LX4211, a dual SGLT1/SGLT2 inhibitor, improved glycemic control in patients with type 2 diabetes in a randomized, placebo-controlled trial

B Zambrowicz, J Freiman, P M Brown, K S Frazier, A Turnage, J Bronner, D Ruff, M Shadoan, P Banks, F Mseeh, D B Rawlins, N C Goodwin, R Mabon, B A Harrison, A Wilson, A Sands, D R Powell, B Zambrowicz, J Freiman, P M Brown, K S Frazier, A Turnage, J Bronner, D Ruff, M Shadoan, P Banks, F Mseeh, D B Rawlins, N C Goodwin, R Mabon, B A Harrison, A Wilson, A Sands, D R Powell

Abstract

Thirty-six patients with type 2 diabetes mellitus (T2DM) were randomized 1:1:1 to receive a once-daily oral dose of placebo or 150 or 300 mg of the dual SGLT1/SGLT2 inhibitor LX4211 for 28 days. Relative to placebo, LX4211 enhanced urinary glucose excretion by inhibiting SGLT2-mediated renal glucose reabsorption; markedly and significantly improved multiple measures of glycemic control, including fasting plasma glucose, oral glucose tolerance, and HbA(1c); and significantly lowered serum triglycerides. LX4211 also mediated trends for lower weight, lower blood pressure, and higher glucagon-like peptide-1 levels. In a follow-up single-dose study in 12 patients with T2DM, LX4211 (300 mg) significantly increased glucagon-like peptide-1 and peptide YY levels relative to pretreatment values, probably by delaying SGLT1-mediated intestinal glucose absorption. In both studies, LX4211 was well tolerated without evidence of increased gastrointestinal side effects. These data support further study of LX4211-mediated dual SGLT1/SGLT2 inhibition as a novel mechanism of action in the treatment of T2DM.

Trial registration: ClinicalTrials.gov NCT00962065 NCT01188863.

Figures

Figure 1
Figure 1
Effect of LX4211 on glycemic parameters and UGE. (a) Chemical structure of LX4211, (2S,3R,4R,5S,6R)-2-(4-chloro-3-(4-ethoxybenzyl)phenyl)-6-(methylthio)tetrahydro-2H-pyran-3,4,5-triol. (b) FPG. This schematic representation of the study design depicts mean changes in FPG levels from baseline (day −1) values throughout the study. (c) HbA1c. Change in HbA1c levels after 28 days of treatment with LX4211 or placebo. (d) OGTT glucose AUCs. AUCs for glucose obtained from OGTTs performed on days −2, 2, 13, and 27. (e) OGTT glucose excursions above fasting (hour 0) values. OGTT glucose excursions were plotted after correcting for differences in FPG by adjusting hour 0 glucose values to 0 mg/dl. Left, data from day −2; right, data from day 27. (f) UGE. UGE was estimated by measuring the total amount of glucose in 24-h urine samples collected on days −1, 1, 14, and 28; the data shown represent the mean change from baseline (day −1) values. The color key identifying each group in bf is at the bottom of the figure. For d and e, AUC values for each LX4211 treatment group on each day were compared with the AUC values of the placebo group on that day. For b,d,f: different from placebo, ***P < 0.001, **P < 0.01; different from 150 mg dose group, ••P < 0.01, •P < 0.05. ♦, n = 11 for the placebo group on days 27 and 28. For e, AUC values on day 27 were significantly lower for each LX4211 treatment group as compared with placebo, P < 0.001. Error bars in c represent 1 SD. All the data are presented as arithmetic means except in c, in which the data are presented as differences in least squares means. AUC, area under the curve for glucose concentration; FPG, fasting plasma glucose; HbA1c, hemoglobin A1c; OGTT, oral glucose tolerance test; UGE, urinary glucose excretion.
Figure 2
Figure 2
Effects of single doses of liquid or solid forms of oral LX4211 on UGE and on circulating levels of GLP-1, PYY, and LX4211. (a) Study design. This schematic representation depicts the treatment sequence; each of three groups of patients (n = 4 per group) received each of three LX4211 formulations on days 1, 6, and 11. Metformin was washed out before day 1, and LX4211 formulations administered on days 1 and 6 were washed out over the 5 days following each of the doses. The levels of UGE, GLP-1, PYY, and LX4211 measured on the day when the LX4211 formulation was administered to 12 patients with type 2 diabetes mellitus (T2DM) were compared with those on day −1 (baseline control levels) measured in the same 12 patients. (b) UGE. UGE was estimated by measuring the total amount of glucose present in 24-h urine samples collected at baseline (day −1) and on the days when each LX4211 formulation was administered; the data shown represent mean changes from baseline values. (ce) Circulating GLP-1 and PYY. These were measured at the same time points on day −1 and on the days when each LX4211 formulation was administered. In ce, arrows show the time points at which meals were provided, and P values are presented at the bottom of each of these panels. (c) Total GLP-1. (d) Active GLP-1. (e) Total PYY. (f) Time course of plasma LX4211 levels after oral administration of liquid or tablet LX4211 forms. The color key identifying each group in bf is at the bottom of the figure. Error bars in b represent 1 SD. All data are presented as arithmetic means. AUC, area under the curve; D/C, discontinue; GLP-1, glucagon-like peptide-1; PK, pharmacokinetic analysis; PYY, peptide YY; UGE, urinary glucose excretion.

