Endothelial-protective effects of a G-protein-biased sphingosine-1 phosphate receptor-1 agonist, SAR247799, in type-2 diabetes rats and a randomized placebo-controlled patient trial

Luc Bergougnan, Grit Andersen, Leona Plum-Mörschel, Maria Francesca Evaristi, Bruno Poirier, Agnes Tardat, Marcel Ermer, Theresa Herbrand, Jorge Arrubla, Hans Veit Coester, Roberto Sansone, Christian Heiss, Olivier Vitse, Fabrice Hurbin, Rania Boiron, Xavier Benain, David Radzik, Philip Janiak, Anthony J Muslin, Lionel Hovsepian, Stephane Kirkesseli, Paul Deutsch, Ashfaq A Parkar, Luc Bergougnan, Grit Andersen, Leona Plum-Mörschel, Maria Francesca Evaristi, Bruno Poirier, Agnes Tardat, Marcel Ermer, Theresa Herbrand, Jorge Arrubla, Hans Veit Coester, Roberto Sansone, Christian Heiss, Olivier Vitse, Fabrice Hurbin, Rania Boiron, Xavier Benain, David Radzik, Philip Janiak, Anthony J Muslin, Lionel Hovsepian, Stephane Kirkesseli, Paul Deutsch, Ashfaq A Parkar

Abstract

Aims: SAR247799 is a G-protein-biased sphingosine-1 phosphate receptor-1 (S1P1 ) agonist designed to activate endothelial S1P1 and provide endothelial-protective properties, while limiting S1P1 desensitization and consequent lymphocyte-count reduction associated with higher doses. The aim was to show whether S1P1 activation can promote endothelial effects in patients and, if so, select SAR247799 doses for further clinical investigation.

Methods: Type-2 diabetes patients, enriched for endothelial dysfunction (flow-mediated dilation, FMD <7%; n = 54), were randomized, in 2 sequential cohorts, to 28-day once-daily treatment with SAR247799 (1 or 5 mg in ascending cohorts), placebo or 50 mg sildenafil (positive control) in a 5:2:2 ratio per cohort. Endothelial function was assessed by brachial artery FMD. Renal function, biomarkers and lymphocytes were measured following 5-week SAR247799 treatment (3 doses) to Zucker diabetic fatty rats and the data used to select the doses for human testing.

Results: The maximum FMD change from baseline vs placebo for all treatments was reached on day 35; mean differences vs placebo were 0.60% (95% confidence interval [CI] -0.34 to 1.53%; P = .203) for 1 mg SAR247799, 1.07% (95% CI 0.13 to 2.01%; P = .026) for 5 mg SAR247799 and 0.88% (95% CI -0.15 to 1.91%; P = .093) for 50 mg sildenafil. Both doses of SAR247799 were well tolerated, did not affect blood pressure, and were associated with minimal-to-no lymphocyte reduction and small-to-moderate heart rate decrease.

Conclusion: These data provide the first human evidence suggesting endothelial-protective properties of S1P1 activation, with SAR247799 being as effective as the clinical benchmark, sildenafil. Further clinical testing of SAR247799, at sub-lymphocyte-reducing doses (≤5 mg), is warranted in vascular diseases associated with endothelial dysfunction.

Trial registration: ClinicalTrials.gov NCT03462017.

Keywords: SAR247799; diabetes; endothelium; flow-mediated dilation; sphingosine-1 phosphate receptor-1.

Conflict of interest statement

At the time of conduct of the studies, L.B., M.F.E., B.P., A.T., O.V., F.H., R.B., X.B., D.R., P.J., A.J.M., L.H., S.K., P.D. and A.A.P. were employees of Sanofi; G.A., L.P., M.E., T.H., J.A. and H.C. were employees of Profil Institute; R.S. and C.H. received personal fees from Profil Institute. Authors affiliated with Sanofi may have equity interest in Sanofi. A.A.P., B.P. and P.J. are inventors of US patent number 9 782 411. C.H. received grants from Deutsche Forschungsgemeinschaft, Mitsubishi Cleansui, Wild Blueberry Association of North America, University of Surrey, Philips, National Processed Raspberry Council, Cranberry Institute and Rheacell, and personal fees from Bayer and Novo Nordisk, all outside of the submitted work.

