Microbial Metabolites and Enzymes Inhibition in Drug Discovery and Development: A Case Study of the Statins, a Class of HMG-CoA Reductase Inhibitors

doi.org/10.26538/tjnpr/v3i4.1

Authors

  • Akolade R. Oladipupo Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Lagos, Nigeria
  • Herbert A. B. Coker Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Lagos, Nigeria
  • Chinwe S. Alaribe Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Lagos, Nigeria
  • Ogacheko D. Okoko Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Lagos, Nigeria

Keywords:

Drug discovery, Statins, Microbial metabolites, HMG-CoA reductase inhibitors Cholesterol-lowering agents,, Akira Endo.

Abstract

The discovery and development of the statins provides an insight into the many facets of the drug discovery process and also shows the time and resources that must be devoted to developing a clinically useful drug from an initial concept. It began from the conceptual search for metabolites that could inhibit an important enzyme in cholesterol biosynthesis, HMG-CoA (3-hydroxy-3-methylglutaryl-CoA) reductase, to the isolation of the first active metabolite citrinin, from a fungus, leading to further search and development. This discovery is a milestone in medicine and a historical portrait, which illustrates the importance of multidisciplinary approach in drug development. The statins are the results of combined efforts of inter alia pharmaceutical chemists, microbiologists, physiologists, and pharmacologists. This paper gives an account of the discovery of the statins, highlighting the clinical importance of cholesterol, the discovery of HMG-CoA reductase, and subsequent search for its inhibitors. It showcases the clinical importance and limitations of the statins and seeks to serve as a guide and inspiration for future discovery and development of novel, potent and safer cholesterol-lowering agents.

References

Maycock CAA, Muhlestein JB, Horne BD, Carlquist JF, Bair TL, Pearson RR, Li Q, Anderson JL. Statin Therapy Is Associated With Reduced Mortality Across All Age Groups of Individuals With Significant Coronary Disease, Including Very Elderly Patients. J Am Coll Cardiol. 2002; 40(10):1777-1785.

World Health Organization (WHO). Cardiovascular Diseases (CVDs). 2017; [cited 2018 July 2]. Available from: http://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds)

Malik VS. Microbial secondary metabolism. Trends Biochem Sci. 1980; 5(3):68-72.

Mousa WK and Raizada MN. The diversity of anti-microbial secondary metabolites produced by fungal endophytes: an interdisciplinary perspective. Front Microbiol. 2013; 4:65.

Ludovici LJ. Fleming. Discoverer of Penicillin. London: Andrew Dankers Limited; 1952.

Waksman SA. Living with the Microbes. New York: Simon & Schuster, Inc; 1954.

Ramsay RR and Tipton KF. Assessment of Enzyme Inhibition: A Review with Examples from the Development of Monoamine Oxidase and Cholinesterase Inhibitory Drugs. Molecules 2017; 22(1192):237.

Hopkins AL and Groom CR. The druggable genome. Nat Rev Drug Discov. 2002; 1:727–730.

Rajagopalan PTR, Zhang Z, McCourt L, Dwyer M, Benkovic SJ, Hammes, GG. Interaction of dihydrofolate reductase with methotrexate: Ensemble and single-molecule kinetics. Proc Natl Acad Sci USA. 2002; 99(21):13481–13486.

Goodsell DS. The Molecular Perspective: Methotrexate. Oncologist 1999; 4(4):340–341.

Béthune MP. Non-nucleoside reverse transcriptase inhibitors (NNRTIs), their discovery, development, and use in the treatment of HIV-1 infection: a review of the last 20 years (1989-2009). Antiviral Res. 2010; 85(1):75–90.

Schauer GD, Huber KD, Leuba SH, Sluis-Cremer N. Mechanism of allosteric inhibition of HIV-1 reverse transcriptase revealed by

single-molecule and ensemble fluorescence. Nucleic Acids Res 2014; 42(18):11687–11696.

Jokanović M and Stojiljković MP. Current understanding of the application of pyridinium oximes as cholinesterase reactivators in treatment of organophosphate poisoning. Eur J Pharmacol. 2006; 553:10–17.