References

    1. Huang E.S., Basu A., O'Grady M., &, Capretta J.C. Projecting the future diabetes population size and related costs for the U.S. Diabetes Care. 2009;32:2225–2229.
    1. The Diabetes Control and Complications Trial Research Group The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. N Engl. J. Med. 1993;329:977–986.
    1. Nathan D.M., Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) Study Research Group et al. Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N. Engl. J. Med. 2005;353:2643–2653.
    1. UK Prospective Diabetes Study (UKPDS) Group Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with Type 2 diabetes (UKPDS 33) Lancet. 1998;352:837–853.
    1. UK Prospective Diabetes Study (UKPDS) Group Effect of intensive blood-glucose control with metformin on complications in overweight patients with Type 2 diabetes (UKPDS 34) Lancet. 1998;352:854–865.
    1. Holman R.R., Paul S.K., Bethel M.A., Matthews D.R., &, Neil H.A. 10-year follow-up of intensive glucose control in type 2 diabetes. N. Engl. J. Med. 2008;359:1577–1589.
    1. Ray al. Effect of intensive control of glucose on cardiovascular outcomes and death in patients with diabetes mellitus: a meta-analysis of randomised controlled trials Lancet 3731765–1772.2009
    1. Wallace T.M., &, Matthews D.R. Coefficient of failure: a methodology for examining longitudinal beta-cell function in Type 2 diabetes. Diabet. Med. 2002;19:465–469.
    1. Fonseca V.A. Defining and characterizing the progression of type 2 diabetes. Diabetes Care. 2009;32 suppl. 2:S151–S156.
    1. Rodbard al. Statement by an American Association of Clinical Endocrinologists/American College of Endocrinology consensus panel on type 2 diabetes mellitus: an algorithm for glycemic control Endocr. Pract 15540–559.2009
    1. Nathan D.M., American Diabetes Association; European Association for Study of Diabetes et al. Medical management of hyperglycemia in type 2 diabetes: a consensus algorithm for the initiation and adjustment of therapy: a consensus statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care. 2009;32:193–203.
    1. Rossetti L., Smith D., Shulman G.I., Papachristou D., &, DeFronzo R.A. Correction of hyperglycemia with phlorizin normalizes tissue sensitivity to insulin in diabetic rats. J. Clin. Invest. 1987;79:1510–1515.
    1. Komoroski al. Dapagliflozin, a novel SGLT2 inhibitor, induces dose-dependent glucosuria in healthy subjects Clin. Pharmacol. Ther 85520–526.2009
    1. Komoroski B., Vachharajani N., Feng Y., Li L., Kornhauser D., &, Pfister M. Dapagliflozin, a novel, selective SGLT2 inhibitor, improved glycemic control over 2 weeks in patients with type 2 diabetes mellitus. Clin. Pharmacol. Ther. 2009;85:513–519.
    1. List J.F., Woo V., Morales E., Tang W., &, Fiedorek F.T. Sodium-glucose cotransport inhibition with dapagliflozin in type 2 diabetes. Diabetes Care. 2009;32:650–657.