© 2020 The Authors. British Journal of Clinical Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.

Figures

FIGURE 1
FIGURE 1
Effects of SAR247799 on lymphocytes, heart rate and renal parameters in diabetic rats. Zucker diabetic fatty (ZDF) rats treated for 5 weeks with chow formulated with control (0), low (L; 0.002% w/w), intermediate (I; 0.007% w/w) or high (H; 0.0245% w/w) doses of SAR247799. Age‐matched lean rats treated with control chow (0) for comparison. n = 8, 12, 14, 12 rats in lean, ZDF 0, ZDF L, ZDF I, ZDF H groups, respectively. Bars are mean ± standard error of the mean when Shapiro–Wilks test did not reject normality hypothesis (A, B) and median ± interquartile range otherwise (C–G). #P < .05, ##P < .01, ###P < .001 for lean vs ZDF control group; A and B by Student t test; E, F, G by Wilcoxon test; C and D baseline comparisons of lean vs the 4 ZDF groups by Kruskal–Wallis followed by Wilcoxon posthoc test; C and D treatment comparisons of lean vs ZDF control group by Wilcoxon test. * P < .05, ** P < .01, *** P < .001 for treated groups vs ZDF control group; A and B by 1‐way ANOVA followed by Dunnett's test; E, F, G and C & D (treatment) by Kruskal–Wallis test followed by Wilcoxon posthoc test
FIGURE 2
FIGURE 2
Effects of SAR247799 on plasma biomarkers in diabetic rats. Zucker diabetic fatty (ZDF) rats following 5‐week treatment with chow formulated with control (0), low (L; 0.002% w/w), intermediate (I; 0.007% w/w), or high (H; 0.0245% w/w) doses of SAR247799. Age‐matched lean rats treated with control chow (0) for comparison. n = 8, 12, 14, 12 rats in lean, ZDF 0, ZDF L, ZDF I, ZDF H groups, respectively. Bars represent mean ± standard error of the mean when Shapiro–Wilks test did not reject normality hypothesis (C) and median ± interquartile range otherwise (A, B, D, E, F, G). ##P < .01, ###P < .001 for lean vs ZDF control group; Student t‐test for C, or Wilcoxon test for A, B, D, E, F, G. * P < .05, ** P < .01, for treated groups vs ZDF control group; Kruskal–Wallis test followed by Wilcoxon posthoc test for A, B, D, E, F, G
FIGURE 3
FIGURE 3
Design and patient (pt) progress through randomized, double‐blinded clinical trial to evaluate SAR247799 on flow‐mediated dilation in type‐2 diabetes patients. (A) Study design. (B) Flow chart of patient progress through trial. of the 122 patients not meeting inclusion criteria, 14 were because flow‐mediated dilation (FMD) was >7%. # 1 patient excluded for unusable FMD data and 1 patient withdrawn. DSM, dose selection meeting; PK, pharmacokinetics; B, baseline; EOS, end of study; BAD, brachial artery diameter; HR, heart rate; BP, blood pressure; PK, pharmacokinetics
FIGURE 4
FIGURE 4
Effect of SAR247799 (1 and 5 mg), sildenafil (50 mg), or placebo on heart rate (A) and lymphocytes (B) in type‐2 diabetes patients. Heart rate and lymphocytes expressed on each day (D1–D28) at time points associated with maximal pharmacodynamic effects (4 and 6.5 h, respectively). Mean ± standard error of the mean; P values shown when < .05 (repeated‐measure model as described under Methods)
FIGURE 5
FIGURE 5
Effect of SAR247799 (1 and 5 mg), sildenafil (50 mg), or placebo on % flow‐mediated dilation (FMD) in type‐2 diabetes patients. FMD data expressed as change from baseline (A), or change from baseline, placebo corrected (B). Bars are mean ± standard error of the mean. Variation in the placebo groups in B (placebo‐corrected FMD change) illustrated by retaining error bars with mean zero (first group at each time point). Precise P‐values for difference vs placebo shown when P < .05; repeated‐measure model as described under Methods. Number of patients with FMD data in each histogram was as follows for placebo/1 mg SAR247799/5 mg SAR247799/50 mg sildenafil, respectively; 12/15/15/10 (D14, D21, D28 and D42) and 12/15/13/10 (D35)

