; McWilliams, J. C.; Lewis, S.; Cowen, J.; Reamer, R. A.; Volante, R. P.; Reider, P. J.; Askin, D. Preparation of a Clinically Investigated Ras Farnesyl Transferase Inhibitor. J. Heterocycl. Chem. 2003, 40 (2), 229– 241, DOI: 10.1002/jhet.5570400206Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXjsVamt7k%253D&md5=ed9c15111665cd350692e581b7ae8386Preparation of a clinically investigated ras farnesyl transferase inhibitorMaligres, Peter E.; Waters, Marjorie S.; Weissman, Steven A.; McWilliams, J. Christopher; Lewis, Stephanie; Cowen, Jennifer; Reamer, Robert A.; Volante, R. P.; Reider, Paul J.; Askin, DavidJournal of Heterocyclic Chemistry (2003), 40 (2), 229-241CODEN: JHTCAD; ISSN:0022-152X. (HeteroCorporation)The synthesis of ras farnesyl-protein transferase inhibitor I is described on a multi-kilogram scale. Retrosynthetic anal. reveals chloromethylimidazole II and a piperazinone III as viable precursors. The 1,5-disubstituted imidazole system was regioselectively assembled via an improved Marckwald imidazole synthesis. A new imidazole dethionation procedure was developed to convert the Marckwald mercaptoimidazole product to the desired imidazole. This methodol. was found to be tolerant of a variety of functional groups providing good to excellent yields of 1,5-disubstituted imidazoles. A new Mitsunobu cyclization strategy was developed to prep. the arylpiperazinone fragment III.
- 40Ashburn, T. T.; Thor, K. B. Drug Repositioning: Identifying and Developing New Uses for Existing Drugs. Nat. Rev. Drug Discovery 2004, 3 (8), 673– 683, DOI: 10.1038/nrd1468Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXmtVOhtL8%253D&md5=0145c02423ab81ace6e3b3d92585ccccDrug repositioning: identifying and developing new uses for existing drugsAshburn, Ted T.; Thor, Karl B.Nature Reviews Drug Discovery (2004), 3 (8), 673-683CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Biopharmaceutical companies attempting to increase productivity through novel discovery technologies have fallen short of achieving the desired results. Repositioning existing drugs for new indications could deliver the productivity increases that the industry needs while shifting the locus of prodn. to biotechnol. companies. More and more companies are scanning the existing pharmacopeia for repositioning candidates, and the no. of repositioning success stories is increasing.
- 41March-Vila, E.; Pinzi, L.; Sturm, N.; Tinivella, A.; Engkvist, O.; Chen, H.; Rastelli, G. On the Integration of In Silico Drug Design Methods for Drug Repurposing. Front. Pharmacol. 2017, 8, 298, DOI: 10.3389/fphar.2017.00298Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFentbbP&md5=cf38db8a704a52cddbd6949caca49783On the integration of in silico drug design methods for drug repurposingMarch-Vila, Eric; Pinzi, Luca; Sturm, Noe; Tinivella, Annachiara; Engkvist, Ola; Chen, Hongming; Rastelli, GiulioFrontiers in Pharmacology (2017), 8 (), 298/1-298/7CODEN: FPRHAU; ISSN:1663-9812. (Frontiers Media S.A.)Drug repurposing has become an important branch of drug discovery. Several computational approaches that help to uncover new repurposing opportunities and aid the discovery process have been put forward, or adapted from previous applications. A no. of successful examples are now available. Overall, future developments will greatly benefit from integration of different methods, approaches and disciplines. Steps forward in this direction are expected to help to clarify, and therefore to rationally predict, new drug-target, target-disease, and ultimately drug-disease assocns.
- 42Wishart, D. S.; Feunang, Y. D.; Guo, A. C.; Lo, E. J.; Marcu, A.; Grant, J. R.; Sajed, T.; Johnson, D.; Li, C.; Sayeeda, Z.; Assempour, N.; Iynkkaran, I.; Liu, Y.; Maciejewski, A.; Gale, N.; Wilson, A.; Chin, L.; Cummings, R.; Le, D.; Pon, A.; Knox, C.; Wilson, M. DrugBank 5.0: A Major Update to the DrugBank Database for 2018. Nucleic Acids Res. 2018, 46 (D1), D1074– D1082, DOI: 10.1093/nar/gkx1037Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitlGisbvI&md5=986b28c7ea546596a26dd3ba38f05feeDrugBank 5.0: a major update to the DrugBank database for 2018Wishart, David S.; Feunang, Yannick D.; Guo, An C.; Lo, Elvis J.; Marcu, Ana; Grant, Jason R.; Sajed, Tanvir; Johnson, Daniel; Li, Carin; Sayeeda, Zinat; Assempour, Nazanin; Iynkkaran, Ithayavani; Liu, Yifeng; Maciejewski, Adam; Gale, Nicola; Wilson, Alex; Chin, Lucy; Cummings, Ryan; Le, Diana; Pon, Allison; Knox, Craig; Wilson, MichaelNucleic Acids Research (2018), 46 (D1), D1074-D1082CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)DrugBank is a web-enabled database contg. comprehensivemol. information about drugs, their mechanisms, their interactions and their targets. First described in 2006, Drug- Bank has continued to evolve over the past 12 years in response to marked improvements to web stds. and changing needs for drug research and development. This year's update, DrugBank 5.0, represents the most significant upgrade to the database in more than 10 years. In many cases, existing data content has grown by 100% or more over the last update. For instance, the total no. of investigational drugs in the database has grown by almost 300%, the no. of drug-drug interactions has grown by nearly 600% and the no. of SNP-assocd. drug effects has grown more than 3000%. Significant improvements have been made to the quantity, quality and consistency of drug indications, drug binding data as well as drug-drug and drug-food interactions. A great deal of brand new data have also been added to DrugBank 5.0. This includes information on the influence of hundreds of drugs on metabolite levels (pharmacometabolomics), gene expression levels (pharmacotranscriptomics) and protein expression levels (pharmacoproteomics). New data have also been added on the status of hundreds of newdrug clin. trials and existing drug repurposing trials. Many other important improvements in the content, interface and performance of the DrugBank website have been made and these should greatly enhance its ease of use, utility and potential applications in many areas of pharmacol. research, pharmaceutical science and drug education.
- 43Berman, H. M. The Protein Data Bank. Nucleic Acids Res. 2000, 28 (1), 235– 242, DOI: 10.1093/nar/28.1.235Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXhvVKjt7w%253D&md5=227fb393f754be2be375ab727bfd05dcThe Protein Data BankBerman, Helen M.; Westbrook, John; Feng, Zukang; Gilliland, Gary; Bhat, T. N.; Weissig, Helge; Shindyalov, Ilya N.; Bourne, Philip E.Nucleic Acids Research (2000), 28 (1), 235-242CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)The Protein Data Bank (PDB; http://www.rcsb.org/pdb/)is the single worldwide archive of structural data of biol. macromols. This paper describes the goals of the PDB, the systems in place for data deposition and access, how to obtain further information, and near-term plans for the future development of the resource.
- 44Buzdar, A. U.; Robertson, J. F. R.; Eiermann, W.; Nabholtz, J.-M. An Overview of the Pharmacology and Pharmacokinetics of the Newer Generation Aromatase Inhibitors Anastrozole, Letrozole, and Exemestane. Cancer 2002, 95 (9), 2006– 2016, DOI: 10.1002/cncr.10908Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XovFSqsL4%253D&md5=00257b7cac0222c6e0b067082949fa42An overview of the pharmacology and pharmacokinetics of the newer generation aromatase inhibitors anastrozole, letrozole, and exemestaneBuzdar, Aman U.; Robertson, John F. R.; Eiermann, Wolfgang; Nabholtz, Jean-MarcCancer (New York, NY, United States) (2002), 95 (9), 2006-2016CODEN: CANCAR; ISSN:0008-543X. (John Wiley & Sons, Inc.)A review. The newer generation, nonsteroidal aromatase inhibitors (AIs) anastrozole and letrozole have shown superior efficacy compared with tamoxifen as first-line treatments and compared with megestrol acetate as second-line therapy in postmenopausal women with advanced breast carcinoma. In an open-label, Phase II trial, it was reported that exemestane showed numerical superiority compared with tamoxifen for objective response and clin. benefit. Because these agents ultimately may be administered for periods of up to 5 yr in the adjuvant setting, it is of increasing importance to assess their tolerability and pharmacol. profiles. In the absence of data from direct clin. comparisons, the published literature was reviewed for the clin. pharmacol., pharmacokinetic characteristics, and selectivity profiles of anastrozole, letrozole, and exemestane. At clin. administered doses, the plasma half-lives of anastrozole (1 mg once daily), letrozole (2.5 mg once daily), and exemestane (25 mg once daily) were 41-48 h, 2-4 days, and 27 h, resp. The time to steady-state plasma levels was 7 days for both anastrozole and exemestane and 60 days for letrozole. Androgenic side effects have been reported only with exemestane. Anastrozole treatment had no impact on plasma lipid levels, whereas both letrozole and exemestane had an unfavorable effect on plasma lipid levels. In indirect comparisons, anastrozole showed the highest degree of selectivity compared with letrozole and exemestane in terms of a lack of effect on adrenosteroidogenesis. All three AIs demonstrated clin. efficacy over preexisting treatments. However, there were differences in terms of pharmacokinetics and effects on lipid levels and adrenosteroidogenesis. The long-term clin. significance of these differences remains to be elucidated.
- 45Grimm, S. W.; Dyroff, M. C. Inhibition of Human Drug Metabolizing Cytochromes P450 by Anastrozole, a Potent and Selective Inhibitor of Aromatase. Drug Metab. Dispos. Biol. Fate Chem. 1997, 25 (5), 598– 602Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK2s3pslWgsg%253D%253D&md5=1c1907907372987ae13d9a99d84fbe5aInhibition of human drug metabolizing cytochromes P450 by anastrozole, a potent and selective inhibitor of aromataseGrimm S W; Dyroff M CDrug metabolism and disposition: the biological fate of chemicals (1997), 25 (5), 598-602 ISSN:0090-9556.Anastrozole (2,2'[5(1H-1,2,4-triazol-1-ylmethyl)-1,3-phenylene]- bis(2-methylproprionitrile)) is a potent third-generation inhibitor of aromatase, currently marketed as a treatment for postmenopausal women with advanced breast cancer. While its potency and selectivity for inhibition of estrogen synthesis has been established in both preclinical and clinical studies, this study used in vitro methods to examine the effects of anastrozole on several drug metabolizing CYP enzymes found in human liver. Human liver microsomes were co-incubated with anastrozole and probe substrates for CYP1A2 (phenacetin), CYP2A6 (coumarin), CYP2C9 (tolbutamide), CYP2D6 (dextromethorphan), and CYP3A (nifedipine). The formation of the CYP-specific metabolites following co-incubation with various anastrozole concentrations was determined to establish IC50 and Ki values for these enzymes. While anastrozole did not inhibit CYP2A6 and CYP2D6 activities at concentrations below 500 microM, this compound inhibited CYP1A2, CYP2C9, and CYP3A activities with Ki values of 8, 10, and 10 microM, respectively. Dixon plots used to determine the Ki values for the inhibition of CYP1A2 and CYP3A activities by anastrozole were biphasic, indicating additional lower affinity Ki values. Major metabolites of anastrozole did not retain the ability to inhibit the metabolism of nifedipine (CYP3A). The results of this study indicate that, although anastrozole can inhibit CYP1A2, 2C9, and 3A-mediated catalytic activities, this compound would not be expected to cause clinically significant interactions with other CYP-metabolized drugs at physiologically relevant concentrations achieved during therapy with Arimidex (Zeneca, Ltd., Macclesfield, UK) 1-mg.
- 46Gobbi, S.; Rampa, A.; Belluti, F.; Bisi, A. Nonsteroidal Aromatase Inhibitors for the Treatment of Breast Cancer: An Update. Anticancer Agents Med. Chem. 2014, 14 (1), 54– 65, DOI: 10.2174/18715206113139990306Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3sfitFOltw%253D%253D&md5=5b359f6fa60cc11f7c96cad85e0f6bc6Nonsteroidal aromatase inhibitors for the treatment of breast cancer: an updateGobbi Silvia; Rampa Angela; Belluti Federica; Bisi AlessandraAnti-cancer agents in medicinal chemistry (2014), 14 (1), 54-65 ISSN:.Estrogens are known to be important in breast cancer growth in both pre- and post-menopausal women. Although circulating estrogen concentrations are very low after menopause, peripheral tissues generate sufficient concentrations to stimulate tumor growth. As aromatase is the rate-limiting enzyme in estrogen biosynthesis, inhibitors of this enzyme represent effective targeted therapy for breast cancer. Three compounds are now FDA approved and have become the first-choice endocrine drugs for postmenopausal breast cancer patients, since they are associated with superior activity and better general tolerability when compared with the estrogen receptor modulator tamoxifen. Nevertheless, some questions concerning the use of aromatase inhibitors for the treatment of breast cancer still need to be addressed, mainly related to their side-effects and the development of resistance, making research in this field still appealing. Many research groups, including our own, are still dealing with the search of new compounds that possess aromatase inhibitory properties. In this review an update of the latest achievements in the field of nonsteroidal aromatase inhibitors will be given.
- 47Rotstein, D. M.; Kertesz, D. J.; Walker, K. A. M.; Swinney, D. C. Stereoisomers of Ketoconazole: Preparation and Biological Activity. J. Med. Chem. 1992, 35 (15), 2818– 2825, DOI: 10.1021/jm00093a015Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK38Xks12rsL0%253D&md5=ef996a2a278c12d93edc4edcdfffef22Stereoisomers of ketoconazole: preparation and biological activityRotstein, David M.; Kertesz, Denis J.; Walker, Keith A. M.; Swinney, David C.Journal of Medicinal Chemistry (1992), 35 (15), 2818-25CODEN: JMCMAR; ISSN:0022-2623.The four stereoisomers of the antifungal agent ketoconazole, (2S,4R)-, (2R,4R)-, (2R,4S)-, and (2S,4S)-(imidazolylmethyl)phenyl{[(acetylpiperazinyl)phenoxy]methyl}dioxolanes I, were prepd. and evaluated for their selectivity in inhibiting a no. of cytochrome P 450 enzymes. Thus, (bromomethyl)phenyldioxolanes II condensed with 4-(N-acetylpiperazino)phenol and imidazole to give I. II were prepd. by bromination of 2',4'-dichloroacetophenone followed by reaction with (S)- and (R)-solketal tosylate. Large differences in selectivity among the isomers were obsd. for inhibition of the cytochromes P 450 involved in steroid biosynthesis, whereas little difference was obsd. for inhibition of those assocd. with hepatic drug metab. The cis-(2S,4R) and trans-(2R,4R) isomers are equipotent in inhibiting corticoid 11β-hydroxylase and much more effective than their antipodes. Little selectivity was obsd. for inhibition of cholesterol side chain cleavage or xenobiotic hydroxylases. These data indicate that the affinity of azoles for cytochrome P 450 enzymes involved in steroid synthesis is high dependent on the stereochem. of the entire mol., whereas binding to drug metabolizing enzymes is a less selective process.
- 48Fleseriu, M.; Castinetti, F. Updates on the Role of Adrenal Steroidogenesis Inhibitors in Cushing’s Syndrome: A Focus on Novel Therapies. Pituitary 2016, 19 (6), 643– 653, DOI: 10.1007/s11102-016-0742-1Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtVKgurY%253D&md5=dd348c530b84890449477e66b23099e0Updates on the role of adrenal steroidogenesis inhibitors in Cushing's syndrome: a focus on novel therapiesFleseriu, Maria; Castinetti, FredericPituitary (2016), 19 (6), 643-653CODEN: PITUF9; ISSN:1386-341X. (Springer)A review. Purpose: Endogenous Cushing's syndrome (CS) is a rare disease that results from exposure to high levels of cortisol; Cushing's disease (CD) is the most frequent form of CS. Patients with CS suffer from a variety of comorbidities that increase the risk of mortality. Surgical resection of the disease-causing lesion is generally the first-line treatment of CS. However, some patients may not be eligible for surgery due to comorbidities, and approx. 25% of patients, esp. those with CD, have recurrent disease. For these patients, adrenal steroidogenesis inhibitors may control cortisol elevation and subsequent symptomatol. CS is rare overall, and clin. studies of adrenal steroidogenesis inhibitors are often small and, in many cases, data are limited regarding the efficacy and safety of these treatments. Our aim was to better characterize the profiles of efficacy and safety of currently available adrenal steroidogenesis inhibitors, including drugs currently in development. Methods: We performed a systematic review of the literature regarding adrenal steroidogenesis inhibitors, focusing on novel drugs. Results: Currently available adrenal steroidogenesis inhibitors, including ketoconazole, metyrapone, etomidate, and mitotane, have variable efficacy and significant side effects, and none are approved by the US Food and Drug Administration for CS. Therefore, there is a clear need for novel, prospectively studied agents that have greater efficacy and a low rate of adverse side effects. Efficacy and safety data of current and emerging adrenal steroidogenesis inhibitors, including osilodrostat (LCI699) and levoketoconazole (COR-003), show promising results that will have to be confirmed in larger-scale phase 3 studies (currently ongoing). Conclusions: The management of CS, and particularly CD, remains challenging. Adrenal steroidogenesis inhibitors can be of major interest to control the hypercortisolism at any time point, either before or after surgery, as discussed in this review.
- 49Brixius-Anderko, S.; Scott, E. E. Structure of Human Cortisol-Producing Cytochrome P450 11B1 Bound to the Breast Cancer Drug Fadrozole Provides Insights for Drug Design. J. Biol. Chem. 2019, 294 (2), 453– 460, DOI: 10.1074/jbc.RA118.006214Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs1Cjurg%253D&md5=d8b75ec9b273e63a2ff26bfbcf4b60efStructure of human cortisol-producing cytochrome P450 11B1 bound to the breast cancer drug fadrozole provides insights for drug designBrixius-Anderko, Simone; Scott, Emily E.Journal of Biological Chemistry (2019), 294 (2), 453-460CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Human cytochrome P 450 11B1 (CYP11B1) is responsible for the final step generating the steroid hormone cortisol, which controls stress and immune responses and glucose homeostasis. CYP11B1 is a promising drug target to manage Cushing's disease, a disorder arising from excessive cortisol prodn. However, the design of selective inhibitors has been hampered because structural information for CYP11B1 is unavailable and the enzyme has high amino acid sequence identity (93%) to a closely related enzyme, the aldosterone-producing CYP11B2. Here the authors report the x-ray crystal structure of human CYP11B1 (at 2.1 Å resoln.) in complex with fadrozole, a racemic compd. normally used to treat breast cancer by inhibiting estrogen-producing CYP19A1. Comparison of fadrozole-bound CYP11B1 with fadrozole-bound CYP11B2 revealed that despite conservation of the active-site residues, the overall structures and active sites had structural rearrangements consistent with distinct protein functions and inhibition. Whereas fadrozole binds to both CYP11B enzymes by coordinating the heme iron, CYP11B2 binds to the R enantiomer of fadrozole, and CYP11B1 binds to the S enantiomer, each with distinct orientations and interactions. These results provide insights into the cross-reactivity of drugs across multiple steroidogenic cytochrome P 450 enzymes, provide a structural basis for understanding human steroidogenesis, and pave the way for the design of more selective inhibitors of each human CYP11B enzyme.
- 50Brixius-Anderko, S.; Scott, E. E. Aldosterone Synthase Structure With Cushing Disease Drug LCI699 Highlights Avenues for Selective CYP11B Drug Design. Hypertension 2021, 78 (3), 751– 759, DOI: 10.1161/HYPERTENSIONAHA.121.17615Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsl2jtLbP&md5=a6eabccb0cc2c351edf10c87841c0510Aldosterone Synthase Structure With Cushing Disease Drug LCI699 Highlights Avenues for Selective CYP11B Drug DesignBrixius-Anderko, Simone; Scott, Emily E.Hypertension (2021), 78 (3), 751-759CODEN: HPRTDN; ISSN:0194-911X. (Lippincott Williams & Wilkins)Primary aldosteronism, the major form of secondary hypertension, develops due to excess steroid hormone aldosterone produced by aldosterone synthase, also known as cytochrome P 450 11B2. CYP11B2 is 93% identical to cortisol-producing CYP11B1, which makes it difficult to design selective drugs. LCI699 (Osilodrostat, Isturisa) was initially developed as a CYP11B2 inhibitor but due to poor selectivity was recently repurposed as the first Food and Drug Administration-approved drug for CYP11B1-mediated Cushing disease. Thus, there is still no effective therapeutic option targeting CYP11B2 for primary aldosteronism. Using a structure/function approach, aspects of LCI699 interaction with CYP11B2 were examd. to facilitate the design of more selective inhibitors. LCI699 effectively binds and inhibits CYP11B2. To det. the structural basis for this interaction, X-ray crystallog. was used to solve the structure of CYP11B2 bound to LCI699. LCI699 binds in the active site with its imidazole nitrogen coordinating the heme iron. LCI699 binding was compared with that of its analog fadrozole to both CYP11B enzymes. Comparison with the CYP11B1 structure reveals distinct CYP11B active site architectures which can be exploited to directed drug design. Exploiting structural differences between the CYP11B enzymes will promote the design of therapeutics for the treatment of primary aldosteronism targeting CYP11B2 while reducing undesirable side effects due to off-target CYP11B1 inhibition.
- 51Strushkevich, N.; Gilep, A. A.; Shen, L.; Arrowsmith, C. H.; Edwards, A. M.; Usanov, S. A.; Park, H.-W. Structural Insights into Aldosterone Synthase Substrate Specificity and Targeted Inhibition. Mol. Endocrinol. 2013, 27 (2), 315– 324, DOI: 10.1210/me.2012-1287Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvFOgur8%253D&md5=808e61fa02a12b63809bf3787ba83318Structural insights into aldosterone synthase substrate specificity and targeted inhibitionStrushkevich, Natallia; Gilep, Andrei A.; Shen, Limin; Arrowsmith, Cheryl H.; Edwards, Aled M.; Usanov, Sergey A.; Park, Hee-WonMolecular Endocrinology (2013), 27 (2), 315-324CODEN: MOENEN; ISSN:0888-8809. (Endocrine Society)Aldosterone is a major mineralocorticoid hormone that plays a key role in the regulation of electrolyte balance and blood pressure. Excess aldosterone levels can arise from dysregulation of the renin-angiotensin-aldosterone system and are implicated in the pathogenesis of hypertension and heart failure. Aldosterone synthase (cytochrome P 450 11B2, CYP11B2) is the sole enzyme responsible for the prodn. of aldosterone in humans. Blocking of aldosterone synthesis by mediating aldosterone synthase activity is thus a recently emerging pharmacol. therapy for hypertension, yet a lack of structural information has limited this approach. Here, we present the crystal structures of human aldosterone synthase in complex with a substrate deoxycorticosterone and an inhibitor fadrozole. The structures reveal a hydrophobic cavity with specific features assocd. with corticosteroid recognition. The substrate binding mode, along with biochem. data, explains the high 11β-hydroxylase activity of aldosterone synthase toward both gluco- and mineralocorticoid formation. The low processivity of aldosterone synthase with a high extent of intermediates release might be one of the mechanisms of controlled aldosterone prodn. from deoxycorticosterone. Although the active site pocket is lined by identical residues between CYP11B isoforms, most of the divergent residues that confer addnl. 18-oxidase activity of aldosterone synthase are located in the I-helix (vicinity of the O2 activation path) and loops around the H-helix (affecting an egress channel closure required for retaining intermediates in the active site). This intrinsic flexibility is also reflected in isoform-selective inhibitor binding. Fadrozole binds to aldosterone synthase in the R-configuration, using part of the active site cavity pointing toward the egress channel. The structural organization of aldosterone synthase provides crit. insights into the mol. mechanism of catalysis and enables rational design of more specific antihypertensive agents.
