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...

Computational Study of Estrogen Receptor-Alpha Antagonist with Three-Dimensional Quantitative Structure-Activity Relationship, Support Vector Regression, and Linear Regression Methods

DOI: 10.1155/2013/743139

Full-Text   Cite this paper   Add to My Lib

Abstract:

Human estrogen receptor (ER) isoforms, ERα and ERβ, have long been an important focus in the field of biology. To better understand the structural features associated with the binding of ERα ligands to ERα and modulate their function, several QSAR models, including CoMFA, CoMSIA, SVR, and LR methods, have been employed to predict the inhibitory activity of 68 raloxifene derivatives. In the SVR and LR modeling, 11 descriptors were selected through feature ranking and sequential feature addition/deletion to generate equations to predict the inhibitory activity toward ERα. Among four descriptors that constantly appear in various generated equations, two agree with CoMFA and CoMSIA steric fields and another two can be correlated to a calculated electrostatic potential of ERα. 1. Introduction Estrogens are critical in the physiology of the female reproductive system, the maintenance of bone density, and cardiovascular health [1, 2]. Estrogen receptors are classified into two isoforms, ERα and ERβ, both of which are members of the nuclear receptor superfamily of ligand-modulated transcription factors [3, 4]. When the natural ligand estradiol or other ligands bind to ERα, complex signaling networks lead to a conformational change, specifically in the activation function (AF)-2 helix (H12), allowing estradiol to bind to chromatin; this, in turn, activates or inhibits responsive genes [5, 6]. ERα and ERβ are the targets of pharmaceutical agents used to fight cancers of the reproductive organs, for example, prostate, uterine, and breast cancer [6, 7]. These pharmaceutical agents are divided into three distinct categories: (i) receptor agonists such as 17β-estradiol, the estrogen receptor’s natural ligand; (ii) antiestrogens, such as the compound ICI 164,384 [5, 8]; and (iii) raloxifene (arylbenzothiophene) [5, 9] and tamoxifen [10], both of which act as agonists as well as antagonists. Raloxifene (compound 25 in Table 1) is a selective estrogen receptor modulator (SERM) providing a safer alternative to estrogen because it is an ER antagonist in mammary tissue and the uterus and also mimics the agonist effects of estrogen on bone and in the cardiovascular system [11]. The U.S. Food and Drug Administration (FDA) recently approved raloxifene for the treatment of osteoporosis [12], and it is also being tested as a preventive drug against breast cancer and coronary heart disease [5, 9]. Because drug resistance and serious side effects, such as venous thromboembolism and fatal stroke, have been reported [13], there is a crucial need for new therapeutic agents. Two