References

    1. Rajendran P, Rengarajan T, Thangavel J, et al. The vascular endothelium and human diseases. Int J Biol Sci. 2013;9(10):1057‐1069.
    1. Taqueti VR, Di Carli MF. Coronary microvascular disease pathogenic mechanisms and therapeutic options: JACC state‐of‐the‐art review. J Am Coll Cardiol. 2018;72(21):2625‐2641.
    1. Marzilli M, Merz CN, Boden WE, et al. Obstructive coronary atherosclerosis and ischemic heart disease: an elusive link! J Am Coll Cardiol. 2012;60(11):951‐956.
    1. van Sloten TT, Henry RM, Dekker JM, et al. Endothelial dysfunction plays a key role in increasing cardiovascular risk in type 2 diabetes: the Hoorn study. Hypertension. 2014;64(6):1299‐1305.
    1. Thuy AV, Reimann CM, Hemdan NY, Gräler MH. Sphingosine 1‐phosphate in blood: function, metabolism, and fate. Cell Physiol Biochem. 2014;34(1):158‐171.
    1. Jozefczuk E, Guzik TJ, Siedlinski M. Significance of sphingosine‐1‐phosphate in cardiovascular physiology and pathology. Pharmacol Res. 2020;156:104793. 10.1016/j.phrs.2020.104793
    1. Cartier A, Hla T. Sphingosine 1‐phosphate: lipid signaling in pathology and therapy. Science. 2019;366:eaar5551. 10.1126/science.aar5551
    1. Guitton J, Bandet CL, Mariko ML, et al. Sphingosine‐1‐phosphate metabolism in the regulation of obesity/type 2 diabetes. Cell. 2020;9:1682. 10.3390/cells9071682
    1. Ng ML, Wadham C, Sukocheva OA. The role of sphingolipid signalling in diabetes‐associated pathologies. Int J Mol Med. 2017;39(2):243‐252.
    1. Raza Z, Saleem U, Naureen Z. Sphingosine 1‐phosphate signaling in ischemia and reperfusion injury. Prostaglandins Other Lipid Mediat. 2020;149:106436. 10.1016/j.prostaglandins.2020.106436
    1. Intapad S. Sphingosine‐1‐phosphate signaling in blood pressure regulation. Am J Physiol Renal Physiol. 2019;317(3):F638‐F640.
    1. Cantalupo A, di Lorenzo A. S1P signaling and De novo biosynthesis in blood pressure homeostasis. J Pharmacol Exp Ther. 2016;358(2):359‐370.
    1. Okajima F. Plasma lipoproteins behave as carriers of extracellular sphingosine 1‐phosphate: is this an atherogenic mediator or an anti‐atherogenic mediator? Biochim Biophys Acta. 2002;1582(1‐3):132‐137.
    1. Frej C, Mendez AJ, Ruiz M, et al. A shift in ApoM/S1P between HDL‐particles in women with type 1 diabetes mellitus is associated with impaired anti‐inflammatory effects of the ApoM/S1P complex. Arterioscler Thromb Vasc Biol. 2017;37(6):1194‐1205.
    1. Diarte‐Añazco EMG, Méndez‐Lara KA, Pérez A, Alonso N, Blanco‐Vaca F, Julve J. Novel insights into the role of HDL‐associated Sphingosine‐1‐phosphate in Cardiometabolic diseases. Int J Mol Sci. 2019;20:6273. 10.3390/ijms20246273
    1. Vaisar T, Couzens E, Hwang A, et al. Type 2 diabetes is associated with loss of HDL endothelium protective functions. PLoS One. 2018. Mar 15:e0192616. 10.1371/journal.pone.0192616 eCollection 2018
    1. Brinck JW, Thomas A, Lauer E, et al. Associated with reduced high‐density lipoprotein Sphingosine‐1‐phosphate content and impaired high‐density lipoprotein cardiac cell protection. Arterioscler Thromb Vasc Biol. 2016;36:817‐824.
    1. Sattler KJE, Elbasan Ş, Keul P, et al. Sphingosine 1‐phosphate levels in plasma and HDL are altered in coronary artery disease. Basic Res Cardiol. 2010;105:821‐832.