- 52Pinzi, L.; Rastelli, G. Identification of Target Associations for Polypharmacology from Analysis of Crystallographic Ligands of the Protein Data Bank. J. Chem. Inf. Model. 2020, 60 (1), 372– 390, DOI: 10.1021/acs.jcim.9b00821Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitlSrtLrF&md5=ad3fd02ec88c96d99b3394a8118a455eIdentification of Target Associations for Polypharmacology from Analysis of Crystallographic Ligands of the Protein Data BankPinzi, Luca; Rastelli, GiulioJournal of Chemical Information and Modeling (2020), 60 (1), 372-390CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)The design of a chem. entity that potently and selectively binds to a biol. target of therapeutic relevance has dominated the scene of drug discovery so far. However, recent findings suggest that multi-target ligands may be endowed with superior efficacy and be less prone to drug resistance. The Protein Data Bank (PDB) provides exptl. validated structural information about targets and bound ligands. Therefore, it represents a valuable source of information to help identifying active sites, understanding pharmacophore requirements, designing novel ligands, and inferring structure-activity relationships. In this study, we performed a large-scale anal. of the PDB by integrating different ligand-based and structure-based approaches, with the aim of identifying promising target assocns. for polypharmacol. based on reported crystal structure information. First, the 2D and 3D similarity profiles of the crystallog. ligands were evaluated using different ligand-based methods. Then, activity data of pairs of similar ligands binding to different targets were inspected by comparing structural information with bioactivity annotations reported in the ChEMBL, BindingDB, BindingMOAD, and PDBbind databases. Afterward, extensive docking screenings of ligands in the identified cross-targets were made in order to validate and refine the ligand-based results. Finally, the therapeutic relevance of the identified target combinations for polypharmacol. was evaluated from comparison with information on therapeutic targets reported in the Therapeutic Target Database (TTD). The results led to the identification of several target assocns. with high therapeutic potential for polypharmacol.
- 53Hawkins, P. C. D.; Skillman, A. G.; Nicholls, A. Comparison of Shape-Matching and Docking as Virtual Screening Tools. J. Med. Chem. 2007, 50 (1), 74– 82, DOI: 10.1021/jm0603365Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xhtlansb%252FF&md5=6f97f5c0cc092b4e225f7c2656c1bcf6Comparison of Shape-Matching and Docking as Virtual Screening ToolsHawkins, Paul C. D.; Skillman, A. Geoffrey; Nicholls, AnthonyJournal of Medicinal Chemistry (2007), 50 (1), 74-82CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Ligand docking is a widely used approach in virtual screening. In recent years a large no. of publications have appeared in which docking tools are compared and evaluated for their effectiveness in virtual screening against a wide variety of protein targets. These studies have shown that the effectiveness of docking in virtual screening is highly variable due to a large no. of possible confounding factors. Another class of method that has shown promise in virtual screening is the shape-based, ligand-centric approach. Several direct comparisons of docking with the shape-based tool ROCS have been conducted using data sets from some of these recent docking publications. The results show that a shape-based, ligand-centric approach is more consistent than, and often superior to, the protein-centric approach taken by docking.
- 54Gaulton, A.; Bellis, L. J.; Bento, A. P.; Chambers, J.; Davies, M.; Hersey, A.; Light, Y.; McGlinchey, S.; Michalovich, D.; Al-Lazikani, B.; Overington, J. P. ChEMBL: A Large-Scale Bioactivity Database for Drug Discovery. Nucleic Acids Res. 2012, 40 (D1), D1100– 1107, DOI: 10.1093/nar/gkr777Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs12htbjN&md5=aedf7793e1ca54b6a4fa272ea3ef7d0eChEMBL: a large-scale bioactivity database for drug discoveryGaulton, Anna; Bellis, Louisa J.; Bento, A. Patricia; Chambers, Jon; Davies, Mark; Hersey, Anne; Light, Yvonne; McGlinchey, Shaun; Michalovich, David; Al-Lazikani, Bissan; Overington, John P.Nucleic Acids Research (2012), 40 (D1), D1100-D1107CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)ChEMBL is an Open Data database contg. binding, functional and ADMET information for a large no. of drug-like bioactive compds. These data are manually abstracted from the primary published literature on a regular basis, then further curated and standardized to maximize their quality and utility across a wide range of chem. biol. and drug-discovery research problems. Currently, the database contains 5.4 million bioactivity measurements for more than 1 million compds. and 5200 protein targets. Access is available through a web-based interface, data downloads and web services at: https://www.ebi.ac.uk/chembldb.
- 55Gaulton, A.; Hersey, A.; Nowotka, M.; Bento, A. P.; Chambers, J.; Mendez, D.; Mutowo, P.; Atkinson, F.; Bellis, L. J.; Cibrián-Uhalte, E.; Davies, M.; Dedman, N.; Karlsson, A.; Magariños, M. P.; Overington, J. P.; Papadatos, G.; Smit, I.; Leach, A. R. The ChEMBL Database in 2017. Nucleic Acids Res. 2017, 45 (D1), D945– D954, DOI: 10.1093/nar/gkw1074Google Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslWhurs%253D&md5=1dcbb641f8c2d11410663f97505a4911The ChEMBL database in 2017Gaulton, Anna; Hersey, Anne; Nowotka, Micha-l; Bento, A. Patricia; Chambers, Jon; Mendez, David; Mutowo, Prudence; Atkinson, Francis; Bellis, Louisa J.; Cibrian-Uhalte, Elena; Davies, Mark; Dedman, Nathan; Karlsson, Anneli; Magarinos, Maria Paula; Overington, John P.; Papadatos, George; Smit, Ines; Leach, Andrew R.Nucleic Acids Research (2017), 45 (D1), D945-D954CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)ChEMBL is an open large-scale bioactivity database (https://www.ebi.ac.uk/chembl), previously described in the 2012 and 2014 Nucleic Acids Research Database Issues. Since then, alongside the continued extn. of data from the medicinal chem. literature, new sources of bioactivity data have also been added to the database. These include: deposited data sets from neglected disease screening; crop protection data; drug metab. and disposition data and bioactivity data from patents. A no. of improvements and new features have also been incorporated. These include the annotation of assays and targets using ontologies, the inclusion of targets and indications for clin. candidates, addn. of metabolic pathways for drugs and calcn. of structural alerts. The ChEMBL data can be accessed via a web-interface, RDF distribution, data downloads and RESTful web-services.
- 56Furet, P.; Batzl, C.; Bhatnagar, A.; Francotte, E.; Rihs, G.; Lang, M. Aromatase Inhibitors: Synthesis, Biological Activity, and Binding Mode of Azole-Type Compounds. J. Med. Chem. 1993, 36 (10), 1393– 1400, DOI: 10.1021/jm00062a012Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3sXktVyksbk%253D&md5=02848c7362f056866b0bd533e63612dfAromatase inhibitors: synthesis, biological activity, and binding mode of azole-type compoundsFuret, P.; Batzl, C.; Bhatnagar, A.; Francotte, E.; Rihs, G.; Lang, M.Journal of Medicinal Chemistry (1993), 36 (10), 1393-400CODEN: JMCMAR; ISSN:0022-2623.The enantiomers of the potent nonsteroidal inhibitor of aromatase, fadrozole hydrochloride, I (R1 = 4-NCC6H4, R2 = H; R1 = H, R2 = 4-NCC6H4), have been sepd. and their abs. configuration detd. by x-ray crystallog. On the basis of a mol. modeling comparison of the active enantiomer I ((R1 = 4-NCC6H4, R2 = H) and one of the most potent steroidal inhibitors reported to date, (19R)-10-thiiranylestr-4-ene-3,17-dione, II, a model describing the relative binding modes of the azole-type and steroidal inhibitors of aromatase at the active site of the enzyme is proposed. It is suggested that the cyanophenyl moiety present in the most active azole inhibitors partially mimics the steroid backbone of the natural substrate for aromatase, androst-4-ene-3,17-dione. The synthesis and biol. testing of novel analogs of fadrozole used to define the accessible and nonaccessible vols. to ligands in the model of the active site of aromatase are reported. Thus, imidazole reacted with SOCl2 and 6-cyano-1-tetralone in CH2Cl2 to give imidazolylcyanotetralin III.
- 57Meyers, K.; Cogan, D. A.; Burke, J.; Arenas, R.; Balestra, M.; Brown, N. F.; Chen, Z.; Cerny, M. A.; Clifford, H. E.; Colombo, F.; Fader, L.; Frederick, K. S.; Guo, X.; Goldberg, D.; Hornberger, K. R.; Kugler, S.; Lord, J.; Marshall, D. R.; Moss, N.; Parmentier, J.-H.; Richman, J. R.; Schmenk, J.; Weldon, S. M.; Yu, M.; Zhang, M. Dihydrobenzisoxazole-4-One Compounds Are Novel Selective Inhibitors of Aldosterone Synthase (CYP11B2) with in Vivo Activity. Bioorg. Med. Chem. Lett. 2018, 28 (5), 979– 984, DOI: 10.1016/j.bmcl.2017.12.015Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhvF2gtLfN&md5=33ded87b78beeef4d6f695007c01a776Dihydrobenzisoxazole-4-one compounds are novel selective inhibitors of aldosterone synthase (CYP11B2) with in vivo activityMeyers, Kenneth; Cogan, Derek A.; Burke, Jennifer; Arenas, Raquel; Balestra, Michael; Brown, Nicholas F.; Cerny, Matthew A.; Clifford, Holly E.; Colombo, Federico; Fader, Lee; Frederick, Kosea S.; Guo, Xin; Hornberger, Keith R.; Kugler, Stanley; Lord, John; Marshall, Daniel R.; Moss, Neil; Richman, Jeremy R.; Schmenk, Jennifer; Weldon, Steven M.; Yu, Maolin; Zhang, Michael; Chen, ZhidongBioorganic & Medicinal Chemistry Letters (2018), 28 (5), 979-984CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)6,7-Dihydro-5H-2,1-benzisoxazol-4-one analogs are potent inhibitors of aldosterone synthase (CYP11B2) with selectivity over the highly homologous enzyme cortisol synthase (CYP11B1). These compds. are unique among inhibitors of CYP11B2 in their lack of a strong-heme binding group such as a pyridine or imidazole. Poor metabolic stability in hepatocyte incubations was found to proceed via a redn. of the isoxazole ring. While the enzyme responsible for the reductive metab. remains unknown, the rate of metab. could be attenuated by the addn. of polar functionality. The in vitro CYP11B2 potency and selectivity were confirmed in vivo in a cynomolgus monkey model by the inhibition of ACTH stimulated aldosterone prodn. without impacting plasma cortisol concns.
- 58McGann, M. FRED Pose Prediction and Virtual Screening Accuracy. J. Chem. Inf. Model. 2011, 51 (3), 578– 596, DOI: 10.1021/ci100436pGoogle Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhvFGqsbk%253D&md5=8f3ae5972583d0a89d50e35a4b01212eFRED Pose Prediction and Virtual Screening AccuracyMcGann, MarkJournal of Chemical Information and Modeling (2011), 51 (3), 578-596CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Results of a previous docking study are reanalyzed and extended to include results from the docking program FRED and a detailed statistical anal. of both structure reprodn. and virtual screening results. FRED is run both in a traditional docking mode and in a hybrid mode that makes use of 16 the structure of a bound ligand in addn. to the protein structure to screen mols. This anal. shows that most docking programs are effective overall but highly inconsistent, tending to do well on one system and poorly on the next. Comparing methods, the difference in mean performance on DUD is found to be statistically significant (95% confidence) 61% of the time when using a global enrichment metric (AUC). Early enrichment metrics are found to have relatively poor statistical power, with 0.5% early enrichment only able to distinguish methods to 95% confidence 14% of the time.
- 59Ghosh, D.; Griswold, J.; Erman, M.; Pangborn, W. Structural Basis for Androgen Specificity and Oestrogen Synthesis in Human Aromatase. Nature 2009, 457 (7226), 219– 223, DOI: 10.1038/nature07614Google Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXitlSitA%253D%253D&md5=7f224e20c34cf5be770bb1c8b33c6e8cStructural basis for androgen specificity and estrogen synthesis in human aromataseGhosh, Debashis; Griswold, Jennifer; Erman, Mary; Pangborn, WalterNature (London, United Kingdom) (2009), 457 (7226), 219-223CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Aromatase cytochrome P 450 is the only enzyme in vertebrates known to catalyze the biosynthesis of all estrogens from androgens. Aromatase inhibitors therefore constitute a frontline therapy for estrogen-dependent breast cancer. In a three-step process, each step requiring 1 mol of O2, 1 mol of NADPH, and coupling with its redox partner cytochrome P 450 reductase, aromatase converts androstenedione, testosterone and 16α-hydroxytestosterone to estrone, 17β-estradiol and 17β,16α-estriol, resp. The first two steps are C19-Me hydroxylation steps, and the third involves the aromatization of the steroid A-ring, unique to aromatase. Whereas most P450s are not highly substrate selective, it is the hallmark androgenic specificity that sets aromatase apart. The structure of this enzyme of the endoplasmic reticulum membrane has remained unknown for decades, hindering elucidation of the biochem. mechanism. Here we present the crystal structure of human placental aromatase, the only natural mammalian, full-length P 450 and P 450 in hormone biosynthetic pathways to be crystd. so far. Unlike the active sites of many microsomal P450s that metabolize drugs and xenobiotics, aromatase has an androgen-specific cleft that binds the androstenedione mol. snugly. Hydrophobic and polar residues exquisitely complement the steroid backbone. The locations of catalytically important residues shed light on the reaction mechanism. The relative juxtaposition of the hydrophobic amino-terminal region and the opening to the catalytic cleft shows why membrane anchoring is necessary for the lipophilic substrates to gain access to the active site. The mol. basis for the enzyme's androgenic specificity and unique catalytic mechanism can be used for developing next-generation aromatase inhibitors.
- 60Yin, L.; Hu, Q.; Hartmann, R. W. Tetrahydropyrroloquinolinone Type Dual Inhibitors of Aromatase/Aldosterone Synthase as a Novel Strategy for Breast Cancer Patients with Elevated Cardiovascular Risks. J. Med. Chem. 2013, 56 (2), 460– 470, DOI: 10.1021/jm301408tGoogle Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVanug%253D%253D&md5=71a8d28d9b81108446073c259a48b498Tetrahydropyrroloquinolinone Type Dual Inhibitors of Aromatase/Aldosterone Synthase as a Novel Strategy for Breast Cancer Patients with Elevated Cardiovascular RisksYin, Lina; Hu, Qingzhong; Hartmann, Rolf W.Journal of Medicinal Chemistry (2013), 56 (2), 460-470CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The application of aromatase inhibitors to postmenopausal breast cancer patients increases the risk of cardiovascular diseases (CVD), which is believed to be caused by the abnormally high concns. of aldosterone as a consequence of the estrogen deficiency. Dual inhibitors of aromatase (CYP19) and aldosterone synthase (CYP11B2) are therefore proposed as a novel strategy for the adjuvant therapy to reduce the CVD risk for these patients. By combining decisive structural features of CYP11B2 and CYP19 inhibitors into a common template, a series of pyridinylmethyl substituted 1,2,5,6-tetrahydropyrrolo[3,2,1-ij]quinolin-4-ones were designed and synthesized. Interestingly, the substituents on the methylene bridge showed strong influences on the inhibitory activities leading to opposite effects, i.e., a given substituent showed an increase in inhibition of one enzyme, while it led to a decrease for the other enzyme. The compromise of this conflict led to compds. 3j, 3k, 3n, and 3p as potent and selective dual inhibitors of CYP19 and CYP11B2, esp. compd. 3p (I), which exhibited IC50 values of 32 and 41 nM for CYP19 and CYP11B2, resp., and a high selectivity toward CYP17 and CYP11B1. This compd. is considered as a candidate for further evaluation in vivo.
- 61Meredith, E. L.; Ksander, G.; Monovich, L. G.; Papillon, J. P. N.; Liu, Q.; Miranda, K.; Morris, P.; Rao, C.; Burgis, R.; Capparelli, M.; Hu, Q.-Y.; Singh, A.; Rigel, D. F.; Jeng, A. Y.; Beil, M.; Fu, F.; Hu, C.-W.; LaSala, D. Discovery and in Vivo Evaluation of Potent Dual CYP11B2 (Aldosterone Synthase) and CYP11B1 Inhibitors. ACS Med. Chem. Lett. 2013, 4 (12), 1203– 1207, DOI: 10.1021/ml400324cGoogle Scholar61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhs1Ckt7vP&md5=3f2301a71dfd1bf91a99ac68f5ca91ffDiscovery and in Vivo Evaluation of Potent Dual CYP11B2 (Aldosterone Synthase) and CYP11B1 InhibitorsMeredith, Erik L.; Ksander, Gary; Monovich, Lauren G.; Papillon, Julien P. N.; Liu, Qian; Miranda, Karl; Morris, Patrick; Rao, Chang; Burgis, Robin; Capparelli, Michael; Hu, Qi-Ying; Singh, Alok; Rigel, Dean F.; Jeng, Arco Y.; Beil, Michael; Fu, Fumin; Hu, Chii-Whei; LaSala, DanielACS Medicinal Chemistry Letters (2013), 4 (12), 1203-1207CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Aldosterone is a key signaling component of the renin-angiotensin-aldosterone system and as such has been shown to contribute to cardiovascular pathol. such as hypertension and heart failure. Aldosterone synthase (CYP11B2) is responsible for the final three steps of aldosterone synthesis and thus is a viable therapeutic target. A series of imidazole derived inhibitors, including clin. candidate I, have been identified through design and structure-activity relationship studies both in vitro and in vivo. Compd. I was also found to be a potent inhibitor of 11β-hydroxylase (CYP11B1), which is responsible for cortisol prodn. Inhibition of CYP11B1 is being evaluated in the clinic for potential treatment of hypercortisol diseases such as Cushing's syndrome.
- 62Bochevarov, A. D.; Harder, E.; Hughes, T. F.; Greenwood, J. R.; Braden, D. A.; Philipp, D. M.; Rinaldo, D.; Halls, M. D.; Zhang, J.; Friesner, R. A. Jaguar: A High-performance Quantum Chemistry Software Program with Strengths in Life and Materials Sciences. Int. J. Quantum Chem. 2013, 113 (18), 2110– 2142, DOI: 10.1002/qua.24481Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtVCqs7%252FP&md5=2fc119bb9e0501e55d7292f7735c24eaJaguar: A high-performance quantum chemistry software program with strengths in life and materials sciencesBochevarov, Art D.; Harder, Edward; Hughes, Thomas F.; Greenwood, Jeremy R.; Braden, Dale A.; Philipp, Dean M.; Rinaldo, David; Halls, Mathew D.; Zhang, Jing; Friesner, Richard A.International Journal of Quantum Chemistry (2013), 113 (18), 2110-2142CODEN: IJQCB2; ISSN:0020-7608. (John Wiley & Sons, Inc.)A review. Jaguar is an ab initio quantum chem. program that specializes in fast electronic structure predictions for mol. systems of medium and large size. Jaguar focuses on computational methods with reasonable computational scaling with the size of the system, such as d. functional theory (DFT) and local second-order Moller-Plesset perturbation theory. The favorable scaling of the methods and the high efficiency of the program make it possible to conduct routine computations involving several thousand MOs. This performance is achieved through a utilization of the pseudospectral approxn. and several levels of parallelization. The speed advantages are beneficial for applying Jaguar in biomol. computational modeling. Addnl., owing to its superior wave function guess for transition-metal-contg. systems, Jaguar finds applications in inorg. and bioinorg. chem. The emphasis on larger systems and transition metal elements paves the way toward developing Jaguar for its use in materials science modeling. The article describes the historical and new features of Jaguar, such as improved parallelization of many modules, innovations in ab initio pKa prediction, and new semiempirical corrections for nondynamic correlation errors in DFT. Jaguar applications in drug discovery, materials science, force field parameterization, and other areas of computational research are reviewed. Timing benchmarks and other results obtained from the most recent Jaguar code are provided. The article concludes with a discussion of challenges and directions for future development of the program. © 2013 Wiley Periodicals, Inc.
- 63Nelson, D. R.; Zeldin, D. C.; Hoffman, S. M.; Maltais, L. J.; Wain, H. M.; Nebert, D. W. Comparison of Cytochrome P450 (CYP) Genes from the Mouse and Human Genomes, Including Nomenclature Recommendations for Genes, Pseudogenes and Alternative-Splice Variants. Pharmacogenetics 2004, 14 (1), 1– 18, DOI: 10.1097/00008571-200401000-00001Google Scholar63https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjs1ags7c%253D&md5=10ee0bacd1f7213153412541bd2d9c95Comparison of cytochrome P450 (CYP) genes from the mouse and human genomes, including nomenclature recommendations for genes, pseudogenes and alternative-splice variantsNelson, David R.; Zeldin, Darryl C.; Hoffman, Susan M. G.; Maltais, Lois J.; Wain, Hester M.; Nebert, Daniel W.Pharmacogenetics (2004), 14 (1), 1-18CODEN: PHMCEE; ISSN:0960-314X. (Lippincott Williams & Wilkins)A review. Completion of both the mouse and human genome sequences in the private and public sectors has prompted comparison between the two species at multiple levels. This review summarizes the cytochrome P 450 (CYP) gene superfamily. For the first time, we have the ability to compare complete sets of CYP genes from two mammals. Use of the mouse as a model mammal, and as a surrogate for human biol., assumes reasonable similarity between the two. It is therefore of interest to catalog the genetic similarities and differences, and to clarify the limits of extrapolation from mouse to human. Data-mining methods have been used to find all the mouse and human CYP sequences; this includes 102 putatively functional genes and 88 pseudogenes in the mouse, and 57 putatively functional genes and 58 pseudogenes in the human. Comparison is made between all these genes, esp. the seven main CYP gene clusters. The seven CYP clusters are greatly expanded in the mouse with 72 functional genes vs. only 27 in the human, while many pseudogenes are present; presumably this phenomenon will be seen in many other gene superfamily clusters. Complete identification of all pseudogene sequences is likely to be clin. important, because some of these highly similar exons can interfere with PCR-based genotyping assays. A naming procedure for each of four categories of CYP pseudogenes is proposed, and we encourage various gene nomenclature committees to consider seriously the adoption and application of this pseudogene nomenclature system.