    1. Wilding J.P., Norwood P., T'joen C., Bastien A., List J.F., &, Fiedorek F.T. A study of dapagliflozin in patients with type 2 diabetes receiving high doses of insulin plus insulin sensitizers: applicability of a novel insulin-independent treatment. Diabetes Care. 2009;32:1656–1662.
    1. Bailey C.J. Renal glucose reabsorption inhibitors to treat diabetes. Trends Pharmacol. Sci. 2011;32:63–71.
    1. Ferrannini E., Ramos S.J., Salsali A., Tang W., &, List J.F. Dapagliflozin monotherapy in type 2 diabetic patients with inadequate glycemic control by diet and exercise: a randomized, double-blind, placebo-controlled, phase 3 trial. Diabetes Care. 2010;33:2217–2224.
    1. Oku al. T-1095, an inhibitor of renal Na+-glucose cotransporters, may provide a novel approach to treating diabetes Diabetes 481794–1800.1999
    1. Arakawa al. Improved diabetic syndrome in C57BL/KsJ-db/db mice by oral administration of the Na(+)-glucose cotransporter inhibitor T-1095 Br. J. Pharmacol 132578–586.2001
    1. Fujimori Y., Katsuno K., Nakashima I., Ishikawa-Takemura Y., Fujikura H., &, Isaji M. Remogliflozin etabonate, in a novel category of selective low-affinity sodium glucose cotransporter (SGLT2) inhibitors, exhibits antidiabetic efficacy in rodent models. J. Pharmacol. Exp. Ther. 2008;327:268–276.
    1. Han al. Dapagliflozin, a selective SGLT2 inhibitor, improves glucose homeostasis in normal and diabetic rats Diabetes 571723–1729.2008
    1. Katsuno K., Fujimori Y., Ishikawa-Takemura Y., &, Isaji M. Long-term treatment with sergliflozin etabonate improves disturbed glucose metabolism in KK-A(y) mice. Eur. J. Pharmacol. 2009;618:98–104.
    1. Musso G., Gambino R., Cassader M., Pagano G. A novel approach to control hyperglycemia in type 2 diabetes: sodium glucose co-transport (SGLT) inhibitors. Systematic review and meta-analysis of randomized trials. Ann. Med. 2012;44:375–393.
    1. Wright E.M. I. Glucose galactose malabsorption. Am. J. Physiol. 1998;275:G879–G882.
    1. Washburn W.N. Evolution of sodium glucose co-transporter 2 inhibitors as anti-diabetic agents. Expert Opin. Ther. Pat. 2009;19:1485–1499.
    1. Zhou al. Dietary resistant starch upregulates total GLP-1 and PYY in a sustained day-long manner through fermentation in rodents Am. J. Physiol. Endocrinol. Metab 295E1160–E1166.2008
    1. Nilsson A.C., Ostman E.M., Holst J.J., &, Björck I.M. Including indigestible carbohydrates in the evening meal of healthy subjects improves glucose tolerance, lowers inflammatory markers, and increases satiety after a subsequent standardized breakfast. J. Nutr. 2008;138:732–739.
    1. Meirelles K., Ahmed T., Culnan D.M., Lynch C.J., Lang C.H., &, Cooney R.N. Mechanisms of glucose homeostasis after Roux-en-Y gastric bypass surgery in the obese, insulin-resistant Zucker rat. Ann. Surg. 2009;249:277–285.
    1. Cummings D.E. Endocrine mechanisms mediating remission of diabetes after gastric bypass surgery. Int. J. Obes. (Lond) 2009;33 suppl. 1:S33–S40.
    1. Goodwin al. Novel L-xylose derivatives as selective sodium-dependent glucose cotransporter 2 (SGLT2) inhibitors for the treatment of type 2 diabetes J. Med. Chem 526201–6204.2009
