389
Views
19
CrossRef citations to date
0
Altmetric
Review

A patent review of myeloperoxidase inhibitors for treating chronic inflammatory syndromes (focus on cardiovascular diseases, 2013-2019)

, &
Pages 595-608 | Received 08 Mar 2020, Accepted 05 Jun 2020, Published online: 18 Jun 2020

References

  • Ermert D, Niemiec MJ, Röhm M, et al. Candida albicans escapes from mouse neutrophils. J Leukoc Biol. 2013;94:223–236.
  • Jacobs L, Nawrot TS, de Geus B, et al. Subclinical responses in healthy cyclists briefly exposed to traffic-related air pollution: an intervention study. Environ Health. 2010;9:64.
  • Metzler KD, Fuchs TA, Nauseef WM, et al. Myeloperoxidase is required for neutrophil extracellular trap formation: implications for innate immunity. Blood. 2011;117:953–959.
  • Nauseef WM. Posttranslational processing of a human myeloid lysosomal protein, myeloperoxidase. Blood. 1987;70:1143–1150.
  • Klebanoff SJ. Myeloperoxidase: friend and foe. J Leukoc Biol. 2005;77:598–625.
  • Klebanoff SJ, Kettle AJ, Rosen H, et al. Myeloperoxidase: a front-line defender against phagocytosed microorganisms. J Leukoc Biol. 2013;93(2):185–198.
  • Nauseef WM. Biosynthesis of human myeloperoxidase. Arch Biochem Biophys. 2018;642:1–9.
  • Fiedler TJ, Davey CA, Fenna RE. X-ray crystal structure and characterization of halide-binding sites of human myeloperoxidase at 1.8 A resolution. J Biol Chem. 2000;275:11964–11971.
  • Van Antwerpen P, Slomianny M-C, Boudjeltia KZ, et al. Glycosylation pattern of mature Dimeric Leukocyte and recombinant Monomeric Myeloperoxidase: glycosylation is required for optimal enzymatic activity. J Biol Chem. 2010;285:16351–16359.
  • Furtmüller PG, Burner U, Obinger C. Reaction of Myeloperoxidase compound I with chloride, bromide, iodide, and thiocyanate. Biochemistry. 1998;37:17923–17930.
  • Kettle AJ, Winterbourn CC. The mechanism of myeloperoxidase-dependent chlorination of monochlorodimedon. Biochim Biophys Acta. 1988;957:185–191.
  • Hoogland H, van Kuilenburg A, van Riel C, et al. Spectral properties of myeloperoxidase compounds II and III. Biochim Biophys Acta. 1987;916:76–82.
  • Paumann-Page M, Furtmüller PG, Hofbauer S, et al. Inactivation of human myeloperoxidase by hydrogen peroxide. Arch Biochem Biophys. 2013;539:51–62.
  • Arnhold J, Furtmüller PG, Obinger C. Redox properties of myeloperoxidase. Redox Rep. 2003;8:179–186.
  • Pereira WE, Hoyano Y, Summons RE, et al. Chlorination studies. II. The reaction of aqueous hypochlorous acid with alpha-amino acids and dipeptides. Biochim Biophys Acta. 1973;313:170–180.
  • Ximenes VF, Maghzal GJ, Turner R, et al. Serotonin as a physiological substrate for myeloperoxidase and its superoxide-dependent oxidation to cytotoxic tryptamine-4,5-dione. Biochem J. 2009;425:285–293.
  • Fu X, Kassim SY, Parks WC, et al. Hypochlorous acid generated by myeloperoxidase modifies adjacent tryptophan and glycine residues in the catalytic domain of matrix metalloproteinase-7 (matrilysin): an oxidative mechanism for restraining proteolytic activity during inflammation. J Biol Chem. 2003;278:28403–28409.
  • Prütz WA. Hypochlorous Acid Interactions with Thiols, Nucleotides, DNA, and other biological substrates. Arch Biochem Biophys. 1996;332:110–120.