    1. Argraves K, Sethi A, Gazzolo P, et al. S1P, dihydro‐S1P and C24:1‐ceramide levels in the HDL‐containing fraction of serum inversely correlate with occurrence of ischemic heart disease. Lipids Health Dis. 2011. May 9;10(1):1‐12. 10.1186/1476-511X-10-70
    1. Knapp M, Baranowski M, Czarnowski D, et al. Plasma sphingosine‐1‐phosphate concentration is reduced in patients with myocardial infarction. Med Sci Monit. 2009;15(9):CR490‐CR493.
    1. Knapp M, Lisowska A, Zabielski P, Musiał W, Baranowski M. Sustained decrease in plasma sphingosine‐1‐phosphate concentration and its accumulation in blood cells in acute myocardial infarction. Prostaglandins Other Lipid Mediat. 2013;106:53‐61.
    1. Denimal D, Monier S, Brindisi MC, et al. Impairment of the ability of HDL from patients with metabolic syndrome but without diabetes mellitus to activate eNOS: correction by S1P enrichment. Arterioscler Thromb Vasc Biol. 2017;37:804‐811.
    1. Coldewey SM, Benetti E, Collino M, et al. Elevation of serum sphingosine‐1‐phosphate attenuates impaired cardiac function in experimental sepsis. Sci Rep. 2016;6:27594. 10.1038/srep27594
    1. Winkler MS, Märtz KB, Nierhaus A, et al. Loss of sphingosine 1‐phosphate (S1P) in septic shock is predominantly caused by decreased levels of high‐density lipoproteins (HDL). J Intensive Care. 2019;7(1):1‐9. 10.1186/s40560-019-0376-2
    1. Jing XD, Wei XM, Deng SB, Du JL, Liu YJ, She Q. The relationship between the high‐density lipoprotein (HDL)‐associated sphingosine‐1‐phosphate (S1P) and coronary in‐stent restenosis. Clin Chim Acta. 2015;446:248‐252.
    1. Soltau I, Mudersbach E, Geissen M, et al. Serum‐Sphingosine‐1‐phosphate concentrations are inversely associated with atherosclerotic diseases in humans. PLoS One. 2016. Dec 14;e0168302. 10.1371/journal.pone.0168302 eCollection 2016
    1. Kiziltunc E, Abaci A, Ozkan S, et al. The relationship between pre‐infarction angina and serum Sphingosine‐1‐phosphate levels. Acta Cardiol Sin. 2014;30(6):546‐552.
    1. Kızıltunç E, Gök M, Topçuoğlu C, et al. Serum sphingosine 1 phosphate levels in patients with and without coronary collateral circulation. Acta Cardiol Sin. 2018;34(5):379‐385.
    1. Rosenson RS, Brewer HB Jr, Ansell BJ, et al. Dysfunctional HDL and atherosclerotic cardiovascular disease. Nat Rev Cardiol. 2016;13(1):48‐60.
    1. Armitage J, Holmes MV, Preiss D. Cholesteryl Ester transfer protein inhibition for preventing cardiovascular events: JACC review topic of the week. J Am Coll Cardiol. 2019;73(4):477‐487.
    1. Boden WE, Probstfield JL, Anderson T, et al. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med. 2011;365(24):2255‐2267.
    1. Galvani S, Hla T. Quality versus quantity: making HDL great again. Arterioscler Thromb Vasc Biol. 2017;37:1018‐1019.
    1. Egom EE, Mamas MA, Soran H. HDL quality or cholesterol cargo: what really matters – spotlight on sphingosine‐1‐phophate‐rich HDL. Curr Op Lipidol. 2013;24(4):351‐356.
    1. Kurano M, Yatomi Y. Sphingosine‐1‐phosphate and atherosclerosis. J Atheroscler Thromb. 2018;25(1):16‐26.
    1. Argraves KM, Gazzolo PJ, Groh EM, et al. High density lipoprotein‐associated sphingosine‐1 phosphate promotes endothelial barrier function. J Biol Chem. 2008;283(36):25074‐25081.