- 64Hare, S. H.; Harvey, A. J. mTOR Function and Therapeutic Targeting in Breast Cancer. Am. J. Cancer Res. 2017, 7 (3), 383– 404Google Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsVGgtLbO&md5=6a8141f05942c0277f1628279ca12594mTOR function and therapeutic targeting in breast cancerHare, Stephen H.; Harvey, Amanda J.American Journal of Cancer Research (2017), 7 (3), 383-404CODEN: AJCRFT; ISSN:2156-6976. (e-Century Publishing Corp.)The mTOR pathway was discovered in the late 1970s after the compd. and natural inhibitor of mTOR, rapamycin was isolated from the bacterium Streptomyces hygroscopicus. mTOR is serine/threonine kinase belonging to the phosphoinositide 3-kinase related kinase (PIKK) family. It forms two distinct complexes; mTORC1 and mTORC2. mTORC1 has a key role in regulating protein synthesis and autophagy while mTORC2 is involved in regulating kinases of the AGC family. mTOR signaling is often over active in multiple cancer types including breast cancer. This can involve mutations in mTOR itself but more commonly, in breast cancer, this is related to an increase in activity of ErbB family receptors or alterations and mutations of PI3K signaling. Rapamycin and its analogs (rapalogues) bind to the intercellular receptor FKBP12, and then predominantly inhibit mTORC1 signaling via an allosteric mechanism. Research has shown that inhibition of mTOR is a useful strategy in tackling cancers, with it acting to slow tumor growth and limit the spread of a cancer. Rapalogues have now made their way into the clinic with the rapalogue everolimus (RAD-001/Afinitor) approved for use in conjunction with exemestane, in post-menopausal breast cancer patients with advanced disease who are HER-2 neg. (normal expression), hormone receptor pos. and whose prior treatment with non-steroidal aromatase inhibitors has failed. Testing across multiple trials has proven that everolimus and other rapalogues are a viable way of treating certain types of cancer. However, rapalogues have shown some drawbacks both in research and clin., with their use often activating feedback pathways that counter their usefulness. As such, new types of inhibitors are being explored that work via different mechanisms, including inhibitors that are ATP competitive with mTOR and which act to perturb signaling from both mTOR complexes.
- 65Hadizadeh, F.; Shafiee, A.; Kazemi, R.; Mohammadi, M. Synthesis of 4-(1-Phenylmethyl-5-Imidazolyl)-1,4-Dihydropyridines as Calcium Channel Antagonists; NISCAIR-CSIR: India, 2002; vol 41B (12), pp 2679– 2682.Google ScholarThere is no corresponding record for this reference.
- 66Millet, R.; Domarkas, J.; Houssin, R.; Gilleron, P.; Goossens, J.-F.; Chavatte, P.; Logé, C.; Pommery, N.; Pommery, J.; Hénichart, J.-P. Potent and Selective Farnesyl Transferase Inhibitors. J. Med. Chem. 2004, 47 (27), 6812– 6820, DOI: 10.1021/jm030502yGoogle Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhtVKrs7%252FK&md5=bc8c296fc3c4c96c4da1d608d5e74502Potent and Selective Farnesyl Transferase InhibitorsMillet, Regis; Domarkas, Juozas; Houssin, Raymond; Gilleron, Pauline; Goossens, Jean-Francois; Chavatte, Philippe; Loge, Cedric; Pommery, Nicole; Pommery, Jean; Henichart, Jean-PierreJournal of Medicinal Chemistry (2004), 47 (27), 6812-6820CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)We recently described a novel series of CA1A2X peptidomimetics as farnesyl transferase inhibitors (FTIs). These compds. possess an N-(4-piperidinyl)benzamide scaffold mimicking A1A2 residue. Extensive exploration of structure-activity relationships revealed that replacement of cysteine by substituted benzylimidazoles provided nanomolar FTIs with in vitro activities (18e, IC50 = 4.60 nM on isolated enzyme, EC50 = 20.0 nM for growth inhibition on a tumor cell line). The mol. docking of 18e and 19e in the active site of the enzyme provided details of key interactions with the protein and showed that the methionine or phenylalanine residue fits into the aryl binding site.
- 67Berthold, M. R.; Cebron, N.; Dill, F.; Gabriel, T. R.; Kötter, T.; Meinl, T.; Ohl, P.; Sieb, C.; Thiel, K.; Wiswedel, B. KNIME: The Konstanz Information Miner. In Data Analysis, Machine Learning and Applications; Studies in Classification, Data Analysis, and Knowledge Organization; Preisach, C.; Burkhardt, H.; Schmidt-Thieme, L.; Decker, R., Eds.; Springer: Berlin, Heidelberg, 2008; pp 319– 326.Google ScholarThere is no corresponding record for this reference.
- 68Schrödinger Release 2018–3: LigPrep; Schrödinger, LLC: New York, NY, 2018.Google ScholarThere is no corresponding record for this reference.
- 69Hawkins, P. C. D.; Skillman, A. G.; Warren, G. L.; Ellingson, B. A.; Stahl, M. T. Conformer Generation with OMEGA: Algorithm and Validation Using High Quality Structures from the Protein Databank and Cambridge Structural Database. J. Chem. Inf. Model. 2010, 50 (4), 572– 584, DOI: 10.1021/ci100031xGoogle Scholar69https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjtlaisrY%253D&md5=fb87ecc9c51eddef63b41fffcd9babeeConformer Generation with OMEGA: Algorithm and Validation Using High Quality Structures from the Protein Databank and Cambridge Structural DatabaseHawkins, Paul C. D.; Skillman, A. Geoffrey; Warren, Gregory L.; Ellingson, Benjamin A.; Stahl, Matthew T.Journal of Chemical Information and Modeling (2010), 50 (4), 572-584CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Here, we present the algorithm and validation for OMEGA, a systematic, knowledge-based conformer generator. The algorithm consists of three phases: assembly of an initial 3D structure from a library of fragments; exhaustive enumeration of all rotatable torsions using values drawn from a knowledge-based list of angles, thereby generating a large set of conformations; and sampling of this set by geometric and energy criteria. Validation of conformer generators like OMEGA has often been undertaken by comparing computed conformer sets to exptl. mol. conformations from crystallog., usually from the Protein Databank (PDB). Such an approach is fraught with difficulty due to the systematic problems with small mol. structures in the PDB. Methods are presented to identify a diverse set of small mol. structures from cocomplexes in the PDB that has maximal reliability. A challenging set of 197 high quality, carefully selected ligand structures from well-solved models was obtained using these methods. This set will provide a sound basis for comparison and validation of conformer generators in the future. Validation results from this set are compared to the results using structures of a set of druglike mols. extd. from the Cambridge Structural Database (CSD). OMEGA is found to perform very well in reproducing the crystallog. conformations from both these data sets using two complementary metrics of success.
- 70Pinzi, L.; Caporuscio, F.; Rastelli, G. Selection of Protein Conformations for Structure-Based Polypharmacology Studies. Drug Discovery Today 2018, 23 (11), 1889– 1896, DOI: 10.1016/j.drudis.2018.08.007Google Scholar70https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsFeqt73F&md5=e0b96eeeb37b37fd4d5e5dea465d9a88Selection of protein conformations for structure-based polypharmacology studiesPinzi, Luca; Caporuscio, Fabiana; Rastelli, GiulioDrug Discovery Today (2018), 23 (11), 1889-1896CODEN: DDTOFS; ISSN:1359-6446. (Elsevier Ltd.)A review. Several drugs exert their therapeutic effect through the modulation of multiple targets. Structure-based approaches hold great promise for identifying compds. with the desired polypharmacol. profiles. These methods use knowledge of the protein binding sites to identify stereoelectronically complementary ligands. The selection of the most suitable protein conformations to be used in the design process is vital, esp. for multitarget drug design in which the same ligand has to be accommodated in multiple binding pockets. Herein, we focus on currently available techniques for the selection of the most suitable protein conformations for multitarget drug design, compare the potential advantages and limitations of each method, and comment on how their combination could help in polypharmacol. drug design.
- 71Pinzi, L.; Rastelli, G. Molecular Docking: Shifting Paradigms in Drug Discovery. Int. J. Mol. Sci. 2019, 20 (18), 4331, DOI: 10.3390/ijms20184331Google Scholar71https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVCns70%253D&md5=9ed9e680a89538b1ef1f11f6cd2b7e68Molecular docking: shifting paradigms in drug discoveryPinzi, Luca; Rastelli, GiulioInternational Journal of Molecular Sciences (2019), 20 (18), 4331CODEN: IJMCFK; ISSN:1422-0067. (MDPI AG)A review. Mol. docking is an established in silico structure-based method widely used in drug discovery. Docking enables the identification of novel compds. of therapeutic interest, predicting ligand-target interactions at a mol. level, or delineating structure-activity relationships (SAR), without knowing a priori the chem. structure of other target modulators. Although it was originally developed to help understanding the mechanisms of mol. recognition between small and large mols., uses and applications of docking in drug discovery have heavily changed over the last years. In this review, we describe how mol. docking was firstly applied to assist in drug discovery tasks. Then, we illustrate newer and emergent uses and applications of docking, including prediction of adverse effects, polypharmacol., drug repurposing, and target fishing and profiling, discussing also future applications and further potential of this technique when combined with emergent techniques, such as artificial intelligence.
- 72Madhavi Sastry, G.; Adzhigirey, M.; Day, T.; Annabhimoju, R.; Sherman, W. Protein and Ligand Preparation: Parameters, Protocols, and Influence on Virtual Screening Enrichments. J. Comput. Aided Mol. Des. 2013, 27 (3), 221– 234, DOI: 10.1007/s10822-013-9644-8Google Scholar72https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXmslalu7c%253D&md5=259a6d547ef3e1310e091fb50fe8de16Protein and ligand preparation: parameters, protocols, and influence on virtual screening enrichmentsMadhavi Sastry, G.; Adzhigirey, Matvey; Day, Tyler; Annabhimoju, Ramakrishna; Sherman, WoodyJournal of Computer-Aided Molecular Design (2013), 27 (3), 221-234CODEN: JCADEQ; ISSN:0920-654X. (Springer)Structure-based virtual screening plays an important role in drug discovery and complements other screening approaches. In general, protein crystal structures are prepd. prior to docking in order to add hydrogen atoms, optimize hydrogen bonds, remove at. clashes, and perform other operations that are not part of the x-ray crystal structure refinement process. In addn., ligands must be prepd. to create 3-dimensional geometries, assign proper bond orders, and generate accessible tautomer and ionization states prior to virtual screening. While the prerequisite for proper system prepn. is generally accepted in the field, an extensive study of the prepn. steps and their effect on virtual screening enrichments has not been performed. In this work, we systematically explore each of the steps involved in prepg. a system for virtual screening. We first explore a large no. of parameters using the Glide validation set of 36 crystal structures and 1,000 decoys. We then apply a subset of protocols to the DUD database. We show that database enrichment is improved with proper prepn. and that neglecting certain steps of the prepn. process produces a systematic degrdn. in enrichments, which can be large for some targets. We provide examples illustrating the structural changes introduced by the prepn. that impact database enrichment. While the work presented here was performed with the Protein Prepn. Wizard and Glide, the insights and guidance are expected to be generalizable to structure-based virtual screening with other docking methods.
Cited By
This article has not yet been cited by other publications.
Article Views
Altmetric
Citations
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 72 other publications.
- 1Nardin, S.; Mora, E.; Varughese, F. M.; D’Avanzo, F.; Vachanaram, A. R.; Rossi, V.; Saggia, C.; Rubinelli, S.; Gennari, A. Breast Cancer Survivorship, Quality of Life, and Late Toxicities. Front. Oncol. 2020, 10, 864, DOI: 10.3389/fonc.2020.008641https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB38nntlaqsw%253D%253D&md5=bface92499349059a608f9c51d724b5eBreast Cancer Survivorship, Quality of Life, and Late ToxicitiesNardin Simone; Mora Edoardo; Varughese Feba Mariam; Vachanaram Ajay Ram; Gennari Alessandra; Nardin Simone; Mora Edoardo; Varughese Feba Mariam; Vachanaram Ajay Ram; Gennari Alessandra; D'Avanzo Francesca; Rossi Valentina; Saggia Chiara; Rubinelli Sara; Gennari AlessandraFrontiers in oncology (2020), 10 (), 864 ISSN:2234-943X.Breast cancer is the most frequent cancer in women: in 2018, almost two million cases have been diagnosed all over the world and it represents the principal cause of death from a neoplastic disease in women. In the past years, breast cancer prognosis has significantly improved over time: currently 5-year survival rates are in the range of 90%, and 10-year survival is about 80%. This improvement has been mostly observed in western countries, due to high coverage and compliance with screening programs, leading to early diagnosis, i.e., when the disease is at a subclinical level, and to an improvement in tumor molecular characterization and innovative systemic treatments. Yet the identification of different biological breast cancer subtypes prompted the development of innovative targeted agents and improved treatment personalization. On the other hand, longer survival rates and increasing proportions of cured patients require dedicated strategies to manage long-term sequelae of breast cancer treatments, with particular attention to quality of life. This review analyzes the most important issues, potentially occurring with cancer treatments, concerning long-term sequelae and quality of life, to define a global approach to breast cancer survivorship.
- 2International Agency for Research on Cancer, Lyon, France. Global Cancer Observatory. https://gco.iarc.fr/.There is no corresponding record for this reference.
- 3Smolarz, B.; Nowak, A. Z.; Romanowicz, H. Breast Cancer─Epidemiology, Classification, Pathogenesis and Treatment (Review of Literature). Cancers 2022, 14 (10), 2569, DOI: 10.3390/cancers141025693https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhsV2ht7%252FF&md5=022928457eab2d2e1b9419b2b659bce9Breast Cancer-Epidemiology, Classification, Pathogenesis and Treatment (Review of Literature)Smolarz, Beata; Nowak, Anna Zadrozna; Romanowicz, HannaCancers (2022), 14 (10), 2569CODEN: CANCCT; ISSN:2072-6694. (MDPI AG)Simple Summary: Breast cancer is the most-commonly diagnosed malignant tumor in women in the world, as well as the first cause of death from malignant tumors. The incidence of breast cancer is constantly increasing in all regions of the world. For this reason, despite the progress in its detection and treatment, which translates into improved mortality rates, it seems necessary to look for new therapeutic methods, predictive and prognostic factors. The article presents a review of the literature on breast carcinoma - a disease affecting women in the world. Abstr.: Breast cancer is the most-commonly diagnosed malignant tumor in women in the world, as well as the first cause of death from malignant tumors. The incidence of breast cancer is constantly increasing in all regions of the world. For this reason, despite the progress in its detection and treatment, which translates into improved mortality rates, it seems necessary to look for new therapeutic methods, and predictive and prognostic factors. Treatment strategies vary depending on the mol. subtype. Breast cancer treatment is multidisciplinary; it includes approaches to locoregional therapy (surgery and radiation therapy) and systemic therapy. Systemic therapies include hormone therapy for hormone-pos. disease, chemotherapy, anti-HER2 therapy for HER2-pos. disease, and quite recently, immunotherapy. Triple neg. breast cancer is responsible for more than 15-20% of all breast cancers. It is of particular research interest as it presents a therapeutic challenge, mainly due to its low response to treatment and its highly invasive nature. Future therapeutic concepts for breast cancer aim to individualize therapy and de-escalate and escalate treatment based on cancer biol. and early response to therapy. The article presents a review of the literature on breast carcinoma-a disease affecting women in the world.
- 4Haque, Md. M.; Desai, K. V. Pathways to Endocrine Therapy Resistance in Breast Cancer. Front. Endocrinol. 2019, 10, 573, DOI: 10.3389/fendo.2019.005734https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MrmtlOgtw%253D%253D&md5=f90b9a06e6fb87d34ab346490bb105e5Pathways to Endocrine Therapy Resistance in Breast CancerHaque Md Moquitul; Desai Kartiki VFrontiers in endocrinology (2019), 10 (), 573 ISSN:1664-2392.Breast cancers with positive expression of Estrogen Receptor (ER+) are treated with anti-hormone/endocrine therapy which targets the activity of the receptor, the half-life of the receptor or the availability of estrogen. This has significantly decreased mortality in women with ER+ breast cancer, however, about 25-30% of treated women run the risk or recurrence due to either intrinsic or acquired resistance to endocrine therapies. While ER itself is a predictor of response to such therapies, there exists a need to find more biomarkers and novel targets to treat resistant tumors. In this review, we summarize the known mechanisms and describe the ability of genomics in unraveling rare mutations and gene rearrangements that may impact the development of resistance and therefore treatment of ER+ breast cancer in the near future.
- 5Sørlie, T.; Perou, C. M.; Tibshirani, R.; Aas, T.; Geisler, S.; Johnsen, H.; Hastie, T.; Eisen, M. B.; Van De Rijn, M.; Jeffrey, S. S.; Thorsen, T.; Quist, H.; Matese, J. C.; Brown, P. O.; Botstein, D.; Lønning, P. E.; Børresen-Dale, A.-L. Gene Expression Patterns of Breast Carcinomas Distinguish Tumor Subclasses with Clinical Implications. Proc. Natl. Acad. Sci. U. S. A. 2001, 98 (19), 10869– 10874, DOI: 10.1073/pnas.1913670985https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXntVGmsbo%253D&md5=5a54045e69bbc92a5294eb5e73934bd7Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implicationsSorlie, Therese; Perou, Charles M.; Tibshirani, Robert; Aas, Turid; Geisler, Stephanie; Johnsen, Hilde; Hastie, Trevor; Eisen, Michael B.; Van de Rijn, Matt; Jeffrey, Stefanie S.; Thorsen, Thor; Quist, Hanne; Matese, John C.; Brown, Patrick O.; Botstein, David; Lonning, Per Eystein; Borresen-Dale, Anne-LiseProceedings of the National Academy of Sciences of the United States of America (2001), 98 (19), 10869-10874CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The purpose of this study was to classify breast carcinomas based on variations in gene expression patterns derived from cDNA microarrays and to correlate tumor characteristics to clin. outcome. A total of 85 cDNA microarray expts. representing 78 cancers, three fibroadenomas, and four normal breast tissues were analyzed by hierarchical clustering. As reported previously, the cancers could be classified into a basal epithelial-like group, an ERBB2-overexpressing group and a normal breast-like group based on variations in gene expression. A novel finding was that the previously characterized luminal epithelial/estrogen receptor-pos. group could be divided into at least two subgroups, each with a distinctive expression profile. These subtypes proved to be reasonably robust by clustering using two different gene sets: first, a set of 456 cDNA clones previously selected to reflect intrinsic properties of the tumors and, second, a gene set that highly correlated with patient outcome. Survival analyses on a subcohort of patients with locally advanced breast cancer uniformly treated in a prospective study showed significantly different outcomes for the patients belonging to the various groups, including a poor prognosis for the basal-like subtype and a significant difference in outcome for the two estrogen receptor-pos. groups.
- 6Musgrove, E. A.; Sutherland, R. L. Biological Determinants of Endocrine Resistance in Breast Cancer. Nat. Rev. Cancer 2009, 9 (9), 631– 643, DOI: 10.1038/nrc27136https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtVegtrzL&md5=e57a6a082e0f41d26422350657201de5Biological determinants of endocrine resistance in breast cancerMusgrove, Elizabeth A.; Sutherland, Robert L.Nature Reviews Cancer (2009), 9 (9), 631-643CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)A review. Endocrine therapies targeting estrogen action (anti-estrogens, such as tamoxifen, and aromatase inhibitors) decrease mortality from breast cancer, but their efficacy is limited by intrinsic and acquired therapeutic resistance. Candidate mol. biomarkers and gene expression signatures of tamoxifen response emphasize the importance of deregulation of proliferation and survival signaling in endocrine resistance. However, definition of the specific genetic lesions and mol. processes that det. clin. endocrine resistance is incomplete. The development of large-scale computational and genetic approaches offers the promise of identifying the mediators of endocrine resistance that may be exploited as potential therapeutic targets and biomarkers of response in the clinic.
- 7Liu, C.-Y.; Wu, C.-Y.; Petrossian, K.; Huang, T.-T.; Tseng, L.-M.; Chen, S. Treatment for the Endocrine Resistant Breast Cancer: Current Options and Future Perspectives. J. Steroid Biochem. Mol. Biol. 2017, 172, 166– 175, DOI: 10.1016/j.jsbmb.2017.07.0017https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFGmtLjK&md5=93d1de1dcc2f66ae08f3f95887c38adaTreatment for the endocrine resistant breast cancer: Current options and future perspectivesLiu, Chun-Yu; Wu, Chia-Yun; Petrossian, Karineh; Huang, Tzu-Ting; Tseng, Ling-Ming; Chen, ShiuanJournal of Steroid Biochemistry and Molecular Biology (2017), 172 (), 166-175CODEN: JSBBEZ; ISSN:0960-0760. (Elsevier Ltd.)Endocrine resistance remains a challenge and an unmet need for managing hormone receptor-pos. breast cancer. The mechanisms of endocrine resistance are multifaceted and are likely to evolve over time following various single or combination therapies. The purpose of this review article is to provide general understanding of mol. basis of endocrine resistance of breast cancer and to offer comprehensive review on current treatment options and potential new treatment strategies for endocrine resistant breast cancers. Last but not the least, we discuss current challenges and future directions for management of endocrine resistant breast cancers.
- 8Hu, Q.; Yin, L.; Hartmann, R. W. Selective Dual Inhibitors of CYP19 and CYP11B2: Targeting Cardiovascular Diseases Hiding in the Shadow of Breast Cancer. J. Med. Chem. 2012, 55 (16), 7080– 7089, DOI: 10.1021/jm30046378https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtFClurjO&md5=67a25d86529e58c06f10a4baf5463542Selective Dual Inhibitors of CYP19 and CYP11B2: Targeting Cardiovascular Diseases Hiding in the Shadow of Breast CancerHu, Qingzhong; Yin, Lina; Hartmann, Rolf W.Journal of Medicinal Chemistry (2012), 55 (16), 7080-7089CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Postmenopausal women are at high risk for cardiovascular diseases because of the estrogen deficiency. As for postmenopausal breast cancer patients, this risk is even higher due to inhibition of estrogens biosyntheses in peripheral tissue by the aromatase (CYP19) inhibitors applied. Because estrogen deficiency results in significantly elevated aldosterone levels, which are a major cause of cardiovascular diseases, dual inhibition of CYP19 and CYP11B2 (aldosterone synthase) is a promising treatment for breast cancer and the coinstantaneous cardiovascular diseases. By combination of important structural features of known CYP19 and CYP11B2 inhibitors, we succeeded in obtaining compds. 3 and 5 as selective dual inhibitors with IC50 values around 50 and 20 nM toward CYP19 and CYP11B2, resp. These compds. showed also good selectivity toward CYP11B1 (selectivity factors (IC50 CYP11B1/IC50 CYP11B2) around 50) and CYP17 (no inhibition).