  • Mongirdienė A, Laukaitienė J, Skipskis V, et al. The effect of oxidant hypochlorous acid on platelet aggregation and dityrosine concentration in chronic heart failure patients and healthy controls. Medicina (Kaunas). 2019;55(5):198.
  • Dennisjr W, Olivieri V, Kruse C. The reaction of nucleotides with aqueous hypochlorous acid. Water Res. 1979;13:357–362.
  • Grotto D, Maria LS, Valentini J, et al. Importance of the lipid peroxidation biomarkers and methodological aspects for malondialdehyde quantification. Quím Nova. 2009;32:169–174.
  • Spickett CM, Jerlich A, Panasenko OM, et al. The reactions of hypochlorous acid, the reactive oxygen species produced by myeloperoxidase, with lipids. Acta Biochim Pol. 2000;47:889–899.
  • Nicholls SJ, Hazen SL. Myeloperoxidase and cardiovascular disease. Arterioscler Thromb Vasc Biol. 2005;25:1102–1111..
  • Green PS, Mendez AJ, Jacob JS, et al. Neuronal expression of myeloperoxidase is increased in Alzheimer’s disease. J Neurochem. 2004;90:724–733.
  • Gellhaar S, Sunnemark D, Eriksson H, et al. Myeloperoxidase-immunoreactive cells are significantly increased in brain areas affected by neurodegeneration in Parkinson’s and Alzheimer’s disease. Cell Tissue Res. 2017;369:445–454.
  • Choi D-K, Pennathur S, Perier C, et al. Ablation of the inflammatory enzyme myeloperoxidase mitigates features of Parkinson’s disease in mice. J Neurosci. 2005;25:6594–6600.
  • Sánchez-López F, Tasset I, Agüera E, et al. Oxidative stress and inflammation biomarkers in the blood of patients with Huntington’s disease. Neurol Res. 2012;34:721–724.
  • Fernandes RMSN, da Silva NP, Sato EI. Increased myeloperoxidase plasma levels in rheumatoid arthritis. Rheumatol Int. 2012;32:1605–1609.
  • Saed GM, Ali-Fehmi R, Jiang ZL, et al. Myeloperoxidase serves as a redox switch that regulates apoptosis in epithelial ovarian cancer. Gynecol Oncol. 2010;116:276–281.
  • Nagra RM, Becher B, Tourtellotte WW, et al. Immunohistochemical and genetic evidence of myeloperoxidase involvement in multiple sclerosis. J Neuroimmunol. 1997;78:97–107.
  • Cascorbi I, Henning S, Brockmöller J, et al. Substantially reduced risk of cancer of the aerodigestive tract in subjects with variant–463A of the myeloperoxidase gene. Cancer Res. 2000;60:644–649.
  • Hung RJ, Boffetta P, Brennan P, et al. Genetic polymorphisms of MPO, COMT, MnSOD, NQO1, interactions with environmental exposures and bladder cancer risk. Carcinogenesis. 2004;25:973–978.
  • Pakakasama S, Chen TT-L, Frawley W, et al. Myeloperoxidase promotor polymorphism and risk of hepatoblastoma. Int J Cancer. 2003;106:205–207.
  • Lee JS, Oh TY, Ahn BO, et al. Involvement of oxidative stress in experimentally induced reflux esophagitis and Barrett’s esophagus: clue for the chemoprevention of esophageal carcinoma by antioxidants. Mutat Res. 2001;480–481:189–200.
  • Yang X, Li Y, Li Y, et al. Oxidative Stress-Mediated Atherosclerosis: mechanisms and Therapies. Front Physiol. 2017;8:600.
  • Fulton DJR, Barman SA. Clarity on the Isoform-specific roles of NADPH oxidases and NADPH oxidase-4 in Atherosclerosis. Arterioscler Thromb Vasc Biol. 2016;36:579–581.
  • Park J-G, Oh GT. The role of peroxidases in the pathogenesis of atherosclerosis. BMB Rep. 2011;44:497–505.
  • Soubhye J, Aldib I, Delporte C, et al. Myeloperoxidase as a target for the treatment of inflammatory syndromes: mechanisms and structure activity relationships of inhibitors. Curr Med Chem. 2016;23:3975–4008.