    1. Potì F, Simoni M, Nofer J‐R. Atheroprotective role of high‐density lipoprotein (HDL)‐associated sphingosine‐1‐phosphate (S1P). Cardiovasc Res. 2014;103(3):395‐404.
    1. Ruiz M, Frej C, Holmér A, Guo LJ, Tran S, Dahlbäck B. High‐density lipoprotein‐associated Apolipoprotein M limits endothelial inflammation by delivering Sphingosine‐1‐phosphate to the Sphingosine‐1‐phosphate receptor 1. Arterioscler Thromb Vasc Biol. 2017;37:118‐129.
    1. Gong M, Wilson M, Kelly T, et al. HDL‐associated estradiol stimulates endothelial NO synthase and vasodilation in an SR‐BI‐dependent manner. J Clin Invest. 2003;111(1587):1579‐1587.
    1. Sukocheva O, Wadham C, Gamble J, Xia P. Sphingosine‐1‐phosphate receptor 1 transmits estrogens' effects in endothelial cells. Steroids. 2015;104:237‐245.
    1. Guo S, Yu Y, Zhang N, et al. Higher level of plasma bioactive molecule sphingosine 1‐phosphate in women is associated with estrogen. Biochim Biophys Acta. 2014. Jun;1841(6):836‐846.
    1. Sattler JKE. Defects of high‐density lipoproteins in coronary artery disease caused by low Sphingosine‐1‐phosphate content. Correction by Sphingosine‐1‐phosphate loading. J Am Coll Cardiol. 2015;66:1470‐1485.
    1. Christensen PM, Liu CH, Swendeman SL, et al. Impaired endothelial barrier function in apolipoprotein M–deficient mice is dependent on sphingosine‐1‐phosphate receptor 1. FASEB J. 2016;30(6):2351‐2359.
    1. Sanchez T. Sphingosine‐1‐phosphate signaling in endothelial disorders. Curr Atheroscler Rep. 2016. 18:31;18(6):1‐13. 10.1007/s11883-016-0586-1
    1. Kappos L, Radue EW, O'Connor P, et al. FREEDOMS study group. A placebo‐controlled trial of oral fingolimod in relapsing multiple sclerosis. N Engl J Med. 2010;362(5):387‐401.
    1. Kappos L, Bar‐Or A, Cree BAC, et al. Siponimod versus placebo in secondary progressive multiple sclerosis (EXPAND): a double‐blind, randomised, phase 3 study. Lancet. 2018;391(10127):1263‐1273.
    1. Hoch M, D'Ambrosio D, Wilbraham D, Brossard P, Dingemanse J. Clinical pharmacology of ponesimod, a selective S1P1 receptor modulator, after uptitration to supratherapeutic doses in healthy subjects. Eur J Pharm Sci. 2014;63:147‐153.
    1. Jain N, Bhatti T. Fingolimod‐associated macular edema: incidence, detection and management. Neurology. 2012;78:672‐680.
    1. Salvadori ML, Budde K, Charpentier B, et al. FTY720 0124 study group. FTY720 versus MMF with cyclosporine in de novo renal transplantation: a 1‐year, randomized controlled trial in Europe and Australasia. Am J Transplant. 2006;6:2912‐2921.
    1. Westhoff TH, Schmidt S, Glander P, et al. The impact of FTY720 (fingolimod) on vasodilatory function and arterial elasticity in renal transplant patients. Nephrol Dial Transplant. 2007;22:2354‐2358.
    1. Poirier B, Briand V, Kadereit D, et al. A G‐protein‐biased S1P1 agonist, SAR247799, protects endothelial cells without affecting lymphocyte numbers. Sci Sign. 2020;13:eaax8050.
    1. Graihle P, Boutarfa‐Madec A, Beauverger P, Janiak P, Parkar AA. A label‐free impedance assay in endothelial cells distinguishes between the activation and desensitization properties of clinical sphingosine‐1 phosphate receptor‐1 agonists. FEBS Open Bio. 2020:2010‐2020. 10.1002/2211-5463.12951