References

[1]  G. E. Christodoulakos, I. V. Lambrinoudaki, and D. C. Botsis, “The cardiovascular effects of selective estrogen receptor modulators,” Annals of the New York Academy of Sciences, vol. 1092, pp. 374–384, 2006.
[2]  E. Bonnelye and J. E. Aubin, “Estrogen receptor-related receptor α: a mediator of estrogen response in bone,” Journal of Clinical Endocrinology and Metabolism, vol. 90, no. 5, pp. 3115–3121, 2005.
[3]  S. J. McPherson, S. J. Ellem, V. Patchev, K. H. Fritzemeier, and G. P. Risbridger, “The role of Erα and ERβ in the prostate: insights from genetic models and isoform-selective ligands,” Ernst Schering Foundation symposium proceedings, vol. 1, pp. 131–147, 2006.
[4]  P. A. Arias-Loza, V. Jazbutyte, K. H. Fritzemeier et al., “Functional effects and molecular mechanisms of subtype-selective ERα and ERβ agonists in the cardiovascular system,” Ernst Schering Foundation symposium proceedings, vol. 1, pp. 87–106, 2006.
[5]  M. Lupien, M. Jeyakumar, E. Hébert et al., “Raloxifene and ICI182,780 increase estrogen receptor-α association with a nuclear compartment via overlapping sets of hydrophobic amino acids in activation function 2 helix 12,” Molecular Endocrinology, vol. 21, no. 4, pp. 797–816, 2007.
[6]  M. Nichols, P. Cheng, Y. Liu, B. Kanterewicz, P. A. Hershberger, and K. S. McCarty, “Breast cancer-derived M543V mutation in helix 12 of estrogen receptor α inverts response to estrogen and SERMs,” Breast Cancer Research and Treatment, vol. 120, no. 3, pp. 761–768, 2010.
[7]  J. Peng, S. Sengupta, and V. C. Jordan, “Potential of selective estrogen receptor modulators as treatments and preventives of breast cancer,” Anti-Cancer Agents in Medicinal Chemistry, vol. 9, no. 5, pp. 481–499, 2009.
[8]  R. B. Riggins, A. Zwart, R. Nehra, and R. Clarke, “The nuclear factor κB inhibitor parthenolide restores ICI 182,780 (Faslodex; fulvestrant)-induced apoptosis in antiestrogen-resistant breast cancer cells,” Molecular Cancer Therapeutics, vol. 4, no. 1, pp. 33–41, 2005.
[9]  K. Visvanathan, R. T. Chlebowski, P. Hurley et al., “American society of clinical oncology clinical practice guideline update on the use of pharmacologic interventions including tamoxifen, raloxifene, and aromatase inhibition for breast cancer risk reduction,” Journal of Clinical Oncology, vol. 27, no. 19, pp. 3235–3258, 2009.
[10]  G. Bertelli, E. Hall, E. Ireland et al., “Long-term endometrial effects in postmenopausal women with early breast cancer participating in the Intergroup Exemestane Study (IES)—a randomised controlled trial of exemestane versus continued tamoxifen after 2-3 years tamoxifen,” Annals of Oncology, vol. 21, no. 3, Article ID mdp358, pp. 498–505, 2009.
[11]  B. Biersack and R. Schobert, “Metallodrug conjugates with steroids and selective estrogen receptor modulators (SERM),” Current Medicinal Chemistry, vol. 16, no. 18, pp. 2324–2337, 2009.
[12]  FDA: FDA approves new uses for Evista, 2007, http://www.fda.gov/bbs/topics/NEWS/2007/NEW01698.html.
[13]  E. Barrett-Connor, L. Mosca, P. Collins et al., “Effects of raloxifene on cardiovascular events and breast cancer in postmenopausal women,” The New England Journal of Medicine, vol. 355, no. 2, pp. 125–137, 2006.
[14]  U. Norinder, “Support vector machine models in drug design: applications to drug transport processes and QSAR using simplex optimisations and variable selection,” Neurocomputing, vol. 55, no. 1-2, pp. 337–346, 2003.
[15]  H. F. Chen, “Computational study of histamine H3-receptor antagonist with support vector machines and three dimension quantitative structure activity relationship methods,” Analytica Chimica Acta, vol. 624, no. 2, pp. 203–209, 2008.
[16]  Z. Cheng, Y. Zhang, and W. Fu, “QSAR study of carboxylic acid derivatives as HIV-1 Integrase inhibitors,” European Journal of Medicinal Chemistry, vol. 45, no. 9, pp. 3970–3980, 2010.
[17]  W. Shi, X. Zhang, and Q. Shen, “Quantitative structure-activity relationships studies of CCR5 inhibitors and toxicity of aromatic compounds using gene expression programming,” European Journal of Medicinal Chemistry, vol. 45, no. 1, pp. 49–54, 2010.
[18]  A. Yan, Y. Chong, L. Wang, X. Hu, and K. Wang, “Prediction of biological activity of Aurora-A kinase inhibitors by multilinear regression analysis and support vector machine,” Bioorganic and Medicinal Chemistry Letters, vol. 21, no. 8, pp. 2238–2243, 2011.