  • Malle E, Marsche G, Arnhold J, et al. Modification of low-density lipoprotein by myeloperoxidase-derived oxidants and reagent hypochlorous acid. Biochim Biophys Acta. 2006;1761:392–415.
  • Abu-Soud HM, Hazen SL. Nitric oxide modulates the catalytic activity of myeloperoxidase. J Biol Chem. 2000;275:5425–5430.
  • Harrison DG. Cellular and molecular mechanisms of endothelial cell dysfunction. J Clin Invest. 1997;100:2153–2157.
  • Zouaoui Boudjeltia K, Daher J, Van Antwerpen P, et al. Exposure of endothelial cells to physiological levels of myeloperoxidase-modified LDL delays pericellular fibrinolysis. PLoS ONE. 2012;7:e38810.
  • Ndrepepa G. Myeloperoxidase – A bridge linking inflammation and oxidative stress with cardiovascular disease. Clin Chim Acta. 2019;493:36–51.
  • Malle E, Furtmüller PG, Sattler W, et al. Myeloperoxidase: a target for new drug development? Br J Pharmacol. 2007;152:838–854.
  • Reiding KR, Franc V, Huitema MG, et al. Neutrophil myeloperoxidase harbors distinct site-specific peculiarities in its glycosylation. J Biol Chem. 2019;294:20233–20245.
  • Jantschko W, Furtmüller PG, Zederbauer M, et al. Exploitation of the unusual thermodynamic properties of human myeloperoxidase in inhibitor design. Biochem Pharmacol. 2005;69:1149–1157.
  • Soubhye J, Meyer F, Furtmüller P, et al. Characterization of chemical features of potent myeloperoxidase inhibitors. Future Med Chem. 2016;8:1163–1177.
  • Aldib I, Soubhye J, Zouaoui Boudjeltia K, et al. Evaluation of new scaffolds of myeloperoxidase inhibitors by rational design combined with high-throughput virtual screening. J Med Chem. 2012;55:7208–7218.
  • Bensalem S, Soubhye J, Aldib I, et al. Inhibition of myeloperoxidase activity by the alkaloids of Peganum harmala L (Zygophyllaceae). J Ethnopharmacol. 2014;154:361–369.
  • Aldib I, Gelbcke M, Soubhye J, et al. Novel bis-arylalkylamines as myeloperoxidase inhibitors: design, synthesis, and structure-activity relationship study. Eur J Med Chem. 2016;123:746–762.
  • Smallheer J, Kick E, Valente M, et al. Macrocyclic Inhibitors of Myeloperoxidase. WO/2018/005336, (2018).
  • Hair PS, Enos AI, Krishna NK, et al. Inhibition of complement activation, myeloperoxidase, NET formation and oxidant activity by PIC1 peptide variants. Li Y. editor PLoS ONE. 2019;14:e0226875..
  • Hair PS, Sass LA, Krishna NK, et al. Inhibition of myeloperoxidase activity in cystic fibrosis sputum by peptide inhibitor of complement C1 (PIC1). Palaniyar N. editor PLoS ONE. 2017;12:e0170203..
  • Krishna N, Cunnion K PIC1 inhibition of myeloperoxidase oxidative activity in an animal model. WO/2019/139886 (2019).
  • Zhang H, Jing X, Shi Y, et al. N-acetyl lysyltyrosylcysteine amide inhibits myeloperoxidase, a novel tripeptide inhibitor. J Lipid Res. 2013;54:3016–3029.
  • Hao Z, Yang S, Hao X, et al. Peptide-based myeloperoxidase inhibitors for use in therapy. EP2485750B1 (2010).
  • Yu G, Liang Y, Huang Z, et al. Inhibition of myeloperoxidase oxidant production by N-acetyl lysyltyrosylcysteine amide reduces brain damage in a murine model of stroke. J Neuroinflammation. 2016;13:119.
  • Yu G, Zheng S, Zhang H. Inhibition of myeloperoxidase by N-acetyl lysyltyrosylcysteine amide reduces experimental autoimmune encephalomyelitis-induced injury and promotes oligodendrocyte regeneration and neurogenesis in a murine model of progressive multiple sclerosis:. NeuroReport. 2018;29:208–213.