    1. Bergougnan L, Armani S, Golor G, et al. First‐in‐human study of the safety, tolerability, pharmacokinetics and pharmacodynamics of single and multiple oral doses of SAR247799, a selective G‐protein‐biased Sphingosine‐1 phosphate receptor‐1 agonist for endothelial protection. Br J Clin Pharm. 2020:1‐14. 10.1111/bcp.14422
    1. Thomas N, Sweeney K, Somayaji V. Meta‐analysis of clinical dose‐response in a large drug development portfolio. Stat Biopharmaceutical Res. 2014;6:302‐317.
    1. Clarkson P, Celermajer DS, Donald AE, et al. Impaired vascular reactivity in insulin dependent diabetes mellitus is related to disease duration and low‐density lipoprotein cholesterol levels. J Am Coll Cardiol. 1996;28(3):573‐579.
    1. Matsuzawa Y, Kwon TG, Lennon RJ, Lerman LO, Lerman A. Prognostic value of flow‐mediated vasodilation in brachial artery and fingertip artery for cardiovascular events: a systematic review and meta‐analysis. J Am Heart Assoc. 2015:4. 10.1161/JAHA.115.002270
    1. Aversa A, Vitale C, Volterrani M, et al. Chronic administration of sildenafil improves markers of endothelial function in men with type 2 diabetes. Diabet Med. 2008;25(1):37‐44.
    1. Balzer J, Rassaf T, Heiss C, et al. Sustained benefits in vascular function through Flavanol‐containing cocoa in medicated diabetic patients. A double‐masked, randomized, controlled trial. J Am Coll Cardiol. 2008;51(22):2141‐2149.
    1. Desouza C, Parulkar A, Lumpkin D, Akers D, Fonseca V. Acute and prolonged effects of sildenafil on brachial artery flow‐mediated dilatation in type 2 diabetes. Diabetes Care. 2002;25(8):1336‐1339.
    1. Deyoung L, Chung E, Kovac JR, Romano W, Brock GB. Daily use of sildenafil improves endothelial function in men with type 2 diabetes. J Androl. 2012;33(2):176‐180.
    1. Halcox JP, Nour KR, Zalos G, et al. The effect of sildenafil on human vascular function, platelet activation, and myocardial ischemia. J Am Coll Cardiol. 2002;40(7):1232‐1240.
    1. Rosano GM, Aversa A, Vitale C, Fabbri A, Fini M, Spera G. Chronic treatment with tadalafil improves endothelial function in men with cardiovascular risk. Eur Urol. 2005;47:214‐222.
    1. Shahin Y, Khan JA, Samuel N, Cheeler I. Angiotensin converting enzyme inhibitors effect on endothelial dysfunction: a meta‐analysis of randomised controlled trials. Atherosclerosis. 2011;216(1):7‐16.
    1. Zhang L, Gong D, Li S, Zhou X. Meta‐analysis of the effects of statin therapy on endothelial function in patients with diabetes mellitus. Atherosclerosis. 2012;223(1):78‐85.
    1. Li S, Wu Y, Yu G, Xia Q, Xu Y. Angiotensin II receptor blockers improve peripheral endothelial function: a meta‐analysis of randomized controlled trials. PloS One. 2014. Mar 3;9(3):e90217. 10.1371/journal.pone.0090217
    1. Clark JB, Palmer CJ, Shaw WN. The diabetic Zucker fatty rat. Proc Soc Exp Biol Med. 1983;173(1):68‐75.
    1. Etgen GJ, Oldham BA. Profiling of Zucker diabetic fatty rats in their progression to the overt diabetic state. Metabolism. 2000;49(5):684‐688.
    1. Ratcliffe B, Pawlak R, Morales F, Harrison C, Gurovich AN. Internal validation of an automated system for brachial and femoral flow mediated dilation. Clin Hypertens. 2017;23:17. 10.1186/s40885-017-0073-1
    1. Southan C, Sharman JL, Benson HE, et al. The IUPHAR/BPS guide to PHARMACOLOGY in 2016: towards curated quantitative interactions between 1300 protein targets and 6000 ligands. Nucleic Acids Res. 2016;44(D1):D1054‐D1068.