- 9Perez, E. A. Safety Profiles of Tamoxifen and the Aromatase Inhibitors in Adjuvant Therapy of Hormone-Responsive Early Breast Cancer. Ann. Oncol. 2007, 18, viii26– viii35, DOI: 10.1093/annonc/mdm2639https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2sjht1ahsQ%253D%253D&md5=4e51288fe254120a955a47510271d03bSafety profiles of tamoxifen and the aromatase inhibitors in adjuvant therapy of hormone-responsive early breast cancerPerez E AAnnals of oncology : official journal of the European Society for Medical Oncology / ESMO (2007), 18 Suppl 8 (), viii26-35 ISSN:.Adjuvant endocrine therapy plays an important role in the management of hormone-receptor-positive early breast cancer, and has increased life expectancy for millions of women. Many patients receive adjuvant treatment for at least 5 years following tumor resection, hence good long-term safety is important for endocrine agents to gain widespread acceptance. Tamoxifen has been used as adjuvant therapy for early breast cancer for many years, and safety data have been well documented, but a poor risk:benefit profile limits treatment duration to 5 years. Increased efficacy over tamoxifen and good tolerability have recently made the third-generation aromatase inhibitors (AIs) the first-choice agents for adjuvant endocrine therapy; however, it is currently not known whether AI therapy, like tamoxifen, will be limited to 5 years. Many side effects of endocrine therapy, such as hot flushes and mood disturbances, are related to estrogen deprivation and are common to tamoxifen and AIs, reflecting the mechanism of action of these drugs. In addition, tamoxifen has estrogenic effects that are beneficial in some tissues: tamoxifen lowers serum cholesterol levels and protects against bone loss and cardiovascular disease, but is also associated with potentially life-threatening side effects, such as endometrial cancer and thromboembolic disease. As AIs lack estrogenic activity, they are not associated with these serious adverse events. Clinical trials comparing AIs with tamoxifen in the adjuvant setting have shown that AIs are well tolerated and are associated with a lower incidence of gynecological symptoms and hot flushes than tamoxifen. However, AIs are associated with musculoskeletal side effects, such as arthralgia, myalgia and bone loss, but these events are preventable or manageable. The effects of AIs on lipid metabolism and the cardiovascular system are still debatable, but placebo-controlled trials provide no evidence to suggest that AIs adversely affect these systems. Furthermore, the AIs allow women to maintain a good quality of life, comparable with women receiving tamoxifen or placebo, and are a cost-effective therapeutic option. Ongoing trials will provide more information regarding the long-term effects of AI therapy and will provide comparative data on the efficacy and safety of the different AIs, thereby helping to determine the optimal treatment strategy for these highly effective and well-tolerated drugs.
- 10Chapman, J.-A. W.; Meng, D.; Shepherd, L.; Parulekar, W.; Ingle, J. N.; Muss, H. B.; Palmer, M.; Yu, C.; Goss, P. E. Competing Causes of Death From a Randomized Trial of Extended Adjuvant Endocrine Therapy for Breast Cancer. JNCI J. Natl. Cancer Inst. 2008, 100 (4), 252– 260, DOI: 10.1093/jnci/djn01410https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1c7isFSltA%253D%253D&md5=afc9fa23e8500381ad33cfabbd5783c8Competing causes of death from a randomized trial of extended adjuvant endocrine therapy for breast cancerChapman Judith-Anne W; Meng Daniel; Shepherd Lois; Parulekar Wendy; Ingle James N; Muss Hyman B; Palmer Michael; Yu Changhong; Goss Paul EJournal of the National Cancer Institute (2008), 100 (4), 252-60 ISSN:.BACKGROUND: Older women with early-stage breast cancer experience higher rates of non-breast cancer-related death. We examined factors associated with cause-specific death in a large cohort of breast cancer patients treated with extended adjuvant endocrine therapy. METHODS: In the MA.17 trial, conducted by the National Cancer Institute of Canada Clinical Trials Group, 5170 breast cancer patients (median age = 62 years; range = 32-94 years) who were disease free after approximately 5 years of adjuvant tamoxifen treatment were randomly assigned to treatment with letrozole (2583 women) or placebo (2587 women). The median follow-up was 3.9 years (range 0-7 years). We investigated the association of 11 baseline factors with the competing risks of death from breast cancer, other malignancies, and other causes. All statistical tests were two-sided likelihood ratio criterion tests. RESULTS: During follow-up, 256 deaths were reported (102 from breast cancer, 50 from other malignancies, 100 from other causes, and four from an unknown cause). Non-breast cancer deaths accounted for 60% of the 252 known deaths (72% for those > or = 70 years and 48% for those < 70 years). Two baseline factors were differentially associated with type of death: cardiovascular disease was associated with a statistically significant increased risk of death from other causes (P.002), and osteoporosis was associated with a statistically significant increased risk of death from other malignancies (P.05). An increased risk of breast cancer-specific death was associated with lymph node involvement (P < .001). Increased risk of death from all three causes was associated with older age (P < .001). CONCLUSIONS: Non-breast cancer-related deaths were more common than breast cancer-specific deaths in this cohort of 5-year breast cancer survivors, especially among older women.
- 11Wang, Y.; Wang, Q.; Zhao, Y.; Gong, D.; Wang, D.; Li, C.; Zhao, H. Protective Effects of Estrogen Against Reperfusion Arrhythmias Following Severe Myocardial Ischemia in Rats. Circ. J. 2010, 74 (4), 634– 643, DOI: 10.1253/circj.CJ-09-022311https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXls1Sksbk%253D&md5=82f084f57eb2caeb3d27f0caad1dbdfdProtective effects of estrogen against reperfusion arrhythmias following severe myocardial ischemia in ratsWang, Yan; Wang, Qi; Zhao, Yingnan; Gong, Dezheng; Wang, Dongmei; Li, Cong; Zhao, HenanCirculation Journal (2010), 74 (4), 634-643CODEN: CJIOBY; ISSN:1346-9843. (Japanese Circulation Society)Background: Female sex hormones may have protective effects against arrhythmias, including reperfusion arrhythmias (RAs), but the mechanisms are still not completely known. Methods and Results: Serial changes in rat hearts (rhythm, apoptosis and the its influencing factors; cardiac vinculin mRNA expression and connexin43 (Cx43) dephosphorylation) were examd. during periods of ischemia-reperfusion with and without estrogen treatment. After reperfusion, although the incidence of arrhythmias became higher in both the vehicle-group and estrogen-group, compared with the ischemia period, estrogen prevented reperfusion-induced upregulation of the incidence of arrhythmias, esp. ventricular premature beats (VPB) and ventricular tachycardia (VT). The duration of VT and fibrillation, and the no. of VPB and VT, were all significantly decreased in the estrogen-group. The expression of cardiac vinculin mRNA decreased significantly in the vehicle-group but not in the estrogen-group. Cx43 dephosphorylation and myocyte apoptosis increased in both groups, but the values for the estrogen-group were all markedly lower than those for the vehicle-group. A selective estrogen receptor (ER) β agonist prevented reperfusion-induced upregulation of the incidence of both VPB and VT significantly; a selective ERα agonist had no significant influence. Conclusions: Estrogen can protect the heart against RAs, at least in part, mediated through gap junctions. Upregulation of ERβ but not ERα mediated most of the estrogen-induced cardioprotection against RA.
- 12Beer, S.; Reincke, M.; Kral, M.; Callies, F.; Strömer, H.; Dienesch, C.; Steinhauer, S.; Ertl, G.; Allolio, B.; Neubauer, S. High-Dose 17β–Estradiol Treatment Prevents Development of Heart Failure Post–Myocardial Infarction in the Rat. Basic Res. Cardiol. 2007, 102 (1), 9– 18, DOI: 10.1007/s00395-006-0608-112https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhvFymu78%253D&md5=2cb6046ecf83dade6c6e81832925ccb9High-dose 17β-estradiol treatment prevents development of heart failure post-myocardial infarction in the ratBeer, Stephanie; Reincke, Martin; Kral, Maike; Callies, Frank; Stroemer, Hinrik; Dienesch, Charlotte; Steinhauer, Sonja; Ertl, Georg; Allolio, Bruno; Neubauer, StefanBasic Research in Cardiology (2007), 102 (1), 9-18CODEN: BRCAB7; ISSN:0300-8428. (Steinkopff Verlag)Prognosis of heart failure remains poor despite therapeutic advances, such as angiotensin converting enzyme inhibition or β-receptor blockade. Thus, more effective forms of treatment are urgently needed. Since estrogens have been shown to modulate migration and proliferation of cardiac fibroblasts and to modulate the expression of estrogen receptors of cardiomyocytes we examd. whether high-dose estrogen treatment can affect post-myocardial infarction left ventricular remodeling. Female rats were treated with 17β-estradiol (7.5 mg/90 d) or placebo for ten weeks, starting two weeks prior to exptl. myocardial infarction. Eight weeks after infarction, in vivo echocardiog. and hemodynamic measurements as well as isolated heart perfusion were performed. In vivo, chronic estrogen treatment almost completely prevented the development of all signs of heart failure that occur in untreated infarcted hearts, such as increased left ventricular diams. (dilatation), reduced fractional shortening (systolic dysfunction) or increased left ventricular end-diastolic pressure (diastolic dysfunction). In vitro, the right- (indicating structural dilatation) and downward (indicating left ventricular dysfunction) shift of left ventricular pressure-vol. curves occurring in untreated infarcted hearts was completely prevented by estrogen. High dose estradiol treatment prevented development of post-MI remodeling, as assessed by in vivo and in vitro parameters of LV dysfunction. Estrogen may hold the potential of becoming a new form of heart failure treatment. However, the mechanisms responsible for this striking and unexpected beneficial action of estrogen in heart failure remain to be elucidated.
- 13Gardner, J. D.; Murray, D. B.; Voloshenyuk, T. G.; Brower, G. L.; Bradley, J. M.; Janicki, J. S. Estrogen Attenuates Chronic Volume Overload Induced Structural and Functional Remodeling in Male Rat Hearts. Am. J. Physiol.-Heart Circ. Physiol. 2010, 298 (2), H497– H504, DOI: 10.1152/ajpheart.00336.2009There is no corresponding record for this reference.
- 14Donaldson, C.; Eder, S.; Baker, C.; Aronovitz, M. J.; Weiss, A. D.; Hall-Porter, M.; Wang, F.; Ackerman, A.; Karas, R. H.; Molkentin, J. D.; Patten, R. D. Estrogen Attenuates Left Ventricular and Cardiomyocyte Hypertrophy by an Estrogen Receptor–Dependent Pathway That Increases Calcineurin Degradation. Circ. Res. 2009, 104 (2), 265– 275, DOI: 10.1161/CIRCRESAHA.108.19039714https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXntFahsw%253D%253D&md5=c59457053ff82df0c1197e958017a27eEstrogen attenuates left ventricular and cardiomyocyte hypertrophy by an estrogen receptor-dependent pathway That Increases Calcineurin DegradationDonaldson, Cameron; Eder, Sarah; Baker, Corey; Aronovitz, Mark J.; Weiss, Alexandra Dabreo; Hall-Porter, Monica; Wang, Feng; Ackerman, Adam; Karas, Richard H.; Molkentin, Jeffery D.; Patten, Richard D.Circulation Research (2009), 104 (2), 265-275CODEN: CIRUAL; ISSN:0009-7330. (Lippincott Williams & Wilkins)Left ventricular (LV) hypertrophy commonly develops in response to chronic hypertension and is a significant risk factor for heart failure and death. The serine-threonine phosphatase calcineurin (Cn)A plays a crit. role in the development of pathol. hypertrophy. Previous exptl. studies in murine models show that estrogen limits pressure overload-induced hypertrophy; our purpose was to explore further the mechanisms underlying this estrogen effect. Wild-type, ovariectomized female mice were treated with placebo or 17β-estradiol (E2), followed by transverse aortic constriction (TAC), to induce pressure overload. At 2 wk, mice underwent physiol. evaluation, immediate tissue harvest, or dispersion of cardiomyocytes. E2 replacement limited TAC-induced LV and cardiomyocyte hypertrophy while attenuating deterioration in LV systolic function and contractility. These E2 effects were assocd. with reduced abundance of CnA. The primary downstream targets of CnA are the nuclear factor of activated T-cell (NFAT) family of transcription factors. In transgenic mice expressing a NFAT-activated promoter/luciferase reporter gene, E2 limited TAC-induced activation of NFAT. Moreover, the inhibitory effects of E2 on LV hypertrophy were absent in CnA knockout mice, supporting the notion that CnA is an important target of E2-mediated inhibition. In cultured rat cardiac myocytes, E2 inhibited agonist-induced hypertrophy while also decreasing CnA abundance and NFAT activation. Agonist stimulation also reduced CnA ubiquitination and degrdn. that was prevented by E2; all in vitro effects of estrogen were reversed by an estrogen receptor (ER) antagonist. These data support that E2 reduces pressure overload induced hypertrophy by an ER-dependent mechanism that increases CnA degrdn., unveiling a novel mechanism by which E2 and ERs regulate pathol. LV and cardiomyocyte growth.
- 15Arias-Loza, P.-A.; Muehlfelder, M.; Elmore, S. A.; Maronpot, R.; Hu, K.; Blode, H.; Hegele-Hartung, C.; Fritzemeier, K. H.; Ertl, G.; Pelzer, T. Differential Effects of 17β-Estradiol and of Synthetic Progestins on Aldosterone-Salt–Induced Kidney Disease. Toxicol. Pathol. 2009, 37 (7), 969– 982, DOI: 10.1177/019262330935047515https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXmvVKrsQ%253D%253D&md5=b57c3c86b1e1591cb504a0513f7c60f4Differential effects of 17β-estradiol and of synthetic progestins on aldosterone-salt-induced kidney diseaseArias-Loza, Paula-Anahi; Muehlfelder, Melanie; Elmore, Susan A.; Maronpot, Robert; Hu, Kai; Blode, Hartmut; Hegele-Hartung, Christa; Fritzemeier, Karl Heinrich; Ertl, Georg; Pelzer, TheoToxicologic Pathology (2009), 37 (7), 969-982CODEN: TOPADD; ISSN:0192-6233. (Sage Publications)Elevated mineralocorticoid levels and female sex hormones have been shown to confer opposing effects on renal injury, but their combined effects are still unknown. Identify the function of estrogens and of different synthetic progestins on aldosterone salt-mediated renal disease. The role of 17β-estradiol, medroxyprogesterone acetate (MPA), and drospirenone during renal injury was studied in Wistar rats subjected to uni-nephrectomy plus aldosterone salt treatment. Aldo-salt treatment of intact, ovariectomized, and estradiol-treated female rats resulted in remnant kidney hypertrophy without structural damage. Co-treatment with MPA, but not with drospirenone, increased kidney hypertrophy, fluid turnover, sodium retention, and potassium excretion. Medroxyprogesterone acetate also caused glomerular, vascular, tubular, and interstitial lesions that were accompanied by increased blood pressure and enhanced NADPH oxidase (p67phox) and sodium channel (α-ENaC) expression. Drospirenone, a progestin with anti-mineralocorticoid function, and spironolactone prevented kidney hypertrophy, hypertension, and sodium retention. Drospirenone and spironolactone also increased renal angiotensin II type 2 receptor expression and relieved aldosterone-induced suppression of serum angiotensin II levels. The choice of specific synthetic progestins has profound implications on the development of kidney injury and renal gene expression under conditions of elevated aldosterone serum levels and salt intake.
- 16Kwan, M. L.; Yao, S.; Laurent, C. A.; Roh, J. M.; Quesenberry, C. P.; Kushi, L. H.; Lo, J. C. Changes in Bone Mineral Density in Women with Breast Cancer Receiving Aromatase Inhibitor Therapy. Breast Cancer Res. Treat. 2018, 168 (2), 523– 530, DOI: 10.1007/s10549-017-4626-516https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhvF2gt7jJ&md5=8f674ba15febd9a0f96926c4942a75f3Changes in bone mineral density in women with breast cancer receiving aromatase inhibitor therapyKwan, Marilyn L.; Yao, Song; Laurent, Cecile A.; Roh, Janise M.; Quesenberry, Charles P., Jr.; Kushi, Lawrence H.; Lo, Joan C.Breast Cancer Research and Treatment (2018), 168 (2), 523-530CODEN: BCTRD6; ISSN:0167-6806. (Springer)Purpose: We assessed bone mineral d. (BMD) change with aromatase inhibitor (AI) treatment in a contemporary cohort of women with breast cancer treated in Kaiser Permanente Northern California. Methods: Percent and estd. annual percent changes in BMD at the total hip and lumbar spine were examd. in 676 women receiving AI therapy who had two serial BMD reports available (at least 1 yr apart) before and after AI initiation (N = 317) or during continued AI therapy (N = 359). BMD changes were examd. at the total hip and lumbar spine and compared by age and clin. subgroups. Results: Women experienced BMD declines after AI initiation or continued therapy, with median annual percent change - 1.2% (interquartile range, IQR - 2.4 to - 0.1%) at the hip and - 1.0% (IQR - 2.3 to 0.1%) at the spine after AI initiation, and - 1.1% (IQR - 2.4 to 0.1%) at the hip and - 0.9% (IQR - 2.4 to 0.5%) at the spine during continued therapy. Higher levels of bone loss were obsd. among younger (< 55 years) compared with older (≥ 75 years) women at the hip (- 1.6% vs. - 0.8%) and at the spine (- 1.5% vs. - 0.5%) after AI initiation, and at the hip (- 1.4% vs. - 1.2%) and at the spine (- 2.4% vs. - 0.001%) during continued therapy. Conclusions: Small but consistent declines in total hip and lumbar spine BMD were present in breast cancer patients following AI therapy initiation or continued AI therapy. Although the overall rates of osteoporosis were low, greater estd. levels of annual bone loss were evident among women < 55 years.
- 17Tian, W.; Wu, M.; Deng, Y. Comparison of Changes in the Lipid Profiles of Eastern Chinese Postmenopausal Women With Early-Stage Breast Cancer Treated With Different Aromatase Inhibitors: A Retrospective Study. Clin. Pharmacol. Drug Dev. 2018, 7 (8), 837– 843, DOI: 10.1002/cpdd.42017https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFGqurfJ&md5=4aeb5a224fb921e1a678a9f84efbccf5Comparison of Changes in the Lipid Profiles of Eastern Chinese Postmenopausal Women With Early-Stage Breast Cancer Treated With Different Aromatase Inhibitors: A Retrospective StudyTian, Wei; Wu, Miaowei; Deng, YongchuanClinical Pharmacology in Drug Development (2018), 7 (8), 837-843CODEN: CPDDAH; ISSN:2160-7648. (John Wiley & Sons, Inc.)Cardiovascular morbidity is closely assocd. with serum lipid level. We aimed to investigate the effects of different aromatase inhibitors, including letrozole, anastrozole, and exemestane, on the lipid profile of eastern Chinese breast cancer patients. We evaluated a retrospective cohort of eastern Chinese postmenopausal women with early-stage breast cancer who received aromatase inhibitors. A total of 116 postmenopausal women with early-stage breast cancer without prior cardiovascular disease were included. Lipid changes at 3, 6, 12, and 24 mo were compared across the endocrine therapy categories. Our data demonstrated that exemestane treatment significantly decreased triglyceride level compared with letrozole after 24 mo. However, the aromatase inhibitors had almost equiv. impacts on high-d. liportein cholesterol, low-d. lipoprotein cholesterol, and triglyceride after long-term aromatase inhibitor treatment. As a small-size retrospective study, our data do not support a judgment about whether one AI or another carries more or less risk in terms of lipid disorders in eastern Chinese breast cancer patients. The exact effects need further randomized, controlled trials to investigate.
- 18Castelli, W. P. Cardiovascular Disease in Women. Am. J. Obstet. Gynecol. 1988, 158 (6), 1553– 1560, DOI: 10.1016/0002-9378(88)90189-518https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaL1c3jvVKqtA%253D%253D&md5=7e37bdc4dfdc154b9e8f1ed6bfe3f223Cardiovascular disease in womenCastelli W PAmerican journal of obstetrics and gynecology (1988), 158 (6 Pt 2), 1553-60, 1566-7 ISSN:0002-9378.Cardiovascular disease is the leading cause of death in American women. Atherosclerotic diseases, primarily myocardial infarction and stroke, are important causes of morbidity and mortality among women. Contrary to popular belief, the actual number of myocardial infarctions in men and women is similar, although women develop cardiovascular disease an average of 10 years later than do men. Total serum cholesterol level is a major indicator of risk of coronary heart disease; for every 1% increase in the total serum cholesterol level, a 2% increase in incidence of coronary heart disease is found. A high level of low-density lipoproteins is an independent risk factor for coronary heart disease in both mean and women, but high triglyceride level is an independent risk factor only in women. Conversely, the higher the level of high-density lipoprotein, the lower the risk of coronary heart disease. To reduce the risk of coronary heart disease, target levels of total serum cholesterol must be lowered from 300 to 200 mg/dl. If the total cholesterol/high-density lipoprotein ratio is greater than or equal to 4.5 of if the low-density lipoprotein concentration is greater than 150 mg/dl, the patient is at high risk for coronary heart disease. Clinical trials that use diet or drugs to lower serum cholesterol levels have consistently shown a 2% reduction in the incidence of coronary heart disease for every 1% reduction in total serum cholesterol level.
- 19Fischer, M. Renin Angiotensin System and Gender Differences in the Cardiovascular System. Cardiovasc. Res. 2002, 53 (3), 672– 677, DOI: 10.1016/S0008-6363(01)00479-519https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XhtleqsLk%253D&md5=b8d7cee6b950772eff8fd78a49878084Renin angiotensin system and gender differences in the cardiovascular systemFischer, Marcus; Baessler, Andrea; Schunkert, HeribertCardiovascular Research (2002), 53 (3), 672-677CODEN: CVREAU; ISSN:0008-6363. (Elsevier Science B.V.)A review. To explain gender-related differences of the cardiovascular system, the renin-angiotensin system experienced intensive exploration. Indeed, the development of hypertension as well as the progression of coronary artery disease and heart failure have 2 factors in common: (1) display distinct gender specific characteristics and (2) are enhanced by the renin-angiotensin system. It is therefore interesting to note that data from exptl. animals, epidemiol. surveys, and clin. investigations suggest that the components of the circulating as well as tissue-based renin-angiotensin system are markedly affected by gender. However, the issue is complicated by counter-regulatory effects of estrogen on the system with the substrate, on one hand, and the processing enzymes as well as the chief receptor, on the other hand. In fact, angiotensinogen is up-regulated particularly by oral administration of estrogen, whereas renin, angiotensin-converting enzyme (ACE), and AT-1 receptor are down-regulated by the hormone. While under well-defined exptl. conditions the net effect of estrogen appears to result in suppression of the renin-angiotensin system, the clin. situation may be more complex. The judgment is further complicated by the difficulty in precisely measuring the activity of the system at the tissue level. Moreover, clin. relevant read-outs for the activity of the renin-angiotensin system may be regulated multifactorially or only indirectly affected by the system. Nevertheless, the undisputable, profound biochem. changes in the renin-angiotensin system related to the estrogen status allow speculation that such interaction explains some of the differences in the cardiovascular system of men and women.
