全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Molecular Docking Studies of Estrone-Coumarin Derivatives as Aromatase and 17β-HSD1 Inhibitors Related to Hormone Receptor Positive (HR+) Breast Cancer

DOI: 10.4236/aer.2022.104006, PP. 83-100

Keywords: Estrone, Coumarins, Aromatase Inhibitors, 17β-HSD1 Inhibitors, Molecular Docking

Full-Text   Cite this paper   Add to My Lib

Abstract:

Hormone Receptor positive (HR+) breast cancer is the most common malignancy in women. New strategies in the treatments have targeted the estrogen biosynthesis pathways including the inhibition of the aromatase and 17β-HSD1 enzymes. The present work, describes the study of a new family of 9 hybrid compounds derived from estrone attached to a coumarin fragment, linked through different lengths of hydrocarbon chains. The activity of these compounds was evaluated by molecular docking with two relevant enzymes in breast cancer (HR+). It has been proposed nine compounds as 17β-HSD1 inhibitors and six as aromatase inhibitors. We found important interactions with key amino acids at the orthosteric site of each enzyme and their score values compared to the crystallographic ligand. The in silico analysis showed good score values in the proposed compounds, where the steroidal portion presented important interactions with Met374 and Tyr155 in aromatase and in 17β-HSD1 respectively. Highlighting Compounds 2, 5 and 8 with an aromatic ring at the C4 position of the coumarin moiety, which favored arene-H type interactions essential for protein-ligand recognition. In addition, the results related to the 17β-HSD1 enzyme demonstrated how the length of the linker influences the interaction; the best score was found for derivative 8 with a chain of 8 methylenes.

