Skip to main content Accessibility help
×
Hostname: page-component-77c89778f8-vsgnj Total loading time: 0 Render date: 2024-07-17T15:37:58.987Z Has data issue: false hasContentIssue false

17 - Structure-based design of potent glycogen phosphorylase inhibitors

from PART III - APPLICATIONS TO DRUG DISCOVERY

Published online by Cambridge University Press:  06 July 2010

Kenneth M. Merz, Jr
Affiliation:
University of Florida
Dagmar Ringe
Affiliation:
Brandeis University, Massachusetts
Charles H. Reynolds
Affiliation:
Johnson & Johnson Pharmaceutical Research & Development
Get access

Summary

INTRODUCTION

Diabetes is a disorder of metabolism and is widely recognized as one of the leading causes of death and disability. It is estimated that more than 180 million people worldwide have diabetes. In the United States, more than 20 million people – about 7.0% of the population – have diabetes. Diabetes is a lifelong condition that, if left untreated, can lead to serious complications such as nerve damage, kidney failure, blindness, and cardiovascular diseases. Type 2 diabetes is a chronic metabolic disorder characterized by fed and fasting hyperglycemia. Glycogen phosphorylase (GP) is a key enzyme in the regulation of glycogen metabolism by catalyzing the breakdown of glycogen to glucose-1-phosphate. In muscle, glucose 1-phosphate is used to generate metabolic energy, whereas in liver it is also converted to glucose for export to peripheral tissues. There are three human isozymes of GP: liver, muscle, and brain, named to denote the tissues in which they are preferentially expressed. The muscle and brain isozymes serve the tissues in which they are found, whereas the liver isozyme meets the glycemic demands of the body as a whole. Previous reports have indicated that GP inhibition can lower blood glucose in diabetic models, thus validating it as a potential therapeutic target for treatment of type 2 diabetes. The liver isozyme of human glycogen phosphorylase (HLGP) is considered to be the preferred target for therapeutic intervention with GP inhibitors because inhibition of muscle or brain GP could lead to undesirable side effects.

Type
Chapter
Information
Drug Design
Structure- and Ligand-Based Approaches
, pp. 257 - 264
Publisher: Cambridge University Press
Print publication year: 2010