  • van der Does AM, Hensbergen PJ, Bogaards SJ, et al. The human lactoferrin-derived peptide hLF1-11 exerts immunomodulatory effects by specific inhibition of myeloperoxidase activity. J Immunol. 2012;188:5012–5019.
  • Liu C, Desikan R, Ying Z, et al. Effects of a novel pharmacologic inhibitor of myeloperoxidase in a mouse atherosclerosis model. Schmidt HHHW. editor PLoS ONE. 2012;7:e50767.
  • Jayaraj P, Narasimhulu CA, Maiseyeu A, et al. Methoxyphenol derivatives as reversible inhibitors of myeloperoxidase as potential antiatherosclerotic agents. Future Med Chem. 2020;12:95–110.
  • Ximenes VF, Paino IMM, Faria-oliveira de OMM, et al. Indole ring oxidation by activated leukocytes prevents the production of hypochlorous acid. Braz J Med Biol Res. 2005;38:1575–1583.
  • Van Antwerpen P, Zouaoui Boudjeltia K. Rational drug design applied to myeloperoxidase inhibition. Free Radic Res. 2015;49:711–720.
  • Soubhye J, Prévost M, Van Antwerpen P, et al. Structure-based design, synthesis, and pharmacological evaluation of 3-(Aminoalkyl)-5-fluoroindoles as myeloperoxidase inhibitors. J Med Chem. 2010;53:8747–8759.
  • Soubhye J, Dufrasne F, Van Antwerpen P 3-Alkyl-5-fluoroindole derivatives as myeloperoxidase inhibitors. WO2010136546 (2010).
  • Soubhye J, Aldib I, Prévost M, et al. Hybrid molecules inhibiting myeloperoxidase activity and serotonin reuptake: a possible new approach of major depressive disorders with inflammatory syndrome: hybrid MPO and 5-HT reuptake inhibitors. J Pharm Pharmacol. 2014. n/a-n/a. doi:10.1111/jphp.12236
  • Soubhye J, Aldib I, Elfving B, et al. Design, synthesis, and structure–activity relationship studies of novel 3-alkylindole derivatives as selective and highly potent myeloperoxidase inhibitors. J Med Chem. 2013;56:3943–3958.
  • Soubhye J, Dufrasne F, Zouaoui Boudjeltia K, et al. Aromatic N-heterocycle derivatives for use as medicine. EP2682119 (2013).
  • A-K T, Sjögren T, Svensson M, et al. 2-thioxanthines are mechanism-based inactivators of myeloperoxidase that block oxidative stress during inflammation. J Biol Chem. 2011;286:37578–37589.
  • Ward J, Spath SN, Pabst B, et al. Mechanistic characterization of a 2-thioxanthine myeloperoxidase inhibitor and selectivity assessment utilizing click chemistry–activity-based protein profiling. Biochemistry. 2013;52:9187–9201.
  • Piek A, Koonen DPY, Schouten E-M, et al. Pharmacological myeloperoxidase (MPO) inhibition in an obese/hypertensive mouse model attenuates obesity and liver damage, but not cardiac remodeling. Sci Rep. 2019;9(1):18765..
  • Nordvall G, Tiden A, Hanson S Thioxanthine derivatives as myeloperoxidase inhibitors. WO/2003/089430 (2003).
  • Barrett E, Aylor KW, Chai W, et al. Inhibition of myeloperoxidase and MPO reverses microvascular insulin resistance in high-fat diet (HFD)-fed Rats. Diabetes. 2018;67:22–LB.
  • Tong X, Zhou D, Savage A, et al. Population pharmacokinetic modeling with enterohepatic circulation for AZD3241 in healthy subjects and patients with multiple system Atrophy. J Clin Pharmacol. 2018;58:1452–1460.
  • Jucaite A, Svenningsson P, Rinne JO, et al. Effect of the myeloperoxidase inhibitor AZD3241 on microglia: a PET study in Parkinson’s disease. Brain. 2015;138:2687–2700.