- 20Roesch, D. M.; Tian, Y.; Zheng, W.; Shi, M.; Verbalis, J. G.; Sandberg, K. Estradiol Attenuates Angiotensin-Induced Aldosterone Secretion in Ovariectomized Rats. Endocrinology 2000, 141 (12), 4629– 4636, DOI: 10.1210/endo.141.12.782220https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXosVSqsbY%253D&md5=bd78f0ea5d5b9cf3f78dda265373702cEstradiol attenuates angiotensin-induced aldosterone secretion in ovariectomized ratsRoesch, Darren M.; Tian, Ying; Zheng, Wei; Shi, Min; Verbalis, Joseph G.; Sandberg, KathrynEndocrinology (2000), 141 (12), 4629-4636CODEN: ENDOAO; ISSN:0013-7227. (Endocrine Society)Estrogen replacement therapy significantly reduces the risk of cardiovascular disease in postmenopausal women. Previous studies indicate that estradiol (E2) decreases angiotensin II (AT) receptor d. in the adrenal and pituitary in NaCl-loaded rats. We used an in vivo model that eliminates the potentially confounding influence of ACTH to det. whether the E2-induced decrease in adrenal AT receptor expression affects aldosterone responses to angiotensin II (Ang II). Female rats were ovariectomized, treated with oil (OVX) or E2(OVX+E2; 10 μg, s.c.) for 14 days, and fed a NaCl-deficient diet for the last 7 days to maximize adrenal AT receptor expression and responsiveness. On days 12-14 rats were treated with dexamethasone (DEX; 25 μg, i.p., every 12 h) to suppress plasma ACTH. On day 14 aldosterone secretion was measured after a 30-min infusion of Ang II (330 ng/min). Ang II infusion increased the peak plasma aldosterone levels to a lesser degree in the OVX+E2 than in the OVX rats (OVX, 1870 pg/mL; OVX+E2, 1010 pg/mL). Ang II-induced ACTH and aldosterone secretion was also studied in rats that were not treated with DEX. In the absence of DEX, the peak plasma aldosterone response was also significantly decreased (OVX, 5360 pg/mL; OVX+E2, 2960 pg/mL). However, E2 also reduced the plasma ACTH response to Ang II (OVX, 220 pg/mL; OVX+E2, 160 pg/mL), suggesting that reduced pituitary ACTH responsiveness to Ang II contributes to the effect of E2 on Ang II-induced aldosterone secretion. Adrenal AT1 binding studies confirmed that E2 significantly reduces adrenal AT1 receptor expression in both the presence and absence of DEX in NaCl-deprived rats. These results indicate that E2-induced decreases in pituitary and adrenal AT1 receptor expression are assocd. with attenuated pituitary ACTH and adrenal aldosterone responses to Ang II and suggest that estrogen replacement therapy may modulate Ang II-stimulated aldosterone secretion as part of its well known cardioprotective actions.
- 21Chappell, M. C.; Gallagher, P. E.; Averill, D. B.; Ferrario, C. M.; Brosnihan, K. B. Estrogen or the AT1 Antagonist Olmesartan Reverses the Development of Profound Hypertension in the Congenic mRen2.Lewis Rat. Hypertension 2003, 42 (4), 781– 786, DOI: 10.1161/01.HYP.0000085210.66399.A321https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXosF2rtb4%253D&md5=7901d1ae6145064874ee2b2793ce3b7aEstrogen or the AT1 antagonist olmesartan reverses the development of profound hypertension in the congenic mRen2.Lewis ratChappell, Mark C.; Gallagher, Patricia E.; Averill, David B.; Ferrario, Carlos M.; Brosnihan, K. BridgetHypertension (2003), 42 (4, Pt. 2), 781-786CODEN: HPRTDN; ISSN:0194-911X. (Lippincott Williams & Wilkins)The influence of estrogen on the regulation of cardiovascular function remains a controversial and complex area of investigation. We assessed the effects of estrogen depletion in the congenic mRen(2).Lewis rat, established from the back-cross of the original (mRen2)-27 transgenic onto the Lewis inbred strain. Ovariectomy of heterozygous mRen(2).Lewis at 4 to 5 wk resulted in a progressive increase in blood pressure compared with the sham surgery congenics at weeks 6 to 11. At 11 wk, the ovariectomized mRen(2).Lewis (OVX) systolic blood pressure averaged 195±3.7 mm Hg vs. 141±4.0 mm Hg for sham. Plasma Angiotensin (Ang) II, serum ACE activity, plasma renin concn., as well as urinary excretion of Ang II, 8-isoprostane F2α, and endothelin-1 were elevated; however, renal mRNA levels of eNOS were suppressed after ovariectomy. Estrogen replacement reduced blood pressure below both the sham and OVX by 11 wk (125±2.9 mm Hg, n=7, P<0.01 vs. OVX and sham). Moreover, the AT1 receptor antagonist olmesartan (CS866; week 12 to 16) essentially normalized blood pressure to 113±5.4 mm Hg (n=6, P<0.01 vs. OVX and sham). The attenuation of the hypertension was still evident 7 wk after complete withdrawal of treatment (124±4.1 mm Hg at week 23). In summary, the OVX mRen.2.Lewis exhibited a rapid and sustained increase in blood pressure. Estrogen or olmesartan lowered pressure by a similar extent. We conclude that the ovary exerts considerable influence on the regulation of the blood pressure in the mRen2.Lewis strain, possibly by limiting activation of the renin-angiotensin system.
- 22Harrison-Bernard, L. M.; Schulman, I. H.; Raij, L. Postovariectomy Hypertension Is Linked to Increased Renal AT1 Receptor and Salt Sensitivity. Hypertension 2003, 42 (6), 1157– 1163, DOI: 10.1161/01.HYP.0000102180.13341.5022https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXpsVGis78%253D&md5=75c66cb3b3beeb4857adbb5aa243bd1dPostovariectomy hypertension is linked to increased renal AT1 receptor and salt sensitivityHarrison-Bernard, Lisa M.; Schulman, Ivonne Hernandez; Raij, LeopoldoHypertension (2003), 42 (6), 1157-1163CODEN: HPRTDN; ISSN:0194-911X. (Lippincott Williams & Wilkins)The functional balance between angiotensin II (Ang II) and nitric oxide (NO) plays a key role in modulating salt sensitivity. Estrogen has been shown to downregulate angiotensin type 1 (AT1) receptor expression and to increase the bioavailability of endothelium-derived NO, which decreases AT1 receptor expression. The present study tests the hypothesis that in the presence of genetic salt sensitivity, deficiency of endogenous estrogens after ovariectomy (OVX) fosters an upregulation of Ang II. Female Dahl salt-resistant (DR), Dahl salt-sensitive (DS), Wistar-Kyoto (WKY), and spontaneously hypertensive (SHR) rats underwent bilateral OVX or sham surgery (SHX) and were fed a normal salt diet (0.5% NaCl) for 14 wk. Systolic blood pressures were measured every 2 wk and were not significantly different between OVX and SHX for DR, WKY, and SHR groups. However, at the end of 14 wk of normal salt diet, hypertension developed in DS OVX but not SHX rats (160±3 vs. 136±3 mm Hg; P<0.05). Hypertension also developed in DS OVX rats pair-fed a normal salt diet (166±7 mm Hg). Development of hypertension in DS OVX rats was prevented by estrogen replacement (132±3 mm Hg), AT1 receptor blockade (119±3 mm Hg), or feeding a very low salt diet (0.1% NaCl; 129±4 mm Hg). Renal AT1 receptor protein expression was significantly elevated 2-fold in DS OVX relative to SHX rats and was prevented by estrogen replacement. These data strongly suggest that after OVX in salt-sensitive rats there is a lower threshold for the hypertensinogenic effect of salt that is linked to an activation of Ang II.
- 23Krishnamurthi, K.; Verbalis, J. G.; Zheng, W.; Wu, Z.; Clerch, L. B.; Sandberg, K. Estrogen Regulates Angiotensin AT1 Receptor Expression via Cytosolic Proteins That Bind to the 52 Leader Sequence of the Receptor mRNA. Endocrinology 1999, 140 (11), 5435– 5438, DOI: 10.1210/endo.140.11.724223https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXmvFemuro%253D&md5=553e5501aa97bd6969ba135d482c7ef2Estrogen regulates angiotensin AT1 receptor expression via cytosolic proteins that bind to the 5' leader sequence of the receptor mRNAKrishnamurthi, Kamakshi; Verbalis, Joseph G.; Zheng, Wei; Wu, Zheng; Clerch, Linda B.; Sandberg, KathrynEndocrinology (1999), 140 (11), 5435-5438CODEN: ENDOAO; ISSN:0013-7227. (Endocrine Society)Two of the most highly recognized factors implicated in the pathogenesis of hypertension, atherosclerosis, congestive heart failure and assocd. cardiovascular disease are the renin-angiotensin system (RAS) and estrogen. A major effect of estrogen results from its influence on the RAS. β-Estradiol (E2) replacement in ovariectomized (OVX) rats significantly decreased type 1 angiotensin (AT1) receptor expression in the pituitary and adrenal, whereas it significantly increased receptor expression in the uterus when compared to OVX controls. Addnl. evidence demonstrated an important influence of estrogen on a recently discovered post-transcriptional mechanism for regulating expression of the AT1 receptor. This mechanism consists of cytosolic RNA binding proteins (BPs) that recognize the 5' leader sequence (5'LS) of the receptor mRNA. The activities of these 5'LS BPs were modulated by estrogen in an inverse manner to AT1 receptor regulation. Moreover, in vitro translation assays in wheat germ lysates suggested that the 5'LS BPs inhibited AT1 receptor translation. Our data therefore indicate that hormonal regulation of AT1 receptors involves modulation of 5'LS BPs by estrogen. These findings may in part account for the obsd. protective effects of estrogen on cardiovascular disease.
- 24Ries, C.; Lucas, S.; Heim, R.; Birk, B.; Hartmann, R. W. Selective Aldosterone Synthase Inhibitors Reduce Aldosterone Formation in Vitro and in Vivo. J. Steroid Biochem. Mol. Biol. 2009, 116 (3–5), 121– 126, DOI: 10.1016/j.jsbmb.2009.04.01324https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXptVGmt70%253D&md5=a03f287e26dc3bfff893dd9f3e5d0541Selective aldosterone synthase inhibitors reduce aldosterone formation in vitro and in vivoRies, Christina; Lucas, Simon; Heim, Ralf; Birk, Barbara; Hartmann, Rolf W.Journal of Steroid Biochemistry and Molecular Biology (2009), 116 (3-5), 121-126CODEN: JSBBEZ; ISSN:0960-0760. (Elsevier Ltd.)Aldosterone plays a crucial role in salt and water homeostasis but in case of pathol. increased plasma aldosterone levels it is also involved in the development and the progression of severe cardiovascular diseases like heart failure and myocardial fibrosis. For the treatment of these diseases we propose inhibition of the aldosterone forming enzyme CYP11B2 as a new pharmacol. strategy. We recently developed in vitro highly potent and selective inhibitors of human CYP11B2, but the evidence of their in vivo activity is still missing. For this purpose, rat aldosterone synthase gene was cloned and expressed in V79MZ cells to establish a new screening assay for the identification of "rat-active" substances. Compds. from the class of heteroaryl substituted 3,4-dihydro-1H-quinolin-2-ones showed a moderate inhibitory effect (65% at 2 μM) on rat CYP11B2 in vitro. Furthermore, it diminished the conversion of deoxycorticosterone to aldosterone in rat adrenals and significantly reduced plasma aldosterone levels in vivo.
- 25Anighoro, A.; Bajorath, J.; Rastelli, G. Polypharmacology: Challenges and Opportunities in Drug Discovery. J. Med. Chem. 2014, 57 (19), 7874– 7887, DOI: 10.1021/jm500646325https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtVShsL3I&md5=5ab36c961a84462d90bc25130d90a281Polypharmacology: Challenges and Opportunities in Drug DiscoveryAnighoro, Andrew; Bajorath, Jurgen; Rastelli, GiulioJournal of Medicinal Chemistry (2014), 57 (19), 7874-7887CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. At present, the legendary magic bullet, i.e., a drug with high potency and selectivity toward a specific biol. target, shares the spotlight with an emerging and alternative polypharmacol. approach. Polypharmacol. suggests that more effective drugs can be developed by specifically modulating multiple targets. It is generally thought that complex diseases such as cancer and central nervous system diseases may require complex therapeutic approaches. In this respect, a drug that "hits" multiple sensitive nodes belonging to a network of interacting targets offers the potential for higher efficacy and may limit drawbacks generally arising from the use of a single-target drug or a combination of multiple drugs. In this review, we will compare advantages and disadvantages of multitarget vs. combination therapies, discuss potential drug promiscuity arising from off-target effects, comment on drug repurposing, and introduce approaches to the computational design of multitarget drugs.
- 26Pinzi, L.; Tinivella, A.; Gagliardelli, L.; Beneventano, D.; Rastelli, G. LigAdvisor: A Versatile and User-Friendly Web-Platform for Drug Design. Nucleic Acids Res. 2021, 49 (W1), W326– W335, DOI: 10.1093/nar/gkab38526https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvFWgtbrO&md5=eee342dee01ed98315cd33d2f25a288dLigAdvisor: a versatile and user-friendly web-platform for drug designPinzi, Luca; Tinivella, Annachiara; Gagliardelli, Luca; Beneventano, Domenico; Rastelli, GiulioNucleic Acids Research (2021), 49 (W1), W326-W335CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)Although several tools facilitating in silico drug design are available, their results are usually difficult to integrate with publicly available information or require further processing to be fully exploited. The rational design of multi-target ligands (polypharmacol.) and the repositioning of known drugs towards unmet therapeutic needs (drug repurposing) have raised increasing attention in drug discovery, although they usually require careful planning of tailored drug design strategies. Computational tools and data-driven approaches can help to reveal novel valuable opportunities in these contexts, as they enable to efficiently mine publicly available chem., biol., clin., and disease-related data. Based on these premises, we developed LigAdvisor, a data-driven webserver which integrates information reported in DrugBank, Protein Data Bank, UniProt, Clin. Trials and Therapeutic Target Database into an intuitive platform, to facilitate drug discovery tasks as drug repurposing, polypharmacol., target fishing and profiling. As designed, LigAdvisor enables easy integration of similarity estn. results with clin. data, thereby allowing a more efficient exploitation of information in different drug discovery contexts. Users can also develop customizable drug design tasks on their own mols., by means of ligand- and target-based search modes, and download their results.
- 27Ankley, G. T.; Kahl, M. D.; Jensen, K. M.; Hornung, M. W.; Korte, J. J.; Makynen, E. A.; Leino, R. L. Evaluation of the Aromatase Inhibitor Fadrozole in a Short-Term Reproduction Assay with the Fathead Minnow (Pimephales Promelas). Toxicol. Sci. 2002, 67 (1), 121– 130, DOI: 10.1093/toxsci/67.1.12127https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XjtlGmsLw%253D&md5=02e4e03e90dee1df37ed1bffe02e6188Evaluation of the aromatase inhibitor fadrozole in a short-term reproduction assay with the fathead minnow (Pimephales promelas)Ankley, Gerald T.; Kahl, Michael D.; Jensen, Kathleen M.; Hornung, Michael W.; Korte, Joseph J.; Makynen, Elizabeth A.; Leino, Richard L.Toxicological Sciences (2002), 67 (1), 121-130CODEN: TOSCF2; ISSN:1096-6080. (Oxford University Press)Cytochrome P 450 aromatase (CYP19) is a key enzyme in vertebrate steroidogenesis, catalyzing the conversion of C19 androgens to C18 estrogens such as β-estradiol (E2). The objective of this study was to assess effects of the CYP19 inhibitor fadrozole on fathead minnow (Pimephales promelas) reproductive endocrinol. and physiol. in a short-term reprodn. assay proposed for identifying specific classes of endocrine-disrupting chems. A concn.-dependent redn. in fecundity was obsd. in fish exposed for 21 days to water concns. of fadrozole ranging from 2 to 50 μg/l. Consistent with the expected mechanism of action, there was a significant inhibition of brain aromatase activity in both male and female fathead minnows exposed to fadrozole. In females, this inhibition was accompanied by a concn.-dependent decrease in plasma E2 and vitellogenin concns.; the latter observation is consistent with the fact that activation of the estrogen receptor by E2 initiates hepatic vitellogenin prodn. in oviparous vertebrates. Histol. assessment of ovaries from females exposed to fadrozole indicated a decrease in mature oocytes and an increase in preovulatory atretic follicles. Exposure of male fathead minnows to fadrozole significantly increased plasma concns. of the androgens testosterone (T) and 11-ketotestosterone (KT) and resulted in a marked accumulation of sperm in the tests. Results of this study indicate that the proposed fathead minnow assay should effectively identify test chems. as potential aromatase inhibitors, both in the context of their reproductive toxicity and the specific mechanism of action. These results also should be of utility in assessing the potential ecol. risk of CYP19 inhibitors, in particular in the context of relating alterations in subcellular indicators of endocrine function (changes in steroids, proteins) to adverse consequences in the whole organism.
- 28Browne, L. J.; Gude, C.; Rodriguez, H.; Steele, R. E.; Bhatnager, A. Fadrozole Hydrochloride: A Potent, Selective, Nonsteroidal Inhibitor of Aromatase for the Treatment of Estrogen-Dependent Disease. J. Med. Chem. 1991, 34 (2), 725– 736, DOI: 10.1021/jm00106a03828https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXhtVWrsLg%253D&md5=a154e38c3cdb14113a3fefbddf8781eeFadrozole hydrochloride: a potent, selective, nonsteroidal inhibitor of aromatase for the treatment of estrogen-dependent diseaseBrowne, L. J.; Gude, C.; Rodriguez, H.; Steele, R. E.; Bhatnager, A.Journal of Medicinal Chemistry (1991), 34 (2), 725-36CODEN: JMCMAR; ISSN:0022-2623.The prepn. of 5-phenyl-5,6,7,8-tetrahydroimidazopyridine derivs. I (R = cyano, Br, CH2OH, Me, CO2H, CO2Et) and their evaluation as inhibitors for aromatase and estrogen prodn. was described; the most potent aromatase inhibitor was (+)-I (R = cyano), i.e. fadrozole (CGS 16949A). The mol. structure-activity relationship was discussed.
- 29Ménard, J.; Pascoe, L. Can the Dextroenantiomer of the Aromatase Inhibitor Fadrozole Be Useful for Clinical Investigation of Aldosterone-Synthase Inhibition?. J. Hypertens. 2006, 24 (6), 993– 997, DOI: 10.1097/01.hjh.0000226183.98439.b329https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xktlartrk%253D&md5=e56a1f5ec586a4dfa515f6f995f731c9Can the dextroenantiomer of the aromatase inhibitor fadrozole be useful for clinical investigation of aldosterone-synthase inhibition?Menard, Joel; Pascoe, LeighJournal of Hypertension (2006), 24 (6), 993-997CODEN: JOHYD3; ISSN:0263-6352. (Lippincott Williams & Wilkins)A review. The beneficial effects of spironolactone, eplerenone, amiloride, and potassium in preventing cardiovascular damage in various exptl. models of salt-induced hypertension can be dissocd. from blood pressure effects, and have drawn attention to the direct genomic and non-genomic actions of aldosterone at the level of the vessels, the heart, and the kidneys. Exposure to endogenous aldosterone could be decreased by direct and specific aldosterone-synthase inhibition. FAD 286A, the dextroenantiomer of the aromatase inhibitor fadrozole, might be a 1st candidate to investigate in humans, the physiol. impact and therapeutic properties of aldosterone-synthase inhibition, esp. in various forms of primary aldosteronism.
- 30Smith, I. E.; Norton, A. Fadrozole and Letrozole in Advanced Breast Cancer: Clinical and Biochemical Effects. Breast Cancer Res. Treat. 1998, 49 (S1), S67– S71, DOI: 10.1023/A:100600502437730https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXnt1Sktbk%253D&md5=34ab654833d3201e0ce7602e3f51984dFadrozole and letrozole in advanced breast cancer: clinical and biochemical effectsSmith, Ian E.; Norton, AlisonBreast Cancer Research and Treatment (1998), 49 (Suppl. 1), S67-S71CODEN: BCTRD6; ISSN:0167-6806. (Kluwer Academic Publishers)A review with 18 refs.
- 31Lamberts, S. W. J.; Bruining, H. A.; Marzouk, H.; Zuiderwijk, J.; Uitterlinden, P.; Blijd, J. J.; Hackeng, W. H. L.; Jong, F. H. D. The New Aromatase Inhibitor CGS-16949A SuppressesAldosterone and Cortisol Production by Human Adrenal Cells in Vitro. J. Clin. Endocrinol. Metab. 1989, 69 (4), 896– 901, DOI: 10.1210/jcem-69-4-89631https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1MXlvVOhur4%253D&md5=9c9846ae461e8242a07d225a8b20ebf6The new aromatase inhibitor CGS-16949A suppresses aldosterone and cortisol production by human adrenal cells in vitroLamberts, S. W. J.; Bruining, H. A.; Marzouk, H.; Zuiderwijk, J.; Uitterlinden, P.; Blijd, J. J.; Hackeng, W. H. L.; De Jong, F. H.Journal of Clinical Endocrinology and Metabolism (1989), 69 (4), 896-901CODEN: JCEMAZ; ISSN:0021-972X.CGS-16949A is a new orally active nonsteroidal aromatase inhibitor which is more than 100-fold more potent than aminoglutethimide. This compd. is an imidazole deriv., and therefore, its possible effect on cytochrome P 450-dependent enzyme activities in the adrenal gland was evaluated. In vitro investigations with dispersed normal and hyperplastic human adrenocortical cells showed that CGS-16949A at 10-7-10-6M is a potent 11β-hydroxylase inhibitor, which inhibits ACTH-stimulated cortisol release to a similar extent as an equimolar concn. of metyrapone (IC50 for both compds., 10-7-5 × 10-7M). Etomidate was a more potent 11 β-hydroxylase inhibitor (IC50, ∼10-8M), while 10-7-10-6M ketoconazole caused (via 17α-hydroxylase inhibition) a similar inhibition of cortisol release as 10-7M CGS-16949A (IC50, 10-7-5 × 10-5M). The 11β-hydroxylase inhibition by CGS-16949A was accompanied by a dose-dependent increase in the release of precursor steroids by the adrenocortical cells in vitro, including deoxycortisol, 17-hydroxyprogesterone, and androstenedione. Aldosterone release was suppressed 50% by 10-8M CGS-16949A, while the IC50 for cortisol in the same cells was 10-7M. Aldosterone release by the dispersed adenoma cells obtained from a patient with primary aldosteronism was also suppressed by CGS-16949A. It is concluded that the new nonsteroidal aromatase inhibitor CGS 16949A is an inhibitor of 11β-hydroxylase which is equipotent to metyrapone. At present it is unclear whether the compd. at the dose that causes complete aromatase inhibition in vivo also affects stress-induced cortisol release in man. Also, CGS-16949A exerts a very potent inhibitory effect on normal aldosterone release (IC50, 10-9M) and on a tumorous aldosterone secretion. CGS-16949A might, therefore, be a drug that can be used in the treatment of primary hyperaldosteronism.
- 32Hu, Q.; Yin, L.; Hartmann, R. W. Aldosterone Synthase Inhibitors as Promising Treatments for Mineralocorticoid Dependent Cardiovascular and Renal Diseases: Miniperspective. J. Med. Chem. 2014, 57 (12), 5011– 5022, DOI: 10.1021/jm401430e32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXotVelsQ%253D%253D&md5=585f8d88a1df8d22382e6a58ab0283b0Aldosterone Synthase Inhibitors as Promising Treatments for Mineralocorticoid Dependent Cardiovascular and Renal DiseasesHu, Qingzhong; Yin, Lina; Hartmann, Rolf W.Journal of Medicinal Chemistry (2014), 57 (12), 5011-5022CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Besides the well-known roles of aldosterone as a mineralocorticoid in regulating homeostasis of electrolytes and vol., recent studies revealed that it is also a potent proinflammation factor inducing reactive oxygen species and up-regulating a panel of fibrosis related genes. Under pathol. circumstances, excessive aldosterone is involved in a lot of chronic diseases, including hypertension, cardiac fibrosis, congestive heart failure, ventricular remodeling, and diabetic nephropathy. Therefore, the inhibition of aldosterone synthase (CYP11B2), which is the pivotal enzyme in aldosterone biosynthesis, was proposed as a superior approach. Expected pharmacodynamic effects have been demonstrated in both animal models and clin. trials after the application of CYP11B2 inhibitors. The importance of selectivity over other steroidogenic CYP enzymes, in particular 11β-hydroxylase (CYP11B1), was also revealed. Recently, much more selective CYP11B2 inhibitors have been reported, which could be promising drug candidates for the treatment of aldosterone related diseases.