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Diabetes, World Health Organization. Fact Sheet No. 312, 2006.
Diabetes overview, National Diabetes Statistics. Fact Sheet, 2005.
Talor, S. I.Deconstructing type 2 diabetes. Cell 1999, 97, 9–12.Google Scholar
Treadway, J. L.; Mendys, P.; Hoover, D. J.Glycogen phosphorylase inhibitors for treatment of type 2 diabetes melltus. Exp. Opin. Invest. Drugs 2001, 10, 439–454.Google Scholar
Jakobsen, P.; Lundbeck, J. M.; Kristiansen, M.; Breinholt, J.; Demuth, H.; Pawlas, J.; Torres Candela, M. P.; Anderson, B.; Westergaard, N.; Lundgren, K.; Asano, N.Iminosugars: potential inhibitors of liver glycogen phosphorylase. Bioorg. Med. Chem. 2001, 9, 733–744.Google Scholar
Martin, W. H.; Hoover, D. J.; Armento, S. J.; Stock, I. A.; McPherson, R. K.; Danley, D. E.; Stevenson, R. W.; Barrett, E. J.; Treadway, J. L.Discovery of a human liver glycogen phosphorylase inhibitor that lowers blood glucose in vivo. Proc. Natl. Acad. Sci. U.S.A. 1998, 95, 1776–1781.Google Scholar
Johnson, L. N.Glycogen phosphorylase: control by phosphorylation and allosteric effectors. FASEB J. 1992, 6, 2274–2282.Google Scholar
Rath, V. L.; Ammirati, M.; LeMotte, P. K.; Fennell, K. F.; Mansour, M. N.; Damley, D. E.; Hynes, T. R.; Schulte, G. K.; Wasilko, D. J.Pandit, Activation of human liver glycogen phosphorylase by alteration of the secondary structure and packing of the catalytic core. Mol. Cell 2000, 6, 139–148.Google Scholar
Oikonomakos, N. G.; Tsitsanou, K. E.; Zographos, S. E.; Skamnaki, V. T.; Goldmann, S.; Bischoff, H.Allosteric inhibition of glycogen phosphorylase a by the potential antidiabetic drug 3-isopropyl 4-(2-chlorophenyl)-1,4-dihydro-1-ethyl-2-methyl-pyridine-3,5,6-tricarboxylate. Protein Sci. 1999, 8, 1930–1945.Google Scholar
Zographos, S. E.; Oikonomakos, N. G.; Tsitsanou, K. E.; Leonidas, D. D.; Chrysina, E. D.; Skamnaki, V. T.; Bischoff, H.; Goldmann, S.; Watson, K. A.; Johnson, L. N.The structure of glycogen phosphorylase b with an alkyl-dihydropyridine-dicarboxylic acid compound, a novel and potent inhibitor. Structure 1997, 5, 1413–1425.Google Scholar
Oikonomakos, N. G.; Schnier, J. B.; Zographos, S. E.; Skamnaki, V. T.; Tsitsanou, K. E.; Johnson, L. N.Flavopiridol inhibits glycogen phosphorylase by binding at the inhibitor site. J. Biol. Chem. 2000, 275, 34566–34573.Google Scholar
Hoover, D. J.; Lefkowitz-Snow, S.; Burgess-Henry, J. L.; Martin, W. H.; Armento, S. J.; Stock, I. A.; McPherson, R. K.; Genereux, P. E.; Gibbs, E. M.; Treadway, J. L.Indole-2-carboxamide inhibitors of human liver glycogen phosphorylase. J. Med. Chem. 1998, 41, 2934–2938.Google Scholar
Oikonomakos, N. G.; Skamnaki, V. T.; Tsitsanou, K. E.; Gavalas, N. G.; Johnson, L. N.A New allosteric site in glycogen phosphorylase b as a target for drug interactions. Structure 2000, 8, 575–584.Google Scholar
Rath, V. L.; Ammirati, M.; Danley, D. E.; Ekstrom, J. L.; Gibbs, E. M.; Hynes, T. R.; Mathiowetz, A. M.; McPherson, R. K.; Olson, T. V.; Treadway, J. L.; Hoover, D. J.Human liver glycogen phosphorylase inhibitors bind at a new allosteric site. Chem. Biol. 2000, 7, 677–682.Google Scholar
Oikonomakos, N. G.; Zographos, S. E.; Skamnaki, V. T.; Archontis, G.The 1.76Å resolution crystal structure of glycogen phosphorylase B complexed with glucose, and CP320626, a potential antidiabetic drug. Bioorg. Med. Chem. 2002, 10, 1313–1319.Google Scholar
Miller, M. D.; Sheridan, R. P.; Kearsley, S. K.SQ: a program for rapidly producing pharmacophorically relevent molecular superpositions. J. Med. Chem. 1999, 42, 1505–1514.Google Scholar
ICM-Pro 2.8 docking module. San-Diego: Molsoft LLC.
Miller, M. D.; Kearley, S. K.; Underwood, D. J.; Sheridan, R. P. J.FLOG: a system to select ‘quasi-flexible' ligands complementary to a receptor of known three-dimensional structure. Comput. Aided Mol. Des. 1994, 8, 153–174.Google Scholar
Crippen, C. M.; Havel, T. F.Distance Geometry and Molecular Conformation. New York, NY: Wiley; 1988.
Halgren, T. A.Merck molecular force field. I. Basis, form, scope, parameterization, and performance of MMFF94. J. Comp. Chem. 1996, 17, 490–519.Google Scholar
Halgren, T. A.Merck molecular force field. II. MMFF94 van der Waals and electrostatic parameters for intermolecular interactions. J. Comp. Chem. 1996, 17, 520–552.Google Scholar
Halgren, T. A.Merck molecular force field. III. Molecular geometries and vibrational frequencies for MMFF94. J. Comp. Chem. 1996, 17, 553–586.Google Scholar
Halgren, T. A.; Nachbar, R. B.Merck molecular force field. IV. Conformational energies and geometries for MMFF94. J. Comp. Chem. 1996, 17, 587–615.Google Scholar
Halgren, T. A.Merck molecular force field. V. Extension of MMFF94 using experimental data, additional computational data, and empirical rules. J. Comp. Chem. 1996, 17, 616–641.Google Scholar
Halgren, T. A. MMFF VI. MMFF94s option for energy minimization studiesJ. Comp. Chem. 1999, 20, 720–729.Google Scholar
Halgren, T. A.MMFF VII. Characterization of MMFF94, MMFF94s, and other widely available force fields for conformational energies and for intermolecular-interaction energies and geometries. J. Comp. Chem. 1999, 20, 730–748.Google Scholar
Thompson, J. D.; Higgins, D. G.; Gibson, T. J.CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choice. Nucleic Acids Res. 1994, 22, 4673–4680.Google Scholar
Lu, Z.; Bohn, J.; Bergeron, R.; Deng, Q.; Ellsworth, K. P.; Geissler, W. M.; Harris, G.; McCann, P. E.; McKeever, B.; Myers, R. W.; Saperstein, R.; Willoughby, C. A.; Yao, J.; Chapman, K. A.A New class of glycogen phosphorylase inhibitors. Bioorg. Med. Chem. Lett. 2003, 13, 4125–4128.Google Scholar
Deng, Q.; Lu, Z.; Bohn, J.; Ellsworth, K. P.; Myers, R. W.; Geissler, W. M.; Harris, G.; Willoughby, C. A.; Chapman, K.; Mckeever, B.; Mosley, R.Modeling aided design of potent glycogen phosphorylase inhibitors. J. Mol. Graph. Model. 2005, 23, 457–464.Google Scholar
Kristiansen, M.; Anderson, B.; Iversen, L. F.; Westergaard, N.Identification, synthesis, and characterization of new glycogen phosphorylase inhibitors binding to the allosteric AMP site. J. Med. Chem. 2004, 47, 3537–3545.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×