Ferreira LG, dos Santos RN, Oliva G, Andricopulo AD. Molecular Docking and Structure-Based Drug Design Strategies. Molecules 2015; 20:1334-1342.

Mendis S, Puska P, Norrving B (Eds). Global Atlas on Cardiovascular Disease Prevention and Control. Geneva: World Health Organization; 2011.

Dam H. Historical introduction to cholesterol. In: Cook RP, editor. Chemistry, Biochemistry and Pathology. New York: Academic Press; 1958. 1–14 p.

Erling N. Nobel Prizes And Notable Discoveries. Singapore: World Scientific Publishing; 2016. 327 p.

Chevreul ME. Recherches chimiques sur plusieurs corps gras, et particulie`rement sur leurs combinations avec les alcalis. Cinquieme Memoire. Des corps qu’on a appele´s adipocire, c’est-a`-dire, de la substance cristallise´e des calculs biliaires humains, du spermace´ti et de la substance grasse des cadavres [Chemical research on several fatty substances, and particularly on their combinations with alkalis. Fifth Memoir. Bodies that have been called adipocere, that is the crystallized substance of human gallstones, spermaceti, and the fatty substance of corpses]. Ann Chim. 1815; 95:5–50.

Brown MS, Goldstein JL. A receptor rmediated pathway for cholesterol homeostasis. Sci. 1986; 232:34–47.

Hanukoglu I. Steroidogenic enzymes: structure, function, and role in regulation of steroid hormone biosynthesis. J Steroid Biochem Mol Biol. 1992; 43(8):779–804.

Payne AH and Hales DB. Overview of steroidogenic enzymes in the pathway from cholesterol to active steroid hormones. Endocr Rev. 2004; 25(6):947–970.

Goldstein JL and Brown HS. Cholesterol: a century of research. HHMI Bull. 2003; 16(3):10–19.

Anitschkow N. Über die Veränderungen der Kaninchenaorta bei experimenteller Cholesterinsteatose [About changes in rabbit aorta in experimental cholesterol steatosis]. Beitr Pathol Anat. 1913; 56:379–404.

Gofman JW, Lindgren FT, Elliott H, Manz W, Hewitt J, Herring V. The role of lipids and lipoproteins in atherosclerosis. Sci. 1950; 111:166–171.

Keys A, Anderson JT, Fidanza F, Keys MH, Swahn B. Effects of diet on blood lipids in man, particularly cholesterol and lipoproteins. Clin Chem. 1955; 1:34–52.

Kannel WB, Dawber TR, Kagan A, Revotskie N, Stokes J. Factors of risk in the development of coronary heart disease—six year follow-up experience: The Framingham Study. Ann Intern Med. 1961; 55:33–50.

Wilson PW, Garrison RJ, Castelli WP, Feinleib M, McNamara PM, Kannel WB. Prevalence of coronary heart disease in the Framingham Offspring Study: role of lipoprotein cholesterols. Am J Cardiol. 1980; 46:649–654.

Bloch K..The biological synthesis of cholesterol. Sci. 1965; 150:19–28.

Lynen F. The biochemical basis of the biosynthesis of cholesterol and fatty acids. Wien Klin Wochenschr 1966; 78:489–497.

Bucher NL, Overath P, Lynen F. Hydroxy---methylglutaryl coenzyme A reductase, cleavage and condensing enzymes in relation to cholesterol formation in rat liver. Biochim Biophys Acta 1960; 40:491–501.

Siperstein MD. Regulation of cholesterol biosynthesis in normal and malignant tissues. Curr. Topics Cell Reg 1970; 2:65-100.

Endo A. A historical perspective on the discovery of statins. Proc Jpn Acad Ser B. 2010; 86:484-493.

Blohm TR and MacKenzie RD. Specific inhibition of cholesterol biosynthesis by a synthetic compound (MER-29). Arch Biochem Biophys. 1959; 85:245–249.

Altschul R, Hoffer A, Stephen JD. Influence of nicotinic acid on serum cholesterol in man. Arch Biochem. 1955; 54:558–559.