[19]  T. A. Grese, S. Cho, D. R. Finley et al., “Structure-activity relationships of selective estrogen receptor modulators: modifications to the 2-arylbenzothiophene core of raloxifene,” Journal of Medicinal Chemistry, vol. 40, no. 2, pp. 146–167, 1997.
[20]  R. D. Cramer, D. E. Patterson, and J. D. Bunce, “Comparative molecular field analysis (CoMFA). 1. Effect of shape on binding of steroids to carrier proteins,” Journal of the American Chemical Society, vol. 110, no. 18, pp. 5959–5967, 1988.
[21]  H. Kubinyi, Comparative Molecular Field Analysis (CoMFA), The Encyclopedia of Computational Chemistry, 1998.
[22]  G. Klebe, U. Abraham, and T. Mietzner, “Molecular similarity indices in a comparative analysis (CoMSIA) of drug molecules to correlate and predict their biological activity,” Journal of Medicinal Chemistry, vol. 37, no. 24, pp. 4130–4146, 1994.
[23]  A. C. Wold S, W. J. Dunn III, U. Edlund et al., “Multivariate data analysis in chemistry,” in Chemometrics: Mathematics and Statistics in Chemistry, B. Kowalski, Ed., Reidel, Dordrecht, The Netherlands, 1984.
[24]  R. D. Cramer III, J. D. Bunce, D. E. Patterson, et al., “Crossvalidation, bootstrapping, and partial least squares compared with multiple regression in conventional QSAR studies,” Quantitative Structure-Activity Relationships, vol. 7, no. 1, pp. 18–25, 1988.
[25]  T. G. Gantchev, H. Ali, and J. E. Van Lier, “Quantitative structure-activity relationships/comparative molecular field analysis (QSAR/CoMFA) for receptor-binding properties of halogenated estradiol derivatives,” Journal of Medicinal Chemistry, vol. 37, no. 24, pp. 4164–4176, 1994.
[26]  P. Wolohan and D. E. Reichert, “CoMFA and docking study of novel estrogen receptor subtype selective ligands,” Journal of Computer-Aided Molecular Design, vol. 17, no. 5-6, pp. 313–328, 2003.
[27]  T. G. Gantchev, H. Ali, and J. E. V. Lier, “Quantitative structure-activity relationships/comparative molecular field analysis (QSAR/CoMFA) for receptor-binding properties of halogenated estradiol derivatives,” Journal of Medicinal Chemistry, vol. 37, no. 24, pp. 4164–4176, 1994.
[28]  H. Peng, F. Long, and C. Ding, “Feature selection based on mutual information: criteria of max-dependency, max-relevance, and min-redundancy,” IEEE Transactions on Pattern Analysis and Machine Intelligence, vol. 27, no. 8, pp. 1226–1238, 2005.
[29]  C. M. Bishop, Pattern Recognition and Machine Learning, Springer, New York, NY, USA, 2006.
[30]  A. M. Brzozowski, A. C. W. Pike, Z. Dauter et al., “Molecular basis of agonism and antagonism in the oestrogen receptor,” Nature, vol. 389, no. 6652, pp. 753–758, 1997.
[31]  L. B. Salum, I. Polikarpov, and A. D. Andricopulo, “Structure-based approach for the study of estrogen receptor binding affinity and subtype selectivity,” Journal of Chemical Information and Modeling, vol. 48, no. 11, pp. 2243–2253, 2008.
[32]  J. Sun, J. Baudry, J. A. Katzenellenbogen, and B. S. Katzenellenbogen, “Molecular basis for the subtype discrimination of the estrogen receptor-β-selective ligand, diarylpropionitrile,” Molecular Endocrinology, vol. 17, no. 2, pp. 247–258, 2003.
[33]  R. W. Hsieh, S. S. Rajan, S. K. Sharma et al., “Identification of ligands with bicyclic scaffolds provides insights into mechanisms of estrogen receptor subtype selectivity,” The Journal of Biological Chemistry, vol. 281, no. 26, pp. 17909–17919, 2006.
[34]  C. C. Chang and C. J. Lin, LIBSVM: A library for Support Vector Machines, 2001, http://www.csie.ntu.edu.tw/~cjlin/libsvm/.
[35]  V. N. Vapnik, The Nature of Statistical Learning Theory, Springer, New York, NY, USA, 1995.
[36]  T. Z. Bao, G. Z. Han, J. Y. Shim, Y. Wen, and X. R. Jiang, “Quantitative structure-activity relationship of various endogenous estrogen metabolites for human estrogen receptor α and β subtypes: insights into the structural determinants favoring a differential subtype binding,” Endocrinology, vol. 147, no. 9, pp. 4132–4150, 2006.
[37]  R. K. Dubey, S. P. Tofovic, and E. K. Jackson, “Cardiovascular Pharmacology of estradiol metabolites,” Journal of Pharmacology and Experimental Therapeutics, vol. 308, no. 2, pp. 403–409, 2004.
[38]  M. Chang, K. W. Peng, I. Kastrati et al., “Activation of estrogen receptor-mediated gene transcription by the equine estrogen metabolite, 4-methoxyequilenin, in human breast cancer cells,” Endocrinology, vol. 148, no. 10, pp. 4793–4802, 2007.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133