- 33Roumen, L.; Peeters, J. W.; Emmen, J. M. A.; Beugels, I. P. E.; Custers, E. M. G.; De Gooyer, M.; Plate, R.; Pieterse, K.; Hilbers, P. A. J.; Smits, J. F. M.; Vekemans, J. A. J.; Leysen, D.; Ottenheijm, H. C. J.; Janssen, H. M.; Hermans, J. J. R. Synthesis, Biological Evaluation, and Molecular Modeling of 1-Benzyl-1H-Imidazoles as Selective Inhibitors of Aldosterone Synthase (CYP11B2). J. Med. Chem. 2010, 53 (4), 1712– 1725, DOI: 10.1021/jm901356d33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtlCgtrY%253D&md5=95b05f5ad57464a12715c4cf15502b83Synthesis, Biological Evaluation, and Molecular Modeling of 1-Benzyl-1H-imidazoles as Selective Inhibitors of Aldosterone Synthase (CYP11B2)Roumen, Luc; Peeters, Joris W.; Emmen, Judith M. A.; Beugels, Ilona P. E.; Custers, Erica M. G.; de Gooyer, Marcel; Plate, Ralf; Pieterse, Koen; Hilbers, Peter A. J.; Smits, Jos F. M.; Vekemans, Jef A. J.; Leysen, Dirk; Ottenheijm, Harry C. J.; Janssen, Henk M.; Hermans, J. J. RobJournal of Medicinal Chemistry (2010), 53 (4), 1712-1725CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Reducing aldosterone action is beneficial in various major diseases such as heart failure. Currently, this is achieved with mineralocorticoid receptor antagonists, however, aldosterone synthase (CYP11B2) inhibitors may offer a promising alternative. In this study, three-dimensional modeling of CYP11B2 was used to model the binding modes of the natural substrate 18-hydroxycorticosterone and the recently published CYP11B2 inhibitor R-fadrozole (I) as a rational guide to design 44 structurally simple and achiral 1-benzyl-1H-imidazoles. Their syntheses, in vitro inhibitor potencies, and in silico docking are described. Some promising CYP11B2 inhibitors were identified, with the novel lead MOERAS115 (II) displaying an IC50 for CYP11B2 of 1.7 nM, and a CYP11B2 (vs. CYP11B1) selectivity of 16.5, comparable to R-fadrozole (IC50 for CYP11B2 6.0 nM, selectivity 19.8). Mol. docking of the inhibitors in the models enabled the generation of posthoc hypotheses on their binding modes, providing a valuable basis for future studies and further design of CYP11B2 inhibitors.
- 34Weldon, S. M.; Cerny, M. A.; Gueneva-Boucheva, K.; Cogan, D.; Guo, X.; Moss, N.; Parmentier, J.-H.; Richman, J. R.; Reinhart, G. A.; Brown, N. F. Selectivity of BI 689648, a Novel, Highly Selective Aldosterone Synthase Inhibitor: Comparison with FAD286 and LCI699 in Nonhuman Primates. J. Pharmacol. Exp. Ther. 2016, 359 (1), 142– 150, DOI: 10.1124/jpet.116.23646334https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvF2jtLrK&md5=584b931b62922f854df7b68b288d5d19Selectivity of BI 689648, a novel, highly selective aldosterone synthase inhibitor: comparison with FAD286 and LCI699 in nonhuman primatesWeldon, Steven M.; Cerny, Matthew A.; Gueneva-Boucheva, Kristina; Cogan, Derek; Guo, Xin; Moss, Neil; Parmentier, Jean-Hugues; Richman, Jeremy R.; Reinhart, Glenn A.; Brown, Nicholas F.Journal of Pharmacology and Experimental Therapeutics (2016), 359 (1), 142-150CODEN: JPETAB; ISSN:1521-0103. (American Society for Pharmacology and Experimental Therapeutics)The mineralocorticoid aldosterone is an important regulator of blood pressure, vol., and electrolyte balance. However, excess aldosterone can be deleterious as a driver of vascular remodeling and tissue fibrosis assocd. with cardiometabolic diseases. Aldosterone synthase (AS) inhibitors (ASI) attenuate the prodn. of aldosterone directly and have been proposed as an alternative to mineralocorticoid receptor antagonists for blocking the pathol. effects of excess aldosterone. Discovery of selective ASIs has been challenging because of the high sequence identity (93%) AS shares with cortisol synthase (CS), and the low identity of rodent AS compared with human (63%). Using cynomolgus (cyno) monkey-based models, we identified BI 689648 [6-(5-methoxymethyl-pyridin-3-yl)-3,4-dihydro-2H-[1,8]naphthyridine-1-carboxylic acid amide], a novel, highly selective ASI that exhibits an in vitro IC50 of 2 nMagainst AS and 300 nm against CS (150-fold selectivity) compared with the recently described ASIs FAD286 [4-(5,6,7,8-tetrahydroimidazo[1,5-a]pyridin-5-yl)benzonitrile] (3 nM AS; 90 nM CS; 40-fold) and LCI699 (4-[(5R)-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-5-yl]-3-fluorobenzonitrile) (10 nM AS; 80 nM CS; 8-fold). After oral administration in cyno monkeys, BI 689648 (5 mg/kg) exhibits a peak plasma concn. of ∼500 nM. For in vivo profiling we used an ACTH-challenge model in which BI 689648 was.20-fold more selective compared with FAD286 and LCI699. Because both FAD286 and LCI699 failed to provide adequate selectivity for CS when tested in patients, the desire for more selective mols. to test the ASI hypothesis remains high. Therefore, highly selective aldosterone synthase inhibitors such as BI 689648 represent an important step forward toward developing ASIs with greater potential for clin. success in cardiometabolic diseases.
- 35Matore, B. W.; Banjare, P.; Singh, J.; Roy, P. P. In Silico Selectivity Modeling of Pyridine and Pyrimidine Based CYP11B1 and CYP11B2 Inhibitors: A Case Study. J. Mol. Graph. Model. 2022, 116, 108238 DOI: 10.1016/j.jmgm.2022.10823835https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhsFKns7rN&md5=6df656ead0f6993c7c6f28f08cbe63caIn silico selectivity modeling of pyridine and pyrimidine based CYP11B1 and CYP11B2 inhibitors: A case studyMatore, Balaji Wamanrao; Banjare, Purusottam; Singh, Jagadish; Roy, Partha PratimJournal of Molecular Graphics & Modelling (2022), 116 (), 108238CODEN: JMGMFI; ISSN:1093-3263. (Elsevier Ltd.)Design of selective drug candidates for highly structural similar targets is a challenging task for researchers. The main objective of this study was to explore the selectivity modeling of pyridine and pyrimidine scaffold towards the highly homologous targets CYP11B1 and CYP11B2 enzymes by in silico (Mol. docking and QSAR) approaches. In this regard, a big dataset (n = 228) of CYP11B1 and CYP11B2 inhibitors were gathered and classified based on heterocyclic ring and the exhaustive anal. was carried out for pyridine and pyrimidinescaffolds. The LibDock algorithm was used to explore the binding pattern, screening, and identify the structural feature responsible for the selectivity of the ligands towards the studied targets. Finally, QSAR anal. was done to explore the correlation between various binding parameters and structural features responsible for the inhibitory activity and selectivity of the ligands in a quant. way. The docking and QSAR anal. clearly revealed and distinguished the importance of structural features, functional groups attached for CYP11B2 and CYP11B1 selectivity for pyridine and pyrimidine analogs. Addnl., the docking anal. highlighted the differentiating amino acids residues for selectivity for ligands for each of the enzymes. The results obtained from this research work will be helpful in designing the selective CYP11B1/CYP11B2 inhibitors.
- 36Baumann, M.; Baxendale, I. R. Sustainable Synthesis of Thioimidazoles via Carbohydrate-Based Multicomponent Reactions. Org. Lett. 2014, 16 (23), 6076– 6079, DOI: 10.1021/ol502845h36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvFOhsbbO&md5=997439ac86734e67b074f3b686748cefSustainable Synthesis of Thioimidazoles via Carbohydrate-Based Multi-component ReactionsBaumann, Marcus; Baxendale, Ian R.Organic Letters (2014), 16 (23), 6076-6079CODEN: ORLEF7; ISSN:1523-7052. (American Chemical Society)The synthesis of diversely functionalized thioimidazoles through a modern variant of the Marckwald reaction is presented. This new protocol utilizes unprotected carbohydrates as well as simple amine salts as sustainable and biorenewable starting materials. Importantly it was discovered that a bifurcated reaction pathway results from using aldoses and ketoses resp., yielding distinct reaction products in a highly selective manner.
- 37Baumann, M.; Baxendale, I. R. A Continuous-Flow Method for the Desulfurization of Substituted Thioimidazoles Applied to the Synthesis of Etomidate Derivatives: A Continuous-Flow Method for the Desulfurization of Substituted Thioimidazoles Applied to the Synthesis of Etomidate Derivatives. Eur. J. Org. Chem. 2017, 2017 (44), 6518– 6524, DOI: 10.1002/ejoc.20170083337https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtlCnsL3I&md5=3de7f2177ab7feedb828041bc89baae0A Continuous-Flow Method for the Desulfurization of Substituted Thioimidazoles Applied to the Synthesis of Etomidate DerivativesBaumann, Marcus; Baxendale, Ian R.European Journal of Organic Chemistry (2017), 2017 (44), 6518-6524CODEN: EJOCFK; ISSN:1099-0690. (Wiley-VCH Verlag GmbH & Co. KGaA)A simple yet robust flow set-up for the efficient desulfurization of a series of thioimidazoles is presented, which generates the corresponding imidazole derivs. I (R = C6H5, 4-BrC6H4, 2,6-F2C6H3,etc.) in high yields. The strategic choice of peristaltic over piston pumps allowed reliable delivery of the heterogeneous stream of the thioimidazole substrate into a T-piece where it reacted with NaNO2 in the presence of acetic acid. This approach enabled the controlled and safe formation of the reactive nitrosonium species without uncontrolled exposure to hazardous nitrous oxide byproducts as obsd. in related batch protocols. The value of the resulting imidazole products was further demonstrated by their conversion into various esters II (R = C6H5, 4-BrC6H4, 2,6-F2C6H3, etc.; R1 = Me, Et, i-Pr, CH2CF3) representing new derivs. of the known analgesic etomidate through an efficient one-pot Corey-Gilman-Ganem oxidn. procedure.
- 38Baumann, M.; Baxendale, I. R. A Continuous Flow Synthesis and Derivatization of 1,2,4-Thiadiazoles. Bioorg. Med. Chem. 2017, 25 (23), 6218– 6223, DOI: 10.1016/j.bmc.2017.01.02238https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXitVSqtbY%253D&md5=e6aa8834be2f90a733b7aebefa802e30A continuous flow synthesis and derivatization of 1,2,4-thiadiazolesBaumann, Marcus; Baxendale, Ian R.Bioorganic & Medicinal Chemistry (2017), 25 (23), 6218-6223CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)A continuous flow process is presented that enables the efficient synthesis and derivatization of 1,2,4-thiadiazole heterocycles. Special attention was given to the safe handling of the versatile yet hazardous trichloromethanesulfenyl chloride reagent including its in-line quenching in order to eliminate malodorous and corrosive byproducts. Based on this flow method gram quantities of 5-chloro-3-phenyl-1,2,4-thiadiazole was safely prepd. allowing for further elaboration of this valuable building block by reaction with different nitrogen-, sulfur- and oxygen-based nucleophiles. This synthetic approach was subsequently applied to generate a series of bromophenyl-5-chloro-1,2,4-thiadiazoles providing a valuable entry towards further structural diversification on this important heterocyclic scaffold.
- 39Maligres, P. E.; Waters, M. S.; Weissman, S. A.; McWilliams, J. C.; Lewis, S.; Cowen, J.; Reamer, R. A.; Volante, R. P.; Reider, P. J.; Askin, D. Preparation of a Clinically Investigated Ras Farnesyl Transferase Inhibitor. J. Heterocycl. Chem. 2003, 40 (2), 229– 241, DOI: 10.1002/jhet.557040020639https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXjsVamt7k%253D&md5=ed9c15111665cd350692e581b7ae8386Preparation of a clinically investigated ras farnesyl transferase inhibitorMaligres, Peter E.; Waters, Marjorie S.; Weissman, Steven A.; McWilliams, J. Christopher; Lewis, Stephanie; Cowen, Jennifer; Reamer, Robert A.; Volante, R. P.; Reider, Paul J.; Askin, DavidJournal of Heterocyclic Chemistry (2003), 40 (2), 229-241CODEN: JHTCAD; ISSN:0022-152X. (HeteroCorporation)The synthesis of ras farnesyl-protein transferase inhibitor I is described on a multi-kilogram scale. Retrosynthetic anal. reveals chloromethylimidazole II and a piperazinone III as viable precursors. The 1,5-disubstituted imidazole system was regioselectively assembled via an improved Marckwald imidazole synthesis. A new imidazole dethionation procedure was developed to convert the Marckwald mercaptoimidazole product to the desired imidazole. This methodol. was found to be tolerant of a variety of functional groups providing good to excellent yields of 1,5-disubstituted imidazoles. A new Mitsunobu cyclization strategy was developed to prep. the arylpiperazinone fragment III.
- 40Ashburn, T. T.; Thor, K. B. Drug Repositioning: Identifying and Developing New Uses for Existing Drugs. Nat. Rev. Drug Discovery 2004, 3 (8), 673– 683, DOI: 10.1038/nrd146840https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXmtVOhtL8%253D&md5=0145c02423ab81ace6e3b3d92585ccccDrug repositioning: identifying and developing new uses for existing drugsAshburn, Ted T.; Thor, Karl B.Nature Reviews Drug Discovery (2004), 3 (8), 673-683CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Biopharmaceutical companies attempting to increase productivity through novel discovery technologies have fallen short of achieving the desired results. Repositioning existing drugs for new indications could deliver the productivity increases that the industry needs while shifting the locus of prodn. to biotechnol. companies. More and more companies are scanning the existing pharmacopeia for repositioning candidates, and the no. of repositioning success stories is increasing.
- 41March-Vila, E.; Pinzi, L.; Sturm, N.; Tinivella, A.; Engkvist, O.; Chen, H.; Rastelli, G. On the Integration of In Silico Drug Design Methods for Drug Repurposing. Front. Pharmacol. 2017, 8, 298, DOI: 10.3389/fphar.2017.0029841https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFentbbP&md5=cf38db8a704a52cddbd6949caca49783On the integration of in silico drug design methods for drug repurposingMarch-Vila, Eric; Pinzi, Luca; Sturm, Noe; Tinivella, Annachiara; Engkvist, Ola; Chen, Hongming; Rastelli, GiulioFrontiers in Pharmacology (2017), 8 (), 298/1-298/7CODEN: FPRHAU; ISSN:1663-9812. (Frontiers Media S.A.)Drug repurposing has become an important branch of drug discovery. Several computational approaches that help to uncover new repurposing opportunities and aid the discovery process have been put forward, or adapted from previous applications. A no. of successful examples are now available. Overall, future developments will greatly benefit from integration of different methods, approaches and disciplines. Steps forward in this direction are expected to help to clarify, and therefore to rationally predict, new drug-target, target-disease, and ultimately drug-disease assocns.
- 42Wishart, D. S.; Feunang, Y. D.; Guo, A. C.; Lo, E. J.; Marcu, A.; Grant, J. R.; Sajed, T.; Johnson, D.; Li, C.; Sayeeda, Z.; Assempour, N.; Iynkkaran, I.; Liu, Y.; Maciejewski, A.; Gale, N.; Wilson, A.; Chin, L.; Cummings, R.; Le, D.; Pon, A.; Knox, C.; Wilson, M. DrugBank 5.0: A Major Update to the DrugBank Database for 2018. Nucleic Acids Res. 2018, 46 (D1), D1074– D1082, DOI: 10.1093/nar/gkx103742https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitlGisbvI&md5=986b28c7ea546596a26dd3ba38f05feeDrugBank 5.0: a major update to the DrugBank database for 2018Wishart, David S.; Feunang, Yannick D.; Guo, An C.; Lo, Elvis J.; Marcu, Ana; Grant, Jason R.; Sajed, Tanvir; Johnson, Daniel; Li, Carin; Sayeeda, Zinat; Assempour, Nazanin; Iynkkaran, Ithayavani; Liu, Yifeng; Maciejewski, Adam; Gale, Nicola; Wilson, Alex; Chin, Lucy; Cummings, Ryan; Le, Diana; Pon, Allison; Knox, Craig; Wilson, MichaelNucleic Acids Research (2018), 46 (D1), D1074-D1082CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)DrugBank is a web-enabled database contg. comprehensivemol. information about drugs, their mechanisms, their interactions and their targets. First described in 2006, Drug- Bank has continued to evolve over the past 12 years in response to marked improvements to web stds. and changing needs for drug research and development. This year's update, DrugBank 5.0, represents the most significant upgrade to the database in more than 10 years. In many cases, existing data content has grown by 100% or more over the last update. For instance, the total no. of investigational drugs in the database has grown by almost 300%, the no. of drug-drug interactions has grown by nearly 600% and the no. of SNP-assocd. drug effects has grown more than 3000%. Significant improvements have been made to the quantity, quality and consistency of drug indications, drug binding data as well as drug-drug and drug-food interactions. A great deal of brand new data have also been added to DrugBank 5.0. This includes information on the influence of hundreds of drugs on metabolite levels (pharmacometabolomics), gene expression levels (pharmacotranscriptomics) and protein expression levels (pharmacoproteomics). New data have also been added on the status of hundreds of newdrug clin. trials and existing drug repurposing trials. Many other important improvements in the content, interface and performance of the DrugBank website have been made and these should greatly enhance its ease of use, utility and potential applications in many areas of pharmacol. research, pharmaceutical science and drug education.
- 43Berman, H. M. The Protein Data Bank. Nucleic Acids Res. 2000, 28 (1), 235– 242, DOI: 10.1093/nar/28.1.23543https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXhvVKjt7w%253D&md5=227fb393f754be2be375ab727bfd05dcThe Protein Data BankBerman, Helen M.; Westbrook, John; Feng, Zukang; Gilliland, Gary; Bhat, T. N.; Weissig, Helge; Shindyalov, Ilya N.; Bourne, Philip E.Nucleic Acids Research (2000), 28 (1), 235-242CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)The Protein Data Bank (PDB; http://www.rcsb.org/pdb/)is the single worldwide archive of structural data of biol. macromols. This paper describes the goals of the PDB, the systems in place for data deposition and access, how to obtain further information, and near-term plans for the future development of the resource.
- 44Buzdar, A. U.; Robertson, J. F. R.; Eiermann, W.; Nabholtz, J.-M. An Overview of the Pharmacology and Pharmacokinetics of the Newer Generation Aromatase Inhibitors Anastrozole, Letrozole, and Exemestane. Cancer 2002, 95 (9), 2006– 2016, DOI: 10.1002/cncr.1090844https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XovFSqsL4%253D&md5=00257b7cac0222c6e0b067082949fa42An overview of the pharmacology and pharmacokinetics of the newer generation aromatase inhibitors anastrozole, letrozole, and exemestaneBuzdar, Aman U.; Robertson, John F. R.; Eiermann, Wolfgang; Nabholtz, Jean-MarcCancer (New York, NY, United States) (2002), 95 (9), 2006-2016CODEN: CANCAR; ISSN:0008-543X. (John Wiley & Sons, Inc.)A review. The newer generation, nonsteroidal aromatase inhibitors (AIs) anastrozole and letrozole have shown superior efficacy compared with tamoxifen as first-line treatments and compared with megestrol acetate as second-line therapy in postmenopausal women with advanced breast carcinoma. In an open-label, Phase II trial, it was reported that exemestane showed numerical superiority compared with tamoxifen for objective response and clin. benefit. Because these agents ultimately may be administered for periods of up to 5 yr in the adjuvant setting, it is of increasing importance to assess their tolerability and pharmacol. profiles. In the absence of data from direct clin. comparisons, the published literature was reviewed for the clin. pharmacol., pharmacokinetic characteristics, and selectivity profiles of anastrozole, letrozole, and exemestane. At clin. administered doses, the plasma half-lives of anastrozole (1 mg once daily), letrozole (2.5 mg once daily), and exemestane (25 mg once daily) were 41-48 h, 2-4 days, and 27 h, resp. The time to steady-state plasma levels was 7 days for both anastrozole and exemestane and 60 days for letrozole. Androgenic side effects have been reported only with exemestane. Anastrozole treatment had no impact on plasma lipid levels, whereas both letrozole and exemestane had an unfavorable effect on plasma lipid levels. In indirect comparisons, anastrozole showed the highest degree of selectivity compared with letrozole and exemestane in terms of a lack of effect on adrenosteroidogenesis. All three AIs demonstrated clin. efficacy over preexisting treatments. However, there were differences in terms of pharmacokinetics and effects on lipid levels and adrenosteroidogenesis. The long-term clin. significance of these differences remains to be elucidated.
- 45Grimm, S. W.; Dyroff, M. C. Inhibition of Human Drug Metabolizing Cytochromes P450 by Anastrozole, a Potent and Selective Inhibitor of Aromatase. Drug Metab. Dispos. Biol. Fate Chem. 1997, 25 (5), 598– 60245https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK2s3pslWgsg%253D%253D&md5=1c1907907372987ae13d9a99d84fbe5aInhibition of human drug metabolizing cytochromes P450 by anastrozole, a potent and selective inhibitor of aromataseGrimm S W; Dyroff M CDrug metabolism and disposition: the biological fate of chemicals (1997), 25 (5), 598-602 ISSN:0090-9556.Anastrozole (2,2'[5(1H-1,2,4-triazol-1-ylmethyl)-1,3-phenylene]- bis(2-methylproprionitrile)) is a potent third-generation inhibitor of aromatase, currently marketed as a treatment for postmenopausal women with advanced breast cancer. While its potency and selectivity for inhibition of estrogen synthesis has been established in both preclinical and clinical studies, this study used in vitro methods to examine the effects of anastrozole on several drug metabolizing CYP enzymes found in human liver. Human liver microsomes were co-incubated with anastrozole and probe substrates for CYP1A2 (phenacetin), CYP2A6 (coumarin), CYP2C9 (tolbutamide), CYP2D6 (dextromethorphan), and CYP3A (nifedipine). The formation of the CYP-specific metabolites following co-incubation with various anastrozole concentrations was determined to establish IC50 and Ki values for these enzymes. While anastrozole did not inhibit CYP2A6 and CYP2D6 activities at concentrations below 500 microM, this compound inhibited CYP1A2, CYP2C9, and CYP3A activities with Ki values of 8, 10, and 10 microM, respectively. Dixon plots used to determine the Ki values for the inhibition of CYP1A2 and CYP3A activities by anastrozole were biphasic, indicating additional lower affinity Ki values. Major metabolites of anastrozole did not retain the ability to inhibit the metabolism of nifedipine (CYP3A). The results of this study indicate that, although anastrozole can inhibit CYP1A2, 2C9, and 3A-mediated catalytic activities, this compound would not be expected to cause clinically significant interactions with other CYP-metabolized drugs at physiologically relevant concentrations achieved during therapy with Arimidex (Zeneca, Ltd., Macclesfield, UK) 1-mg.