Thorp JM and Warig WS. Modification of Metabolism and distribution of lipids by ethyl chlorophenoxyisobutyrate. Nature 1962; 194:948–49.

Endo A. The discovery and development of HMG-CoA reductase inhibitors. J Lipid Res 1992; 33:1569–82.

Endo A. A gift from nature: the birth of the statins. Nat Med. 2008; 14:1050–1052.

Endo A and Kuroda M. Citrinin, an inhibitor of cholesterol synthesis. J Antibiot (Japan) 1976; 29:841–843.

Tanzawa K, Kuroda M, Endo A. Time-dependent, irreversible inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A reductase by the antibiotic citrinin. Biochim Biophys Acta 1977; 488:97–101.

Endo A, Tsujita T, Kuroda M, Tanzawa K. Inhibition of cholesterol synthesis in vitro and in vivo by ML-236A and ML-236B, competitive inhibitors of 3-hydroxy-3-methylglutarylcoenzyme A reductase. Eur J Biochem. 1977; 77:31–36.

Endo A, Kuroda M, Tanzawa K. Competitive inhibition of 3-hydroxy-3-methylglutaryl coenzyme A reductase by ML-236A and ML 236B fungal metabolites, having hypocholesterolemic activity. FEBS Lett. 1976; 72:323–326.

Tsujita Y, Kuroda M, Tanzawa K, Kitano N, Endo A. Hypolipidemic effects in dogs of ML-236B, a competitive inhibitor of 3-hydroxy-3-methylglutaryl-coenzyme A reductase. Atheroscler. 1979; 32:307–313.

Kuroda M, Tsujita Y, Tanzawa K, Endo A. Hypolipidemic effects in monkeys of ML-236B, a competitive inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A reductase. Lipids 1979; 14:585–589.

Brown AG, Smale TC, King TJ, Hasenkamp R, Thompson RH. Crystal and molecular structure of compactin, a new antifungal metabolite from Penicillium brevicompactum. J Chem Soc, Perkin Trans. 1976; 1:1165–1170.

Fears RB, Richards DH, Ferres H. The effect of compactin, a potent inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A reductase activity, on cholesterogenesis and serum cholesterol levels in rats and chicks. Atheroscler. 1980; 35:439–449.

Alberts AW, Chen J, Kuron G, Hunt V, Huff J, Hoffman C, Rothrock J, Lopez M, Joshua H, Harris E, Patchett A, Monaghan R, Currie S, Stapley E, Albers-Schonbergy, Hensens G, Hirshfieldt J, Hoogsteent K, Liescht J, Springer J. Mevinolin: a highly potent competitive inhibitor of hydroxymethylglutaryl-coenzyme A reductase and a cholesterol-lowering agent. Proc Natl Acad Sci USA. 1980; 77:3957–3961.

Endo A. Monacolin K, a new hypocholesterolemic agent produced by a Monascus species. J Antibiot. 1979; 32:852–854.

Vagelos PR and Galambos L. Medicine, Science and Merck. Cambridge: Cambridge University Press; 2004. 1–301 p.

Brown MS, Dana SE, Goldstein JL. Regulation of 3-hydroxy-3-methylglutaryl coenzyme A reductase activity in cultured human fibroblasts. Comparison of cells from a normal subject and from a patient with homozygous familial hypercholesterolemia. J Biol Chem. 1974; 249:789–796.

Kishida Y, Naito A, Iwado S, Terahara A , Tsujita Y. Research and Development of Pravastatin. Yakugaku Zasshi 1991; 111(9):469-487.

Williams O, Jacks A, Davis J, Martinez S. Case 10: Merck (A): Mevacor. In Allan Afuah editor. Innovation Management - Strategies, Implementation, and Profits. Oxford: Oxford University Press; 1998.

Fong CW. Statins in therapy: Understanding their hydrophilicity, lipophilicity, binding to 3-hydroxy-3-methylglutaryl-CoA reductase, ability to cross the blood brain barrier and metabolic stability based on electrostatic molecular orbital studies. Eur J Med Chem. 2014; 85:661-674.

Istvan ES and Deisenhofer J. Structural Mechanism for Statin Inhibition of HMG-CoA Reductase. Sci. 2001; 292(5519):1160–164.