    1. Alexander SP, Christopoulos A, Davenport AP, et al. The concise guide to pharmacology 2019/20: G protein‐coupled receptors; 2019:S21‐S141.
    1. Evans CE, Iruela‐Arispe ME, Zhao Y‐Y. Mechanisms of endothelial regeneration and vascular repair and their application to regenerative medicine. Am J Pathology. 2020. 10.1016/j.ajpath.2020.10.001
    1. Stirban A, Laude D, Elghozi JL, et al. Acute effects of sildenafil on flow mediated dilatation and cardiovascular autonomic nerve function in type 2 diabetic patients. Diabetes Metab Res Rev. 2009;25(2):136‐143.
    1. Halcox JP, Donald AE, Ellins E, et al. Endothelial function predicts progression of carotid intima‐media thickness. Circulation. 2009;119(7):1005‐1012.
    1. Ceconi C, Fox KM, Remme WJ, et al. ACE inhibition with perindopril and endothelial function. Results of a substudy of the EUROPA study: PERTINENT. Cardiovasc Res. 2007;73(1):237‐246.
    1. Taddei S, Virdis A, Ghiadoni L, et al. Effect of calcium antagonist or beta blockade treatment on nitric oxide‐dependent vasodilation and oxidative stress in essential hypertensive patients. J Hypertens. 2001;19(8):1379‐1386.
    1. Rubinshtein R, Kuvin JT, Soffler M, et al. Assessment of endothelial function by non‐invasive peripheral arterial tonometry predicts late cardiovascular adverse events. Eur Heart J. 2010;31(9):1142‐1148.
    1. Barter PJ, Caulfield M, Eriksson M, et al. Effects of torcetrapib in patients at high risk for coronary events. N Engl J Med. 2007;357(21):2109‐2122.
    1. Simic B, Hermann M, Shaw SG, et al. Torcetrapib impairs endothelial function in hypertension. Eur Heart J. 2012;33(13):1615‐1624.
    1. Lüscher TF, Taddei S, Kaski J‐C, et al. Vascular effects and safety of dalcetrapib in patients with or at risk of coronary heart disease: the dal‐VESSEL randomized clinical trial. Eur Heart J. 2012;33(7):857‐865.
    1. Schwartz GG, Olsson AG, Abt M, et al. Dal‐OUTCOMES investigators. Effects of dalcetrapib in patients with a recent acute coronary syndrome. N Engl J Med. 2012;367(22):2089‐2099.
    1. Charakida M, Masi S, Lüscher TF, Kastelein JJ, Deanfield JE. Assessment of atherosclerosis: the role of flow‐mediated dilatation. Eur Heart J. 2010;31:2854‐2861.
    1. Kuno Y, Iyoda M, Shibata T, Hirai Y, Akizawa T. Sildenafil, a phosphodiesterase type 5 inhibitor, attenuates diabetic nephropathy in non‐insulin‐dependent Otsuka long‐Evans Tokushima fatty rats. Br J Pharmacol. 2011;162(6):1389‐1400.
    1. Jeong KH, Lee TW, Ihm CG, Lee SH, Moon JY, Lim SJ. Effects of sildenafil on oxidative and inflammatory injuries of the kidney in streptozotocin‐induced diabetic rats. Am J Nephrol. 2009;29(3):274‐282.
    1. Igarashi J, Michel T. Sphingosine‐1‐phosphate and modulation of vascular tone. Cardiovasc Res. 2009;82(2):212‐220.
    1. Toblli JE, Cao G, Angerosa M, Rivero M. Long‐term phosphodiesterase 5 inhibitor administration reduces inflammatory markers and heat‐shock proteins in cavernous tissue of Zucker diabetic fatty rat (ZDF/fa/fa). Int J Impot Res. 2015;27(5):182‐190.
    1. El‐Mahdy NA, El‐Sayad ME, El‐Kadem AH. Combination of telmisartan with sildenafil ameliorate progression of diabetic nephropathy in streptozotocin‐induced diabetic model. Biomed Pharmacother. 2016;81:136‐144.

Source: PubMed

3
Předplatit