  • AZD3241 PET MSA trial, phase 2, randomized,12 week safety and tolerability trial with pet in MSA patients [Internet]. [cited 2017 Sep 25]. Available from: https://clinicaltrials.gov/ct2/show/NCT02388295?term=Myeloperoxidase+inhibitor&draw=2&rank=3.
  • Gan L-M, Lagerström-Fermér M, Ericsson H, et al. Safety, tolerability, pharmacokinetics and effect on serum uric acid of the myeloperoxidase inhibitor AZD4831 in a randomized, placebo-controlled, phase I study in healthy volunteers. Br J Clin Pharmacol. 2019;85:762–770.
  • MPO inhibitor A_Zeneca for HFpEF [Internet]. [cited 2020 Feb 5]. Available from: https://clinicaltrials.gov/ct2/show/NCT03611153.
  • Li Y, Ganesh T, Diebold BA, et al. Thioxo-dihydroquinazolin-one compounds as novel inhibitors of myeloperoxidase. ACS Med Chem Lett. 2015;6:1047–1052.
  • Lambeth JD, Ganesh T, Smith SM, et al. Quinazoline derivatives, compositions, and uses related thereto. US20160243121 (2016).
  • Wurtz NR, Viet A, Shaw SA, et al. Potent triazolopyridine Myeloperoxidase inhibitors. ACS Med Chem Lett. 2018;9:1175–1180.
  • Quoc Viet A, Wurtz NR Thioether triazolopyridine and triazolopyrimidine inhibitors of Myeloperoxidase. US20190031623 (2019).
  • Ruggeri R 2-Thiopyrimidinones. WO/2016/178113 (2016).
  • Ruggeri RB, Buckbinder L, Bagley SW, et al. Discovery of 2-(6-(5-Chloro-2-methoxyphenyl)-4-oxo-2-thioxo-3,4-dihydropyrimidin-1(2 H)-yl)acetamide (PF-06282999): A highly selective mechanism-based myeloperoxidase inhibitor for the treatment of cardiovascular diseases. J Med Chem. 2015;58:8513–8528.
  • Dong JQ, Varma MV, Wolford A, et al. Pharmacokinetics and disposition of the thiouracil derivative PF-06282999, an orally bioavailable, irreversible inactivator of Myeloperoxidase enzyme, across animals and humans. Drug Metab Dispos. 2016;44:209–219.
  • Dong JQ, Gosset JR, Fahmi OA, et al. Examination of the human Cytochrome P4503A4 induction potential of PF-06282999, an irreversible Myeloperoxidase inactivator: integration of Preclinical, in Silico, and biomarker methodologies in the prediction of the clinical outcome. Drug Metab Dispos. 2017;45:501–511.
  • Moscovitz JE, Lin Z, Johnson N, et al. Induction of human cytochrome P450 3A4 by the irreversible myeloperoxidase inactivator PF-06282999 is mediated by the pregnane X receptor. Xenobiotica. 2018;48:647–655.
  • Study A To evaluate the safety and effects on the body of an investigational drug using an endotoxin-induced inflammatory response model (POM) [Internet]. Available from: https://clinicaltrials.gov/ct2/show/NCT01965600?term=PF-06282999&draw=2&rank=3.
  • Myeloperoxidase: AY. Its role for host defense, inflammation, and neutrophil function. Arch Biochem Biophys. 2018;640:47–52.
  • Lazarević-Pasti T, Leskovac A, Vasić V. Myeloperoxidase Inhibitors as Potential Drugs. Curr Drug Metab. 2015;16:168–190.
  • Soubhye J, Chikh Alard I, Aldib I, et al. Discovery of novel potent reversible and irreversible Myeloperoxidase inhibitors using virtual screening procedure. J Med Chem. 2017;60:6563–6586.
  • Hasanpour Z, Javanmard S, Gharaaty M, et al. Association between serum myeloperoxidase levels and coronary artery disease in patients without diabetes, hypertension, obesity, and hyperlipidemia. Adv Biomed Res. 2016;5:103.