- 46Gobbi, S.; Rampa, A.; Belluti, F.; Bisi, A. Nonsteroidal Aromatase Inhibitors for the Treatment of Breast Cancer: An Update. Anticancer Agents Med. Chem. 2014, 14 (1), 54– 65, DOI: 10.2174/1871520611313999030646https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3sfitFOltw%253D%253D&md5=5b359f6fa60cc11f7c96cad85e0f6bc6Nonsteroidal aromatase inhibitors for the treatment of breast cancer: an updateGobbi Silvia; Rampa Angela; Belluti Federica; Bisi AlessandraAnti-cancer agents in medicinal chemistry (2014), 14 (1), 54-65 ISSN:.Estrogens are known to be important in breast cancer growth in both pre- and post-menopausal women. Although circulating estrogen concentrations are very low after menopause, peripheral tissues generate sufficient concentrations to stimulate tumor growth. As aromatase is the rate-limiting enzyme in estrogen biosynthesis, inhibitors of this enzyme represent effective targeted therapy for breast cancer. Three compounds are now FDA approved and have become the first-choice endocrine drugs for postmenopausal breast cancer patients, since they are associated with superior activity and better general tolerability when compared with the estrogen receptor modulator tamoxifen. Nevertheless, some questions concerning the use of aromatase inhibitors for the treatment of breast cancer still need to be addressed, mainly related to their side-effects and the development of resistance, making research in this field still appealing. Many research groups, including our own, are still dealing with the search of new compounds that possess aromatase inhibitory properties. In this review an update of the latest achievements in the field of nonsteroidal aromatase inhibitors will be given.
- 47Rotstein, D. M.; Kertesz, D. J.; Walker, K. A. M.; Swinney, D. C. Stereoisomers of Ketoconazole: Preparation and Biological Activity. J. Med. Chem. 1992, 35 (15), 2818– 2825, DOI: 10.1021/jm00093a01547https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK38Xks12rsL0%253D&md5=ef996a2a278c12d93edc4edcdfffef22Stereoisomers of ketoconazole: preparation and biological activityRotstein, David M.; Kertesz, Denis J.; Walker, Keith A. M.; Swinney, David C.Journal of Medicinal Chemistry (1992), 35 (15), 2818-25CODEN: JMCMAR; ISSN:0022-2623.The four stereoisomers of the antifungal agent ketoconazole, (2S,4R)-, (2R,4R)-, (2R,4S)-, and (2S,4S)-(imidazolylmethyl)phenyl{[(acetylpiperazinyl)phenoxy]methyl}dioxolanes I, were prepd. and evaluated for their selectivity in inhibiting a no. of cytochrome P 450 enzymes. Thus, (bromomethyl)phenyldioxolanes II condensed with 4-(N-acetylpiperazino)phenol and imidazole to give I. II were prepd. by bromination of 2',4'-dichloroacetophenone followed by reaction with (S)- and (R)-solketal tosylate. Large differences in selectivity among the isomers were obsd. for inhibition of the cytochromes P 450 involved in steroid biosynthesis, whereas little difference was obsd. for inhibition of those assocd. with hepatic drug metab. The cis-(2S,4R) and trans-(2R,4R) isomers are equipotent in inhibiting corticoid 11β-hydroxylase and much more effective than their antipodes. Little selectivity was obsd. for inhibition of cholesterol side chain cleavage or xenobiotic hydroxylases. These data indicate that the affinity of azoles for cytochrome P 450 enzymes involved in steroid synthesis is high dependent on the stereochem. of the entire mol., whereas binding to drug metabolizing enzymes is a less selective process.
- 48Fleseriu, M.; Castinetti, F. Updates on the Role of Adrenal Steroidogenesis Inhibitors in Cushing’s Syndrome: A Focus on Novel Therapies. Pituitary 2016, 19 (6), 643– 653, DOI: 10.1007/s11102-016-0742-148https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtVKgurY%253D&md5=dd348c530b84890449477e66b23099e0Updates on the role of adrenal steroidogenesis inhibitors in Cushing's syndrome: a focus on novel therapiesFleseriu, Maria; Castinetti, FredericPituitary (2016), 19 (6), 643-653CODEN: PITUF9; ISSN:1386-341X. (Springer)A review. Purpose: Endogenous Cushing's syndrome (CS) is a rare disease that results from exposure to high levels of cortisol; Cushing's disease (CD) is the most frequent form of CS. Patients with CS suffer from a variety of comorbidities that increase the risk of mortality. Surgical resection of the disease-causing lesion is generally the first-line treatment of CS. However, some patients may not be eligible for surgery due to comorbidities, and approx. 25% of patients, esp. those with CD, have recurrent disease. For these patients, adrenal steroidogenesis inhibitors may control cortisol elevation and subsequent symptomatol. CS is rare overall, and clin. studies of adrenal steroidogenesis inhibitors are often small and, in many cases, data are limited regarding the efficacy and safety of these treatments. Our aim was to better characterize the profiles of efficacy and safety of currently available adrenal steroidogenesis inhibitors, including drugs currently in development. Methods: We performed a systematic review of the literature regarding adrenal steroidogenesis inhibitors, focusing on novel drugs. Results: Currently available adrenal steroidogenesis inhibitors, including ketoconazole, metyrapone, etomidate, and mitotane, have variable efficacy and significant side effects, and none are approved by the US Food and Drug Administration for CS. Therefore, there is a clear need for novel, prospectively studied agents that have greater efficacy and a low rate of adverse side effects. Efficacy and safety data of current and emerging adrenal steroidogenesis inhibitors, including osilodrostat (LCI699) and levoketoconazole (COR-003), show promising results that will have to be confirmed in larger-scale phase 3 studies (currently ongoing). Conclusions: The management of CS, and particularly CD, remains challenging. Adrenal steroidogenesis inhibitors can be of major interest to control the hypercortisolism at any time point, either before or after surgery, as discussed in this review.
- 49Brixius-Anderko, S.; Scott, E. E. Structure of Human Cortisol-Producing Cytochrome P450 11B1 Bound to the Breast Cancer Drug Fadrozole Provides Insights for Drug Design. J. Biol. Chem. 2019, 294 (2), 453– 460, DOI: 10.1074/jbc.RA118.00621449https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs1Cjurg%253D&md5=d8b75ec9b273e63a2ff26bfbcf4b60efStructure of human cortisol-producing cytochrome P450 11B1 bound to the breast cancer drug fadrozole provides insights for drug designBrixius-Anderko, Simone; Scott, Emily E.Journal of Biological Chemistry (2019), 294 (2), 453-460CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Human cytochrome P 450 11B1 (CYP11B1) is responsible for the final step generating the steroid hormone cortisol, which controls stress and immune responses and glucose homeostasis. CYP11B1 is a promising drug target to manage Cushing's disease, a disorder arising from excessive cortisol prodn. However, the design of selective inhibitors has been hampered because structural information for CYP11B1 is unavailable and the enzyme has high amino acid sequence identity (93%) to a closely related enzyme, the aldosterone-producing CYP11B2. Here the authors report the x-ray crystal structure of human CYP11B1 (at 2.1 Å resoln.) in complex with fadrozole, a racemic compd. normally used to treat breast cancer by inhibiting estrogen-producing CYP19A1. Comparison of fadrozole-bound CYP11B1 with fadrozole-bound CYP11B2 revealed that despite conservation of the active-site residues, the overall structures and active sites had structural rearrangements consistent with distinct protein functions and inhibition. Whereas fadrozole binds to both CYP11B enzymes by coordinating the heme iron, CYP11B2 binds to the R enantiomer of fadrozole, and CYP11B1 binds to the S enantiomer, each with distinct orientations and interactions. These results provide insights into the cross-reactivity of drugs across multiple steroidogenic cytochrome P 450 enzymes, provide a structural basis for understanding human steroidogenesis, and pave the way for the design of more selective inhibitors of each human CYP11B enzyme.
- 50Brixius-Anderko, S.; Scott, E. E. Aldosterone Synthase Structure With Cushing Disease Drug LCI699 Highlights Avenues for Selective CYP11B Drug Design. Hypertension 2021, 78 (3), 751– 759, DOI: 10.1161/HYPERTENSIONAHA.121.1761550https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsl2jtLbP&md5=a6eabccb0cc2c351edf10c87841c0510Aldosterone Synthase Structure With Cushing Disease Drug LCI699 Highlights Avenues for Selective CYP11B Drug DesignBrixius-Anderko, Simone; Scott, Emily E.Hypertension (2021), 78 (3), 751-759CODEN: HPRTDN; ISSN:0194-911X. (Lippincott Williams & Wilkins)Primary aldosteronism, the major form of secondary hypertension, develops due to excess steroid hormone aldosterone produced by aldosterone synthase, also known as cytochrome P 450 11B2. CYP11B2 is 93% identical to cortisol-producing CYP11B1, which makes it difficult to design selective drugs. LCI699 (Osilodrostat, Isturisa) was initially developed as a CYP11B2 inhibitor but due to poor selectivity was recently repurposed as the first Food and Drug Administration-approved drug for CYP11B1-mediated Cushing disease. Thus, there is still no effective therapeutic option targeting CYP11B2 for primary aldosteronism. Using a structure/function approach, aspects of LCI699 interaction with CYP11B2 were examd. to facilitate the design of more selective inhibitors. LCI699 effectively binds and inhibits CYP11B2. To det. the structural basis for this interaction, X-ray crystallog. was used to solve the structure of CYP11B2 bound to LCI699. LCI699 binds in the active site with its imidazole nitrogen coordinating the heme iron. LCI699 binding was compared with that of its analog fadrozole to both CYP11B enzymes. Comparison with the CYP11B1 structure reveals distinct CYP11B active site architectures which can be exploited to directed drug design. Exploiting structural differences between the CYP11B enzymes will promote the design of therapeutics for the treatment of primary aldosteronism targeting CYP11B2 while reducing undesirable side effects due to off-target CYP11B1 inhibition.
- 51Strushkevich, N.; Gilep, A. A.; Shen, L.; Arrowsmith, C. H.; Edwards, A. M.; Usanov, S. A.; Park, H.-W. Structural Insights into Aldosterone Synthase Substrate Specificity and Targeted Inhibition. Mol. Endocrinol. 2013, 27 (2), 315– 324, DOI: 10.1210/me.2012-128751https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvFOgur8%253D&md5=808e61fa02a12b63809bf3787ba83318Structural insights into aldosterone synthase substrate specificity and targeted inhibitionStrushkevich, Natallia; Gilep, Andrei A.; Shen, Limin; Arrowsmith, Cheryl H.; Edwards, Aled M.; Usanov, Sergey A.; Park, Hee-WonMolecular Endocrinology (2013), 27 (2), 315-324CODEN: MOENEN; ISSN:0888-8809. (Endocrine Society)Aldosterone is a major mineralocorticoid hormone that plays a key role in the regulation of electrolyte balance and blood pressure. Excess aldosterone levels can arise from dysregulation of the renin-angiotensin-aldosterone system and are implicated in the pathogenesis of hypertension and heart failure. Aldosterone synthase (cytochrome P 450 11B2, CYP11B2) is the sole enzyme responsible for the prodn. of aldosterone in humans. Blocking of aldosterone synthesis by mediating aldosterone synthase activity is thus a recently emerging pharmacol. therapy for hypertension, yet a lack of structural information has limited this approach. Here, we present the crystal structures of human aldosterone synthase in complex with a substrate deoxycorticosterone and an inhibitor fadrozole. The structures reveal a hydrophobic cavity with specific features assocd. with corticosteroid recognition. The substrate binding mode, along with biochem. data, explains the high 11β-hydroxylase activity of aldosterone synthase toward both gluco- and mineralocorticoid formation. The low processivity of aldosterone synthase with a high extent of intermediates release might be one of the mechanisms of controlled aldosterone prodn. from deoxycorticosterone. Although the active site pocket is lined by identical residues between CYP11B isoforms, most of the divergent residues that confer addnl. 18-oxidase activity of aldosterone synthase are located in the I-helix (vicinity of the O2 activation path) and loops around the H-helix (affecting an egress channel closure required for retaining intermediates in the active site). This intrinsic flexibility is also reflected in isoform-selective inhibitor binding. Fadrozole binds to aldosterone synthase in the R-configuration, using part of the active site cavity pointing toward the egress channel. The structural organization of aldosterone synthase provides crit. insights into the mol. mechanism of catalysis and enables rational design of more specific antihypertensive agents.
- 52Pinzi, L.; Rastelli, G. Identification of Target Associations for Polypharmacology from Analysis of Crystallographic Ligands of the Protein Data Bank. J. Chem. Inf. Model. 2020, 60 (1), 372– 390, DOI: 10.1021/acs.jcim.9b0082152https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitlSrtLrF&md5=ad3fd02ec88c96d99b3394a8118a455eIdentification of Target Associations for Polypharmacology from Analysis of Crystallographic Ligands of the Protein Data BankPinzi, Luca; Rastelli, GiulioJournal of Chemical Information and Modeling (2020), 60 (1), 372-390CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)The design of a chem. entity that potently and selectively binds to a biol. target of therapeutic relevance has dominated the scene of drug discovery so far. However, recent findings suggest that multi-target ligands may be endowed with superior efficacy and be less prone to drug resistance. The Protein Data Bank (PDB) provides exptl. validated structural information about targets and bound ligands. Therefore, it represents a valuable source of information to help identifying active sites, understanding pharmacophore requirements, designing novel ligands, and inferring structure-activity relationships. In this study, we performed a large-scale anal. of the PDB by integrating different ligand-based and structure-based approaches, with the aim of identifying promising target assocns. for polypharmacol. based on reported crystal structure information. First, the 2D and 3D similarity profiles of the crystallog. ligands were evaluated using different ligand-based methods. Then, activity data of pairs of similar ligands binding to different targets were inspected by comparing structural information with bioactivity annotations reported in the ChEMBL, BindingDB, BindingMOAD, and PDBbind databases. Afterward, extensive docking screenings of ligands in the identified cross-targets were made in order to validate and refine the ligand-based results. Finally, the therapeutic relevance of the identified target combinations for polypharmacol. was evaluated from comparison with information on therapeutic targets reported in the Therapeutic Target Database (TTD). The results led to the identification of several target assocns. with high therapeutic potential for polypharmacol.
- 53Hawkins, P. C. D.; Skillman, A. G.; Nicholls, A. Comparison of Shape-Matching and Docking as Virtual Screening Tools. J. Med. Chem. 2007, 50 (1), 74– 82, DOI: 10.1021/jm060336553https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xhtlansb%252FF&md5=6f97f5c0cc092b4e225f7c2656c1bcf6Comparison of Shape-Matching and Docking as Virtual Screening ToolsHawkins, Paul C. D.; Skillman, A. Geoffrey; Nicholls, AnthonyJournal of Medicinal Chemistry (2007), 50 (1), 74-82CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Ligand docking is a widely used approach in virtual screening. In recent years a large no. of publications have appeared in which docking tools are compared and evaluated for their effectiveness in virtual screening against a wide variety of protein targets. These studies have shown that the effectiveness of docking in virtual screening is highly variable due to a large no. of possible confounding factors. Another class of method that has shown promise in virtual screening is the shape-based, ligand-centric approach. Several direct comparisons of docking with the shape-based tool ROCS have been conducted using data sets from some of these recent docking publications. The results show that a shape-based, ligand-centric approach is more consistent than, and often superior to, the protein-centric approach taken by docking.
- 54Gaulton, A.; Bellis, L. J.; Bento, A. P.; Chambers, J.; Davies, M.; Hersey, A.; Light, Y.; McGlinchey, S.; Michalovich, D.; Al-Lazikani, B.; Overington, J. P. ChEMBL: A Large-Scale Bioactivity Database for Drug Discovery. Nucleic Acids Res. 2012, 40 (D1), D1100– 1107, DOI: 10.1093/nar/gkr77754https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs12htbjN&md5=aedf7793e1ca54b6a4fa272ea3ef7d0eChEMBL: a large-scale bioactivity database for drug discoveryGaulton, Anna; Bellis, Louisa J.; Bento, A. Patricia; Chambers, Jon; Davies, Mark; Hersey, Anne; Light, Yvonne; McGlinchey, Shaun; Michalovich, David; Al-Lazikani, Bissan; Overington, John P.Nucleic Acids Research (2012), 40 (D1), D1100-D1107CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)ChEMBL is an Open Data database contg. binding, functional and ADMET information for a large no. of drug-like bioactive compds. These data are manually abstracted from the primary published literature on a regular basis, then further curated and standardized to maximize their quality and utility across a wide range of chem. biol. and drug-discovery research problems. Currently, the database contains 5.4 million bioactivity measurements for more than 1 million compds. and 5200 protein targets. Access is available through a web-based interface, data downloads and web services at: https://www.ebi.ac.uk/chembldb.
- 55Gaulton, A.; Hersey, A.; Nowotka, M.; Bento, A. P.; Chambers, J.; Mendez, D.; Mutowo, P.; Atkinson, F.; Bellis, L. J.; Cibrián-Uhalte, E.; Davies, M.; Dedman, N.; Karlsson, A.; Magariños, M. P.; Overington, J. P.; Papadatos, G.; Smit, I.; Leach, A. R. The ChEMBL Database in 2017. Nucleic Acids Res. 2017, 45 (D1), D945– D954, DOI: 10.1093/nar/gkw107455https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslWhurs%253D&md5=1dcbb641f8c2d11410663f97505a4911The ChEMBL database in 2017Gaulton, Anna; Hersey, Anne; Nowotka, Micha-l; Bento, A. Patricia; Chambers, Jon; Mendez, David; Mutowo, Prudence; Atkinson, Francis; Bellis, Louisa J.; Cibrian-Uhalte, Elena; Davies, Mark; Dedman, Nathan; Karlsson, Anneli; Magarinos, Maria Paula; Overington, John P.; Papadatos, George; Smit, Ines; Leach, Andrew R.Nucleic Acids Research (2017), 45 (D1), D945-D954CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)ChEMBL is an open large-scale bioactivity database (https://www.ebi.ac.uk/chembl), previously described in the 2012 and 2014 Nucleic Acids Research Database Issues. Since then, alongside the continued extn. of data from the medicinal chem. literature, new sources of bioactivity data have also been added to the database. These include: deposited data sets from neglected disease screening; crop protection data; drug metab. and disposition data and bioactivity data from patents. A no. of improvements and new features have also been incorporated. These include the annotation of assays and targets using ontologies, the inclusion of targets and indications for clin. candidates, addn. of metabolic pathways for drugs and calcn. of structural alerts. The ChEMBL data can be accessed via a web-interface, RDF distribution, data downloads and RESTful web-services.
- 56Furet, P.; Batzl, C.; Bhatnagar, A.; Francotte, E.; Rihs, G.; Lang, M. Aromatase Inhibitors: Synthesis, Biological Activity, and Binding Mode of Azole-Type Compounds. J. Med. Chem. 1993, 36 (10), 1393– 1400, DOI: 10.1021/jm00062a01256https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3sXktVyksbk%253D&md5=02848c7362f056866b0bd533e63612dfAromatase inhibitors: synthesis, biological activity, and binding mode of azole-type compoundsFuret, P.; Batzl, C.; Bhatnagar, A.; Francotte, E.; Rihs, G.; Lang, M.Journal of Medicinal Chemistry (1993), 36 (10), 1393-400CODEN: JMCMAR; ISSN:0022-2623.The enantiomers of the potent nonsteroidal inhibitor of aromatase, fadrozole hydrochloride, I (R1 = 4-NCC6H4, R2 = H; R1 = H, R2 = 4-NCC6H4), have been sepd. and their abs. configuration detd. by x-ray crystallog. On the basis of a mol. modeling comparison of the active enantiomer I ((R1 = 4-NCC6H4, R2 = H) and one of the most potent steroidal inhibitors reported to date, (19R)-10-thiiranylestr-4-ene-3,17-dione, II, a model describing the relative binding modes of the azole-type and steroidal inhibitors of aromatase at the active site of the enzyme is proposed. It is suggested that the cyanophenyl moiety present in the most active azole inhibitors partially mimics the steroid backbone of the natural substrate for aromatase, androst-4-ene-3,17-dione. The synthesis and biol. testing of novel analogs of fadrozole used to define the accessible and nonaccessible vols. to ligands in the model of the active site of aromatase are reported. Thus, imidazole reacted with SOCl2 and 6-cyano-1-tetralone in CH2Cl2 to give imidazolylcyanotetralin III.
- 57Meyers, K.; Cogan, D. A.; Burke, J.; Arenas, R.; Balestra, M.; Brown, N. F.; Chen, Z.; Cerny, M. A.; Clifford, H. E.; Colombo, F.; Fader, L.; Frederick, K. S.; Guo, X.; Goldberg, D.; Hornberger, K. R.; Kugler, S.; Lord, J.; Marshall, D. R.; Moss, N.; Parmentier, J.-H.; Richman, J. R.; Schmenk, J.; Weldon, S. M.; Yu, M.; Zhang, M. Dihydrobenzisoxazole-4-One Compounds Are Novel Selective Inhibitors of Aldosterone Synthase (CYP11B2) with in Vivo Activity. Bioorg. Med. Chem. Lett. 2018, 28 (5), 979– 984, DOI: 10.1016/j.bmcl.2017.12.01557https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhvF2gtLfN&md5=33ded87b78beeef4d6f695007c01a776Dihydrobenzisoxazole-4-one compounds are novel selective inhibitors of aldosterone synthase (CYP11B2) with in vivo activityMeyers, Kenneth; Cogan, Derek A.; Burke, Jennifer; Arenas, Raquel; Balestra, Michael; Brown, Nicholas F.; Cerny, Matthew A.; Clifford, Holly E.; Colombo, Federico; Fader, Lee; Frederick, Kosea S.; Guo, Xin; Hornberger, Keith R.; Kugler, Stanley; Lord, John; Marshall, Daniel R.; Moss, Neil; Richman, Jeremy R.; Schmenk, Jennifer; Weldon, Steven M.; Yu, Maolin; Zhang, Michael; Chen, ZhidongBioorganic & Medicinal Chemistry Letters (2018), 28 (5), 979-984CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)6,7-Dihydro-5H-2,1-benzisoxazol-4-one analogs are potent inhibitors of aldosterone synthase (CYP11B2) with selectivity over the highly homologous enzyme cortisol synthase (CYP11B1). These compds. are unique among inhibitors of CYP11B2 in their lack of a strong-heme binding group such as a pyridine or imidazole. Poor metabolic stability in hepatocyte incubations was found to proceed via a redn. of the isoxazole ring. While the enzyme responsible for the reductive metab. remains unknown, the rate of metab. could be attenuated by the addn. of polar functionality. The in vitro CYP11B2 potency and selectivity were confirmed in vivo in a cynomolgus monkey model by the inhibition of ACTH stimulated aldosterone prodn. without impacting plasma cortisol concns.