References

[1]  DeSantis, C.E., Ma, J., Goding Sauer, A., Newman, L.A. and Jemal, A. (2017) Breast Cancer Statistics, 2017, Racial Disparity in Mortality by State. CA: A Cancer Journal for Clinicians, 67, 439-448.
https://doi.org/10.3322/caac.21412
[2]  Brueggemeier, R.W., Hackett, J.C. and Diaz-Cruz, E.S. (2005) Aromatase Inhibitors in the Treatment of Breast Cancer. Endocrine Reviews, 26, 331-345.
https://doi.org/10.1210/er.2004-0015
[3]  Ghosh, D., Griswold, J., Erman, M. and Pangborn, W. (2009) Structural Basis for Androgen Specificity and Oestrogen Synthesis in Human Aromatase. Nature, 457, 219-223.
https://doi.org/10.1038/nature07614
[4]  Roberts, K., Rochefort, J., Bleau, G. and Chapdelaine, A. (1980) Plasma Estrone Sulfate Levels in Postmenopausal Women. Steroids, 35, 179-187.
https://doi.org/10.1016/0039-128X(80)90101-4
[5]  Aka, J.A., Mazumdar, M. and Lin, S.-X. (2009) Reductive 17β-Hydroxysteroid Dehydrogenases in the Sulfatase Pathway: Critical in the Cell Proliferation of Breast Cancer. Molecular and Cellular Endocrinology, 301, 183-190.
https://doi.org/10.1016/j.mce.2008.10.042
[6]  Aka, J.A., Mazumdar, M., Chen, C.Q., Poirier, D. and Lin, S.X. (2010) 17β-Hydroxysteroid Dehydrogenase Type 1 Stimulates Breast Cancer by Dihydrotestosterone Inactivation in Addition to Estradiol Production. Molecular Endocrinology, 24, 832-845.
https://doi.org/10.1210/me.2009-0468
[7]  Thomas, M.P. and Potter, B.V. (2013) The Structural Biology of Oestrogen Metabolism. The Journal of Steroid Biochemistry and Molecular Biology, 137, 27-49.
https://doi.org/10.1016/j.jsbmb.2012.12.014
[8]  Ramsay, R.R., Popovic-Nikolic, M.R., Nikolic, K., Uliassi, E. and Bolognesi, M.L. (2018) A Perspective on Multi-Target Drug Discovery and Design for Complex Diseases. Clinical and Translational Medicine, 7, Article No. 3.
https://doi.org/10.1186/s40169-017-0181-2
[9]  Tremblay, M.R., Auger, S. and Poirier, D. (1995) Synthesis of 16-(Bromoalkyl)-Estradiols Having Inhibitory Effect on Human Placental Estradiol 17β-Hydroxysteroid Dehydrogenase (17-β-HSD Type 1). Bioorganic & Medicinal Chemistry, 3, 505-523.
https://doi.org/10.1016/0968-0896(95)00041-E
[10]  Pelletier, J.D. and Poirier, D. (1996) Synthesis and Evaluation of Estradiol Derivatives with 16α-(Bromoalkylamide), 16α-(Bromoalkyl) or 16α-(Bromoalkynyl) Side Chain as Inhibitors of 17β-Hydroxysteroid Dehydrogenase Type 1 without Estrogenic Activity. Bioorganic & Medicinal Chemistry, 4, 1617-1628.
https://doi.org/10.1016/0968-0896(96)00154-X
[11]  Poirier, D., Boivin, R.P., Tremblay, M.R., Bérubé, M., Qiu, W., et al. (2005) Estradiol-Adenosine Hybrid Compounds Designed to Inhibit Type 1 17β-Hydroxysteroid Dehydrogenase. Journal of Medicinal Chemistry, 48, 8134-8147.
https://doi.org/10.1021/jm058235e
[12]  Karthikeyan, C., Solomon, V.R., Lee, H. and Trivedi, P. (2013) Design, Synthesis and Biological Evaluation of Some Isatin-Linked Chalcones as Novel Anti-Breast Cancer Agents: A Molecular Hybridization Approach. Biomedicine & Preventive Nutrition, 3, 325-330.
https://doi.org/10.1016/j.bionut.2013.04.001
[13]  Abu Almaaty, A.H., Elgrahy, N.A., Fayad, E., Abu Ali, O.A., Mahdy, A.R.E., et al. (2021) Design, Synthesis and Anticancer Evaluation of Substituted Cinnamic Acid Bearing 2-Quinolone Hybrid Derivatives. Molecules, 26, Article No. 4724.
https://doi.org/10.3390/molecules26164724
[14]  Ghosh, D., Lo, J., Morton, D., Valette, D., Xi, J., et al. (2012) Novel Aromatase Inhibitors by Structure-Guided Design. Journal of Medicinal Chemistry, 55, 8464-8476.
https://doi.org/10.1021/jm300930n
[15]  Balewski, U., Szulta, S., Jalińska, A. and Kornicka, A. (2021) A Mini-Review: Recent Advances in Coumarin-Metal Complexes with Biological Properties. Frontiers in Chemistry, 9, Article 781779.
https://doi.org/10.3389/fchem.2021.781779
[16]  Luna-Guevara, M.L., Luna-Guevara, J.J., Hernández-Carranza, P., Ruíz-Espinosa, H. and Ochoa-Velasco, C.E. (2018) Phenolic Compounds: A Good Choice against Chronic Degenerative Diseases. Studies in Natural Products Chemistry, 59, 79-108.