  • Teng N, Maghzal GJ, Talib J, et al. The roles of myeloperoxidase in coronary artery disease and its potential implication in plaque rupture. Redox Rep. 2017;22:51–73.
  • Zeng L, Mathew AV, Byun J, et al. Myeloperoxidase-derived oxidants damage artery wall proteins in an animal model of chronic kidney disease–accelerated atherosclerosis. J Biol Chem. 2018;293:7238–7249.
  • Soubhye J, Gelbcke M, Van Antwerpen P, et al. From dynamic combinatorial chemistry to in vivo evaluation of reversible and irreversible Myeloperoxidase inhibitors. ACS Med Chem Lett. 2017;8:206–210.
  • Lemke T, Williams D. Principles of drug discovery. Foye’s principles of medicinal chemistry. 5th ed. ed. Lippincott, Philadelphia: Williams & Wilkins; 2002.
  • Long L, Song Y. Myeloperoxidase: A promising therapeutic target in prevention of atherosclerosis. Int J Cardiol. 2018;256:15.
  • Wang W, Ren D, Wang C, et al. Inhibition of myeloperoxidase might be a novel therapeutic target in the treatment of atherosclerosis. Int J Cardiol. 2018;256:33.
  • Senders ML, Mulder WJM. Targeting myeloperoxidase in inflammatory atherosclerosis. Eur Heart J. 2018;39:3311–3313.
  • Lusis AJ. Atherosclerosis. Nature. 2000;407:233–241.
  • Insull W. The pathology of Atherosclerosis: plaque development and plaque responses to medical treatment. Am J Med. 2009;122:S3–S14.
  • Aviram M, Fuhrman B. LDL oxidation by arterial wall macrophages depends on the oxidative status in the lipoprotein and in the cells: role of prooxidants vs antioxidants. Mol Cell Biochem. 1998;188:149–159.
  • Rifai N. Tietz Textbook of Clinical Chemistry and Molecular Diagnostics. 6th ed. Saunders, USA; 2017.
  • Breese BB. The use of the fluorescent antibody technic for identification of group A streptococci in pediatric practice. Am J Public Health Nations Health. 1968;58:2295–2305.
  • Li X, Fang P, Li Y, et al. Mitochondrial reactive oxygen species mediate Lysophosphatidylcholine-induced Endothelial cell activation. Arterioscler Thromb Vasc Biol. 2016;36:1090–1100
  • Williams KJ, Tabas I. The response-to-retention hypothesis of early Atherogenesis. Arterioscler Thromb Vasc Biol. 1995;15:551–561.
  • Pulli B, Ali M, Forghani R, et al. Measuring Myeloperoxidase activity in biological samples. Johnson R. editor PLoS ONE. 2013;8:e67976..
  • Hosseini M, Rezvani HR, Aroua N, et al. Targeting Myeloperoxidase disrupts mitochondrial redox balance and overcomes Cytarabine resistance in human Acute Myeloid Leukemia. Cancer Res. 2019;79:5191–5203.
  • Wadghiri YZ, Hoang DM, Leporati A, et al. High-resolution imaging of Myeloperoxidase activity sensors in human cerebrovascular disease. Sci Rep. 2018;8:7687.
  • Roth Flach RJ, Su C, Bollinger E, et al. Myeloperoxidase inhibition in mice alters atherosclerotic lesion composition. Bader M. editor PLoS ONE. 2019;14:e0214150.
  • Rashid I, Maghzal GJ, Chen Y-C, et al. Myeloperoxidase is a potential molecular imaging and therapeutic target for the identification and stabilization of high-risk atherosclerotic plaque. Eur Heart J. 2018;39:3301–3310.
  • Kaindlstorfer C, Sommer P, Georgievska B, et al. Failure of neuroprotection despite microglial suppression by delayed-start Myeloperoxidase inhibition in a model of advanced multiple system Atrophy: clinical Implications. Neurotox Res. 2015;28:185–194.
  • AstraZeneca's open innovation pharmacology toolbox. [Internet]. [cited 2014 Nov]. Available from: https://openinnovation.astrazeneca.com/clinical-compound-bank.html.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.