- 58McGann, M. FRED Pose Prediction and Virtual Screening Accuracy. J. Chem. Inf. Model. 2011, 51 (3), 578– 596, DOI: 10.1021/ci100436p58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhvFGqsbk%253D&md5=8f3ae5972583d0a89d50e35a4b01212eFRED Pose Prediction and Virtual Screening AccuracyMcGann, MarkJournal of Chemical Information and Modeling (2011), 51 (3), 578-596CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Results of a previous docking study are reanalyzed and extended to include results from the docking program FRED and a detailed statistical anal. of both structure reprodn. and virtual screening results. FRED is run both in a traditional docking mode and in a hybrid mode that makes use of 16 the structure of a bound ligand in addn. to the protein structure to screen mols. This anal. shows that most docking programs are effective overall but highly inconsistent, tending to do well on one system and poorly on the next. Comparing methods, the difference in mean performance on DUD is found to be statistically significant (95% confidence) 61% of the time when using a global enrichment metric (AUC). Early enrichment metrics are found to have relatively poor statistical power, with 0.5% early enrichment only able to distinguish methods to 95% confidence 14% of the time.
- 59Ghosh, D.; Griswold, J.; Erman, M.; Pangborn, W. Structural Basis for Androgen Specificity and Oestrogen Synthesis in Human Aromatase. Nature 2009, 457 (7226), 219– 223, DOI: 10.1038/nature0761459https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXitlSitA%253D%253D&md5=7f224e20c34cf5be770bb1c8b33c6e8cStructural basis for androgen specificity and estrogen synthesis in human aromataseGhosh, Debashis; Griswold, Jennifer; Erman, Mary; Pangborn, WalterNature (London, United Kingdom) (2009), 457 (7226), 219-223CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Aromatase cytochrome P 450 is the only enzyme in vertebrates known to catalyze the biosynthesis of all estrogens from androgens. Aromatase inhibitors therefore constitute a frontline therapy for estrogen-dependent breast cancer. In a three-step process, each step requiring 1 mol of O2, 1 mol of NADPH, and coupling with its redox partner cytochrome P 450 reductase, aromatase converts androstenedione, testosterone and 16α-hydroxytestosterone to estrone, 17β-estradiol and 17β,16α-estriol, resp. The first two steps are C19-Me hydroxylation steps, and the third involves the aromatization of the steroid A-ring, unique to aromatase. Whereas most P450s are not highly substrate selective, it is the hallmark androgenic specificity that sets aromatase apart. The structure of this enzyme of the endoplasmic reticulum membrane has remained unknown for decades, hindering elucidation of the biochem. mechanism. Here we present the crystal structure of human placental aromatase, the only natural mammalian, full-length P 450 and P 450 in hormone biosynthetic pathways to be crystd. so far. Unlike the active sites of many microsomal P450s that metabolize drugs and xenobiotics, aromatase has an androgen-specific cleft that binds the androstenedione mol. snugly. Hydrophobic and polar residues exquisitely complement the steroid backbone. The locations of catalytically important residues shed light on the reaction mechanism. The relative juxtaposition of the hydrophobic amino-terminal region and the opening to the catalytic cleft shows why membrane anchoring is necessary for the lipophilic substrates to gain access to the active site. The mol. basis for the enzyme's androgenic specificity and unique catalytic mechanism can be used for developing next-generation aromatase inhibitors.
- 60Yin, L.; Hu, Q.; Hartmann, R. W. Tetrahydropyrroloquinolinone Type Dual Inhibitors of Aromatase/Aldosterone Synthase as a Novel Strategy for Breast Cancer Patients with Elevated Cardiovascular Risks. J. Med. Chem. 2013, 56 (2), 460– 470, DOI: 10.1021/jm301408t60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVanug%253D%253D&md5=71a8d28d9b81108446073c259a48b498Tetrahydropyrroloquinolinone Type Dual Inhibitors of Aromatase/Aldosterone Synthase as a Novel Strategy for Breast Cancer Patients with Elevated Cardiovascular RisksYin, Lina; Hu, Qingzhong; Hartmann, Rolf W.Journal of Medicinal Chemistry (2013), 56 (2), 460-470CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The application of aromatase inhibitors to postmenopausal breast cancer patients increases the risk of cardiovascular diseases (CVD), which is believed to be caused by the abnormally high concns. of aldosterone as a consequence of the estrogen deficiency. Dual inhibitors of aromatase (CYP19) and aldosterone synthase (CYP11B2) are therefore proposed as a novel strategy for the adjuvant therapy to reduce the CVD risk for these patients. By combining decisive structural features of CYP11B2 and CYP19 inhibitors into a common template, a series of pyridinylmethyl substituted 1,2,5,6-tetrahydropyrrolo[3,2,1-ij]quinolin-4-ones were designed and synthesized. Interestingly, the substituents on the methylene bridge showed strong influences on the inhibitory activities leading to opposite effects, i.e., a given substituent showed an increase in inhibition of one enzyme, while it led to a decrease for the other enzyme. The compromise of this conflict led to compds. 3j, 3k, 3n, and 3p as potent and selective dual inhibitors of CYP19 and CYP11B2, esp. compd. 3p (I), which exhibited IC50 values of 32 and 41 nM for CYP19 and CYP11B2, resp., and a high selectivity toward CYP17 and CYP11B1. This compd. is considered as a candidate for further evaluation in vivo.
- 61Meredith, E. L.; Ksander, G.; Monovich, L. G.; Papillon, J. P. N.; Liu, Q.; Miranda, K.; Morris, P.; Rao, C.; Burgis, R.; Capparelli, M.; Hu, Q.-Y.; Singh, A.; Rigel, D. F.; Jeng, A. Y.; Beil, M.; Fu, F.; Hu, C.-W.; LaSala, D. Discovery and in Vivo Evaluation of Potent Dual CYP11B2 (Aldosterone Synthase) and CYP11B1 Inhibitors. ACS Med. Chem. Lett. 2013, 4 (12), 1203– 1207, DOI: 10.1021/ml400324c61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhs1Ckt7vP&md5=3f2301a71dfd1bf91a99ac68f5ca91ffDiscovery and in Vivo Evaluation of Potent Dual CYP11B2 (Aldosterone Synthase) and CYP11B1 InhibitorsMeredith, Erik L.; Ksander, Gary; Monovich, Lauren G.; Papillon, Julien P. N.; Liu, Qian; Miranda, Karl; Morris, Patrick; Rao, Chang; Burgis, Robin; Capparelli, Michael; Hu, Qi-Ying; Singh, Alok; Rigel, Dean F.; Jeng, Arco Y.; Beil, Michael; Fu, Fumin; Hu, Chii-Whei; LaSala, DanielACS Medicinal Chemistry Letters (2013), 4 (12), 1203-1207CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Aldosterone is a key signaling component of the renin-angiotensin-aldosterone system and as such has been shown to contribute to cardiovascular pathol. such as hypertension and heart failure. Aldosterone synthase (CYP11B2) is responsible for the final three steps of aldosterone synthesis and thus is a viable therapeutic target. A series of imidazole derived inhibitors, including clin. candidate I, have been identified through design and structure-activity relationship studies both in vitro and in vivo. Compd. I was also found to be a potent inhibitor of 11β-hydroxylase (CYP11B1), which is responsible for cortisol prodn. Inhibition of CYP11B1 is being evaluated in the clinic for potential treatment of hypercortisol diseases such as Cushing's syndrome.
- 62Bochevarov, A. D.; Harder, E.; Hughes, T. F.; Greenwood, J. R.; Braden, D. A.; Philipp, D. M.; Rinaldo, D.; Halls, M. D.; Zhang, J.; Friesner, R. A. Jaguar: A High-performance Quantum Chemistry Software Program with Strengths in Life and Materials Sciences. Int. J. Quantum Chem. 2013, 113 (18), 2110– 2142, DOI: 10.1002/qua.2448162https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtVCqs7%252FP&md5=2fc119bb9e0501e55d7292f7735c24eaJaguar: A high-performance quantum chemistry software program with strengths in life and materials sciencesBochevarov, Art D.; Harder, Edward; Hughes, Thomas F.; Greenwood, Jeremy R.; Braden, Dale A.; Philipp, Dean M.; Rinaldo, David; Halls, Mathew D.; Zhang, Jing; Friesner, Richard A.International Journal of Quantum Chemistry (2013), 113 (18), 2110-2142CODEN: IJQCB2; ISSN:0020-7608. (John Wiley & Sons, Inc.)A review. Jaguar is an ab initio quantum chem. program that specializes in fast electronic structure predictions for mol. systems of medium and large size. Jaguar focuses on computational methods with reasonable computational scaling with the size of the system, such as d. functional theory (DFT) and local second-order Moller-Plesset perturbation theory. The favorable scaling of the methods and the high efficiency of the program make it possible to conduct routine computations involving several thousand MOs. This performance is achieved through a utilization of the pseudospectral approxn. and several levels of parallelization. The speed advantages are beneficial for applying Jaguar in biomol. computational modeling. Addnl., owing to its superior wave function guess for transition-metal-contg. systems, Jaguar finds applications in inorg. and bioinorg. chem. The emphasis on larger systems and transition metal elements paves the way toward developing Jaguar for its use in materials science modeling. The article describes the historical and new features of Jaguar, such as improved parallelization of many modules, innovations in ab initio pKa prediction, and new semiempirical corrections for nondynamic correlation errors in DFT. Jaguar applications in drug discovery, materials science, force field parameterization, and other areas of computational research are reviewed. Timing benchmarks and other results obtained from the most recent Jaguar code are provided. The article concludes with a discussion of challenges and directions for future development of the program. © 2013 Wiley Periodicals, Inc.
- 63Nelson, D. R.; Zeldin, D. C.; Hoffman, S. M.; Maltais, L. J.; Wain, H. M.; Nebert, D. W. Comparison of Cytochrome P450 (CYP) Genes from the Mouse and Human Genomes, Including Nomenclature Recommendations for Genes, Pseudogenes and Alternative-Splice Variants. Pharmacogenetics 2004, 14 (1), 1– 18, DOI: 10.1097/00008571-200401000-0000163https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjs1ags7c%253D&md5=10ee0bacd1f7213153412541bd2d9c95Comparison of cytochrome P450 (CYP) genes from the mouse and human genomes, including nomenclature recommendations for genes, pseudogenes and alternative-splice variantsNelson, David R.; Zeldin, Darryl C.; Hoffman, Susan M. G.; Maltais, Lois J.; Wain, Hester M.; Nebert, Daniel W.Pharmacogenetics (2004), 14 (1), 1-18CODEN: PHMCEE; ISSN:0960-314X. (Lippincott Williams & Wilkins)A review. Completion of both the mouse and human genome sequences in the private and public sectors has prompted comparison between the two species at multiple levels. This review summarizes the cytochrome P 450 (CYP) gene superfamily. For the first time, we have the ability to compare complete sets of CYP genes from two mammals. Use of the mouse as a model mammal, and as a surrogate for human biol., assumes reasonable similarity between the two. It is therefore of interest to catalog the genetic similarities and differences, and to clarify the limits of extrapolation from mouse to human. Data-mining methods have been used to find all the mouse and human CYP sequences; this includes 102 putatively functional genes and 88 pseudogenes in the mouse, and 57 putatively functional genes and 58 pseudogenes in the human. Comparison is made between all these genes, esp. the seven main CYP gene clusters. The seven CYP clusters are greatly expanded in the mouse with 72 functional genes vs. only 27 in the human, while many pseudogenes are present; presumably this phenomenon will be seen in many other gene superfamily clusters. Complete identification of all pseudogene sequences is likely to be clin. important, because some of these highly similar exons can interfere with PCR-based genotyping assays. A naming procedure for each of four categories of CYP pseudogenes is proposed, and we encourage various gene nomenclature committees to consider seriously the adoption and application of this pseudogene nomenclature system.
- 64Hare, S. H.; Harvey, A. J. mTOR Function and Therapeutic Targeting in Breast Cancer. Am. J. Cancer Res. 2017, 7 (3), 383– 40464https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsVGgtLbO&md5=6a8141f05942c0277f1628279ca12594mTOR function and therapeutic targeting in breast cancerHare, Stephen H.; Harvey, Amanda J.American Journal of Cancer Research (2017), 7 (3), 383-404CODEN: AJCRFT; ISSN:2156-6976. (e-Century Publishing Corp.)The mTOR pathway was discovered in the late 1970s after the compd. and natural inhibitor of mTOR, rapamycin was isolated from the bacterium Streptomyces hygroscopicus. mTOR is serine/threonine kinase belonging to the phosphoinositide 3-kinase related kinase (PIKK) family. It forms two distinct complexes; mTORC1 and mTORC2. mTORC1 has a key role in regulating protein synthesis and autophagy while mTORC2 is involved in regulating kinases of the AGC family. mTOR signaling is often over active in multiple cancer types including breast cancer. This can involve mutations in mTOR itself but more commonly, in breast cancer, this is related to an increase in activity of ErbB family receptors or alterations and mutations of PI3K signaling. Rapamycin and its analogs (rapalogues) bind to the intercellular receptor FKBP12, and then predominantly inhibit mTORC1 signaling via an allosteric mechanism. Research has shown that inhibition of mTOR is a useful strategy in tackling cancers, with it acting to slow tumor growth and limit the spread of a cancer. Rapalogues have now made their way into the clinic with the rapalogue everolimus (RAD-001/Afinitor) approved for use in conjunction with exemestane, in post-menopausal breast cancer patients with advanced disease who are HER-2 neg. (normal expression), hormone receptor pos. and whose prior treatment with non-steroidal aromatase inhibitors has failed. Testing across multiple trials has proven that everolimus and other rapalogues are a viable way of treating certain types of cancer. However, rapalogues have shown some drawbacks both in research and clin., with their use often activating feedback pathways that counter their usefulness. As such, new types of inhibitors are being explored that work via different mechanisms, including inhibitors that are ATP competitive with mTOR and which act to perturb signaling from both mTOR complexes.
- 65Hadizadeh, F.; Shafiee, A.; Kazemi, R.; Mohammadi, M. Synthesis of 4-(1-Phenylmethyl-5-Imidazolyl)-1,4-Dihydropyridines as Calcium Channel Antagonists; NISCAIR-CSIR: India, 2002; vol 41B (12), pp 2679– 2682.There is no corresponding record for this reference.
- 66Millet, R.; Domarkas, J.; Houssin, R.; Gilleron, P.; Goossens, J.-F.; Chavatte, P.; Logé, C.; Pommery, N.; Pommery, J.; Hénichart, J.-P. Potent and Selective Farnesyl Transferase Inhibitors. J. Med. Chem. 2004, 47 (27), 6812– 6820, DOI: 10.1021/jm030502y66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhtVKrs7%252FK&md5=bc8c296fc3c4c96c4da1d608d5e74502Potent and Selective Farnesyl Transferase InhibitorsMillet, Regis; Domarkas, Juozas; Houssin, Raymond; Gilleron, Pauline; Goossens, Jean-Francois; Chavatte, Philippe; Loge, Cedric; Pommery, Nicole; Pommery, Jean; Henichart, Jean-PierreJournal of Medicinal Chemistry (2004), 47 (27), 6812-6820CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)We recently described a novel series of CA1A2X peptidomimetics as farnesyl transferase inhibitors (FTIs). These compds. possess an N-(4-piperidinyl)benzamide scaffold mimicking A1A2 residue. Extensive exploration of structure-activity relationships revealed that replacement of cysteine by substituted benzylimidazoles provided nanomolar FTIs with in vitro activities (18e, IC50 = 4.60 nM on isolated enzyme, EC50 = 20.0 nM for growth inhibition on a tumor cell line). The mol. docking of 18e and 19e in the active site of the enzyme provided details of key interactions with the protein and showed that the methionine or phenylalanine residue fits into the aryl binding site.
- 67Berthold, M. R.; Cebron, N.; Dill, F.; Gabriel, T. R.; Kötter, T.; Meinl, T.; Ohl, P.; Sieb, C.; Thiel, K.; Wiswedel, B. KNIME: The Konstanz Information Miner. In Data Analysis, Machine Learning and Applications; Studies in Classification, Data Analysis, and Knowledge Organization; Preisach, C.; Burkhardt, H.; Schmidt-Thieme, L.; Decker, R., Eds.; Springer: Berlin, Heidelberg, 2008; pp 319– 326.There is no corresponding record for this reference.
- 68Schrödinger Release 2018–3: LigPrep; Schrödinger, LLC: New York, NY, 2018.There is no corresponding record for this reference.
- 69Hawkins, P. C. D.; Skillman, A. G.; Warren, G. L.; Ellingson, B. A.; Stahl, M. T. Conformer Generation with OMEGA: Algorithm and Validation Using High Quality Structures from the Protein Databank and Cambridge Structural Database. J. Chem. Inf. Model. 2010, 50 (4), 572– 584, DOI: 10.1021/ci100031x69https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjtlaisrY%253D&md5=fb87ecc9c51eddef63b41fffcd9babeeConformer Generation with OMEGA: Algorithm and Validation Using High Quality Structures from the Protein Databank and Cambridge Structural DatabaseHawkins, Paul C. D.; Skillman, A. Geoffrey; Warren, Gregory L.; Ellingson, Benjamin A.; Stahl, Matthew T.Journal of Chemical Information and Modeling (2010), 50 (4), 572-584CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Here, we present the algorithm and validation for OMEGA, a systematic, knowledge-based conformer generator. The algorithm consists of three phases: assembly of an initial 3D structure from a library of fragments; exhaustive enumeration of all rotatable torsions using values drawn from a knowledge-based list of angles, thereby generating a large set of conformations; and sampling of this set by geometric and energy criteria. Validation of conformer generators like OMEGA has often been undertaken by comparing computed conformer sets to exptl. mol. conformations from crystallog., usually from the Protein Databank (PDB). Such an approach is fraught with difficulty due to the systematic problems with small mol. structures in the PDB. Methods are presented to identify a diverse set of small mol. structures from cocomplexes in the PDB that has maximal reliability. A challenging set of 197 high quality, carefully selected ligand structures from well-solved models was obtained using these methods. This set will provide a sound basis for comparison and validation of conformer generators in the future. Validation results from this set are compared to the results using structures of a set of druglike mols. extd. from the Cambridge Structural Database (CSD). OMEGA is found to perform very well in reproducing the crystallog. conformations from both these data sets using two complementary metrics of success.
- 70Pinzi, L.; Caporuscio, F.; Rastelli, G. Selection of Protein Conformations for Structure-Based Polypharmacology Studies. Drug Discovery Today 2018, 23 (11), 1889– 1896, DOI: 10.1016/j.drudis.2018.08.00770https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsFeqt73F&md5=e0b96eeeb37b37fd4d5e5dea465d9a88Selection of protein conformations for structure-based polypharmacology studiesPinzi, Luca; Caporuscio, Fabiana; Rastelli, GiulioDrug Discovery Today (2018), 23 (11), 1889-1896CODEN: DDTOFS; ISSN:1359-6446. (Elsevier Ltd.)A review. Several drugs exert their therapeutic effect through the modulation of multiple targets. Structure-based approaches hold great promise for identifying compds. with the desired polypharmacol. profiles. These methods use knowledge of the protein binding sites to identify stereoelectronically complementary ligands. The selection of the most suitable protein conformations to be used in the design process is vital, esp. for multitarget drug design in which the same ligand has to be accommodated in multiple binding pockets. Herein, we focus on currently available techniques for the selection of the most suitable protein conformations for multitarget drug design, compare the potential advantages and limitations of each method, and comment on how their combination could help in polypharmacol. drug design.
- 71Pinzi, L.; Rastelli, G. Molecular Docking: Shifting Paradigms in Drug Discovery. Int. J. Mol. Sci. 2019, 20 (18), 4331, DOI: 10.3390/ijms2018433171https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVCns70%253D&md5=9ed9e680a89538b1ef1f11f6cd2b7e68Molecular docking: shifting paradigms in drug discoveryPinzi, Luca; Rastelli, GiulioInternational Journal of Molecular Sciences (2019), 20 (18), 4331CODEN: IJMCFK; ISSN:1422-0067. (MDPI AG)A review. Mol. docking is an established in silico structure-based method widely used in drug discovery. Docking enables the identification of novel compds. of therapeutic interest, predicting ligand-target interactions at a mol. level, or delineating structure-activity relationships (SAR), without knowing a priori the chem. structure of other target modulators. Although it was originally developed to help understanding the mechanisms of mol. recognition between small and large mols., uses and applications of docking in drug discovery have heavily changed over the last years. In this review, we describe how mol. docking was firstly applied to assist in drug discovery tasks. Then, we illustrate newer and emergent uses and applications of docking, including prediction of adverse effects, polypharmacol., drug repurposing, and target fishing and profiling, discussing also future applications and further potential of this technique when combined with emergent techniques, such as artificial intelligence.
- 72Madhavi Sastry, G.; Adzhigirey, M.; Day, T.; Annabhimoju, R.; Sherman, W. Protein and Ligand Preparation: Parameters, Protocols, and Influence on Virtual Screening Enrichments. J. Comput. Aided Mol. Des. 2013, 27 (3), 221– 234, DOI: 10.1007/s10822-013-9644-872https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXmslalu7c%253D&md5=259a6d547ef3e1310e091fb50fe8de16Protein and ligand preparation: parameters, protocols, and influence on virtual screening enrichmentsMadhavi Sastry, G.; Adzhigirey, Matvey; Day, Tyler; Annabhimoju, Ramakrishna; Sherman, WoodyJournal of Computer-Aided Molecular Design (2013), 27 (3), 221-234CODEN: JCADEQ; ISSN:0920-654X. (Springer)Structure-based virtual screening plays an important role in drug discovery and complements other screening approaches. In general, protein crystal structures are prepd. prior to docking in order to add hydrogen atoms, optimize hydrogen bonds, remove at. clashes, and perform other operations that are not part of the x-ray crystal structure refinement process. In addn., ligands must be prepd. to create 3-dimensional geometries, assign proper bond orders, and generate accessible tautomer and ionization states prior to virtual screening. While the prerequisite for proper system prepn. is generally accepted in the field, an extensive study of the prepn. steps and their effect on virtual screening enrichments has not been performed. In this work, we systematically explore each of the steps involved in prepg. a system for virtual screening. We first explore a large no. of parameters using the Glide validation set of 36 crystal structures and 1,000 decoys. We then apply a subset of protocols to the DUD database. We show that database enrichment is improved with proper prepn. and that neglecting certain steps of the prepn. process produces a systematic degrdn. in enrichments, which can be large for some targets. We provide examples illustrating the structural changes introduced by the prepn. that impact database enrichment. While the work presented here was performed with the Protein Prepn. Wizard and Glide, the insights and guidance are expected to be generalizable to structure-based virtual screening with other docking methods.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsptsci.3c00183.
Results of the similarity estimations performed with the LigAdvisor Web server; results of the similarity estimations performed with respect to compounds with activity annotation on CYP19A1, CYP11B2, and CYP11B1, reported in the DrugBank, PDB, and ChEMBL databases; docking scores of the investigated compounds into the PDB crystal structures 3EQM (CYP19A1), 6M7X (CYP11B1), and 4FDH (CYP11B2); binding mode predicted for (S)- and (R)-fadrozole into the CYP19A1 binding site (PDB ID: 3EQM); titration curves of compound X21 against CYP19A1, CYP1A2, and CYP3A4, with their respective controls; antiproliferative in vitro activity of fadrozole on human MCF-7 (Estrogen Receptor+) at 24, 48, and 72 h; and titration curves of compound X21 and the reference compounds E-4031 and tetrodoxin, against hERG and Nav1.5 (manual patch clamp assays), respectively (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.