https://doi.org/10.1016/B978-0-444-64179-3.00003-7
[17]  He, Q., Liu, J., Lan, J.S., Ding, J., Sun, Y., et al. (2018) Coumarin-Dithiocarbamate Hybrids as Novel Multitarget AChE and MAO-B Inhibitors against Alzheimer’s Disease: Design, Synthesis and Biological Evaluation. Bioorganic Chemistry, 81, 512-528.
https://doi.org/10.1016/j.bioorg.2018.09.010
[18]  Sharma, A., Tiwari, M. and Supuran, C.T. (2013) Novel Coumarins and Benzocoumarins Acting as Isoform-Selective Inhibitors against the Tumor-Associated Carbonic Anhydrase IX. Journal of Enzyme Inhibition and Medicinal Chemistry, 29, 292-296.
https://doi.org/10.3109/14756366.2013.777334
[19]  Touisni, N., Maresca, A., McDonald, P.C., Lou, Y., Scozzafava, A., et al. (2011) Glycosyl Coumarin Carbonic Anhydrase IX and XII Inhibitors Strongly Attenuate the Growth of Primary Breast Tumors. Journal of Medicinal Chemistry, 54, 8271-8277.
https://doi.org/10.1021/jm200983e
[20]  Ahmad, I. and Shagufta (2015) Recent Developments in Steroidal and Nonsteroidal Aromatase Inhibitors for the Chemoprevention of Estrogen-Dependent Breast Cancer. European Journal of Medicinal Chemistry, 102, 375-386.
https://doi.org/10.1016/j.ejmech.2015.08.010
[21]  Selcer, K.W., Jagannathan, S., Rhodes, M.E. and Li, P.K. (1996) Inhibition of Placental Estrone Sulfatase Activity and MCF-7 Breast Cancer Cell Proliferation by Estrone-3-Amino Derivatives. The Journal of Steroid Biochemistry and Molecular Biology, 59, 83-91.
https://doi.org/10.1016/S0960-0760(96)00089-1
[22]  Recanatini, M., Bisi, A., Cavalli, A., Belluti, F., Gobbi, S., et al. (2001) A New Class of Nonsteroidal Aromatase Inhibitors: Design and Synthesis of Chromone and Xanthone Derivatives and Inhibition of the P450 Enzymes Aromatase and 17α-Hydroxylase/C17,20-Lyase. Journal of Medicinal Chemistry, 44, 672-680.
https://doi.org/10.1021/jm000955s
[23]  Leonetti, F., Favia, A., Rao, A., Aliano, R., Paluszcak, A., et al. (2004) Design, Synthesis, and 3D QSAR of Novel Potent and Selective Aromatase Inhibitors. Journal of Medicinal Chemistry, 47, 6792-6803.
https://doi.org/10.1021/jm049535j
[24]  Ghodsi, R. and Hemmateenejad, B. (2016) QSAR Study of Diarylalkylimidazole and Diarylalkyltriazole Aromatase Inhibitors. Medicinal Chemistry Research, 25, 834-842.
https://doi.org/10.1007/s00044-016-1530-1
[25]  Chan, H.S., Shan, H., Dahoun, T., Vogel, H. and Yuan, S. (2019) Advancing Drug Discovery via Artificial Intelligence. Trends in Pharmacological Sciences, 40, 592-604.
https://doi.org/10.1016/j.tips.2019.06.004
[26]  Schlander, M., Hernandez-Villafuerte, K., Cheng, C.Y., Mestre-Ferrandiz, J. and Baumann, M. (2021) How Much Does It Cost to Research and Develop a New Drug? A Systematic Review and Assessment. PharmacoEconomics, 39, 1243-1269.
https://doi.org/10.1007/s40273-021-01065-y
[27]  Medina-Franco, J.L., Martinez-Mayorga, K., Fernández-de Gortari, E., Kirchmair, J. and Bajorath, J. (2021) Rationality over Fashion and Hype in Drug Design. F1000Research, 10, Article No. 397.
https://doi.org/10.12688/f1000research.52676.1
[28]  Prieto-Martínez, F.D., Arciniega, M. and Medina-Franco, J.L. (2018) Acoplamiento Molecular: Avances Recientes y Retos. TIP Revista Especializada en Ciencias Químico-Biológicas, 21, 65-87.
https://doi.org/10.22201/fesz.23958723e.2018.0.143
[29]  Naqvi, A.A.T., Mohammad, T., Hasan, G.M. and Hassan, M.I. (2018) Advancements in Docking and Molecular Dynamics Simulations towards Ligand-Receptor Interactions and Structure-function Relationships. Current Topics in Medicinal Chemistry, 18, 1755-1768.
https://doi.org/10.2174/1568026618666181025114157
[30]  Pinzi, L. and Rastelli, G. (2019) Molecular Docking: Shifting Paradigms in Drug Discovery. International Journal of Molecular Sciences, 20, Article No. 4331.
https://doi.org/10.3390/ijms20184331
[31]  Ghosh, D., Egbuta, C. and Lo, J. (2018) Testosterone Complex and Non-Steroidal Ligands of Human Aromatase. The Journal of Steroid Biochemistry and Molecular Biology, 181, 11-19.