McTaggart F. Comparative pharmacology of rosuvastatin. Atheroscler Suppl. 2003; 4(1):9–14.

Schachter M. Chemical, pharmacokinetic and pharmacodynamic properties of statins: an update. Fund Clin Pharmacol. 2004; 19:117–25.

Simon J. The $10 billion pill. Fortune 2003; 147(1):58-62, 66-68.

Pfizer. Doing Things Differently: Pfizer 2008 Annual Review. New York: Pfizer Inc; 2009. 15 p.

Roche VF. Teachers' Topics: Antihyperlipidemic Statins: A Self-Contained, Clinically Relevant Medicinal Chemistry Lesson. Am J Pharm Educ. 2005; 69(4):546–560.

Black DM. A general assessment of the safety of HMG CoA reductase inhibitors (statins). Curr Atheroscler Rep. 2002; 4:34–41.

Bang CN and Okin PM. Statin treatment, new-onset diabetes, and other adverse effects: a systematic review. Curr Cardiol Rep. 2014; 16(3):461.

Manji H. Drug-induced neuropathies. Handb Clin Neurol. 2013; 115:729-742.

Bełtowski J, Wójcicka G, Jamroz-Wiśniewska A. Adverse effects of statins—mechanisms and consequences. Curr Drug Saf. 2009; 4:209-228.

Harper CR and Jacobson TA. The broad spectrum of statin myopathy: from myalgia to rhabdomyolysis. Curr Opin Lipidol. 2007; 18(4):401-408.

Tuccori M, Lapi F, Testi A. Statin-associated psychiatric adverse events: a case/non-case evaluation of an Italian database of spontaneous adverse drug reaction reporting. Curr Drug Saf. 2008; 31:1115-1123.

Davignon J. Beneficial cardiovascular pleiotropic effects of statins. Circul. 2004; 109:39–43.

Blum A and Shamburek R. The pleiotropic effects of statins on endothelial function, vascular inflammation, immunomodulation and thrombogenesis. Atheroscler. 2009; 203(2):325–330.

Wolozin B, Wang SW, Li NC, Lee A, Lee TA, Kazis LE. Simvastatin is associated with a reduced incidence of dementia and Parkinson's disease. BMC Med. 2007; 5(1):20.

Khurana V, Bejjanki HR, Caldito G, Owens MW. Statins reduce the risk of lung cancer in humans: a large case-control study of US veterans. Chest 2007; 131(5):1282–1288.

Klein BE, Klein R, Lee KE, Grady LM. Statin use and incident nuclear cataract. JAMA 2006; 295(23):2752–2758.

Drapala A, Sikora M, Ufnal M. Statins, the renin-angiotensin-aldosterone system and hypertension - a tale of another beneficial effect of statins. J Renin Angiotensin Aldosterone Syst. 2014; 15(3):250–258.

Mondul AM, Han M, Humphreys EB, Meinhold CL, Walsh PC, Platz EA. Association of statin use with pathological tumor characteristics and prostate cancer recurrence after surgery. J Urol. 2011; 185(4):1268–1273.

Steinberg D. An interpretive history of the cholesterol controversy, part V. The discovery of the statins and the end of the controversy. J Lipid Res. 2006; 47:1339–351.

Thompson GR. Lasker award for the discoverer of statins. Br J Cardiol. 2008; 15:294–295.

National Inventors Hall of Fame (NIHF). Akira Endo. [cited 2018 Dec 10]. Available from: http://www.invent.org/inductees/akira-endo

Downloads

Published

2019-04-01

How to Cite

R. Oladipupo, A., A. B. Coker, H., S. Alaribe, C., & D. Okoko, O. (2019). Microbial Metabolites and Enzymes Inhibition in Drug Discovery and Development: A Case Study of the Statins, a Class of HMG-CoA Reductase Inhibitors: doi.org/10.26538/tjnpr/v3i4.1. Tropical Journal of Natural Product Research (TJNPR), 3(4), 99–106. Retrieved from https://www.tjnpr.org/index.php/home/article/view/936