https://doi.org/10.1016/j.jsbmb.2018.02.009
[32]  Bhuvaneswari, K., Sivaguru, P. and Lalitha, A. (2020) Synthesis, Anticancer Evaluation, and Docking Studies of Some Novel Azo Chromene Derivatives. Journal of the Chinese Chemical Society, 67, 1877-1886.
https://doi.org/10.1002/jccs.201900481
[33]  Li, J., Tang, Y., Li, W. and Tu, Y. (2020) Mechanistic Insights into the Regio- and Stereoselectivities of Testosterone and Dihydrotestosterone Hydroxylation Catalyzed by CYP3A4 and CYP19A1. Chemistry—A European Journal, 26, 6214-6223.
https://doi.org/10.1002/chem.201905272
[34]  Egli, M. and Sarkhel, S. (2006) Lone Pair-Aromatic Interactions: To Stabilize or Not to Stabilize. Accounts of Chemical Research, 40, 197-205.
https://doi.org/10.1021/ar068174u
[35]  Hayakawa, D., Sawada, N., Watanabe, Y. and Gouda, H. (2020) A Molecular Interaction Field Describing Nonconventional Intermolecular Interactions and Its Application to Protein-Ligand Interaction Prediction. Journal of Molecular Graphics and Modelling, 96, Article ID: 107515.
https://doi.org/10.1016/j.jmgm.2019.107515
[36]  Breton, R., Housset, D., Mazza, C. and Fontecilla-Camps, J.C. (1996) The Structure of a Complex of Human 17β-Hydroxysteroid Dehydrogenase with Estradiol and NADP+ Identifies Two Principal Targets for the Design of Inhibitors. Structure, 4, 905-915.
https://doi.org/10.1016/S0969-2126(96)00098-6
[37]  Hernández-López, H., Leyva-Ramos, S., Azael Gómez-Durán, C.F., Pedraza-Alvarez, A., Rodríguez-Gutiérrez, I.R., et al. (2020) Synthesis of 1,4-Biphenyl-Triazole Derivatives as Possible 17β-HSD1 Inhibitors: An in Silico Study. ACS Omega, 5, 14061-14068.
https://doi.org/10.1021/acsomega.0c01519
[38]  Tremblay, M.R., Lin, S.X. and Poirier, D. (2001) Chemical Synthesis of 16β-Propylaminoacyl Derivatives of Estradiol and Their Inhibitory Potency on Type 1 17β-Hydroxysteroid Dehydrogenase and Binding Affinity on Steroid Receptors. Steroids, 66, 821-831.
https://doi.org/10.1016/S0039-128X(01)00116-7
[39]  Qiu, W., Campbell, R.L., Gangloff, A., Dupuis, P., Boivin, R.P., et al. (2002) A Concerted, Rational Design of Type 1 17β-Hydroxysteroid Dehydrogenase Inhibitors: Estradiol-Adenosine Hybrids with High Affinity. The FASEB Journal, 16, 1695-1624.
https://doi.org/10.1096/fj.02-0026fje
[40]  Salah, M., Abdelsamie, A.S. and Frotscher, M. (2019) Inhibitors of 17β-Hydroxysteroid Dehydrogenase Type 1, 2 and 14: Structures, Biological Activities and Future Challenges. Molecular and Cellular Endocrinology, 489, 66-81.
https://doi.org/10.1016/j.mce.2018.10.001
[41]  Li, T., Stephen, P., Zhu, D., Shi, R. and Lin, S. (2019) Crystal Structures of Human 17β-Hydroxysteroid Dehydrogenase Type 1 Complexed with Estrone and NADP+ Reveal the Mechanism of Substrate Inhibition. The FEBS Journal, 286, 2155-2166.
https://doi.org/10.1111/febs.14784
[42]  Chen, C., Joo, J.C., Brown, G., Stolnikova, E., Halavaty, A.S., et al. (2014) Structure-Based Mutational Studies of Substrate Inhibition of Betaine Aldehyde Dehydrogenase BetB from Staphylococcus aureus. Applied and Environmental Microbiology, 80, 3992-4002.
https://doi.org/10.1128/AEM.00215-14
[43]  Efimov, I., Basran, J., Sun, X., Chauhan, N., Chapman, S.K., et al. (2012) The Mechanism of Substrate Inhibition in Human Indoleamine 2,3-Dioxygenase. Journal of the American Chemical Society, 134, 3034-3041.
https://doi.org/10.1021/ja208694g
[44]  Mazumdar, M., Fournier, D., Zhu, D.W., Cadot, C., Poirier, D., et al. (2009) Binary and Ternary Crystal Structure Analyses of a Novel Inhibitor with 17β-HSD Type 1: A Lead Compound for Breast Cancer Therapy. Biochemical Journal, 424, 357-366.
https://doi.org/10.1042/BJ20091020
[45]  Ghosh, D. and Vihko, P. (2001) Molecular Mechanisms of Estrogen Recognition and 17-Keto Reduction by Human 17β-Hydroxysteroid Dehydrogenase 1. Chemico-Biological Interactions, 130, 637-650.
https://doi.org/10.1016/S0009-2797(00)00255-6

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133