    1. Samtani M.N. Simple pharmacometric tools for oral anti-diabetic drug development: competitive landscape for oral non-insulin therapies in type 2 diabetes. Biopharm. Drug Dispos. 2010;31:162–177.
    1. Ikumi Y., Kida T., Sakuma S., Yamashita S., &, Akashi M. Polymer-phloridzin conjugates as an anti-diabetic drug that inhibits glucose absorption through the Na+/glucose cotransporter (SGLT1) in the small intestine. J. Control. Release. 2008;125:42–49.
    1. Sakuma al. Carboxyl group-terminated polyamidoamine dendrimers bearing glucosides inhibit intestinal hexose transporter-mediated D-glucose uptake Eur. J. Pharm. Biopharm 75366–374.2010
    1. Ballantyne G.H. Peptide YY(1-36) and peptide YY(3-36): Part I. Distribution, release and actions. Obes. Surg. 2006;16:651–658.
    1. Longo W.E., Ballantyne G.H., Savoca P.E., Adrian T.E., Bilchik A.J., &, Modlin I.M. Short-chain fatty acid release of peptide YY in the isolated rabbit distal colon. Scand. J. Gastroenterol. 1991;26:442–448.
    1. Steinert R.E., Gerspach A.C., Gutmann H., Asarian L., Drewe J., &, Beglinger C. The functional involvement of gut-expressed sweet taste receptors in glucose-stimulated secretion of glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) Clin. Nutr. 2011;30:524–532.
    1. Steinert R.E., Frey F., Töpfer A., Drewe J., &, Beglinger C. Effects of carbohydrate sugars and artificial sweeteners on appetite and the secretion of gastrointestinal satiety peptides. Br. J. Nutr. 2011;105:1320–1328.
    1. Abbott al. The inhibitory effects of peripheral administration of peptide YY(3-36) and glucagon-like peptide-1 on food intake are attenuated by ablation of the vagal-brainstem-hypothalamic pathway Brain Res 1044127–131.2005
    1. Koda al. The role of the vagal nerve in peripheral PYY3-36-induced feeding reduction in rats Endocrinology 1462369–2375.2005
    1. Balkan B., &, Li X. Portal GLP-1 administration in rats augments the insulin response to glucose via neuronal mechanisms. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2000;279:R1449–R1454.
    1. Wan S., Coleman F.H., &, Travagli R.A. Glucagon-like peptide-1 excites pancreas-projecting preganglionic vagal motoneurons. Am. J. Physiol. Gastrointest. Liver Physiol. 2007;292:G1474–G1482.
    1. Ahrén B. Sensory nerves contribute to insulin secretion by glucagon-like peptide-1 in mice. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2004;286:R269–R272.
    1. Baggio L.L., &, Drucker D.J. Biology of incretins: GLP-1 and GIP. Gastroenterology. 2007;132:2131–2157.
    1. Chao E.C., &, Henry R.R. SGLT2 inhibition–a novel strategy for diabetes treatment. Nat. Rev. Drug Discov. 2010;9:551–559.
    1. Goldring W., &, Welsh C. The effects on renal activity of the oral administration of phlorizin in man. J. Clin. Invest. 1934;13:749–752.
    1. Crespy al. Bioavailability of phloretin and phloridzin in rats J. Nutr 1313227–3230.2001
    1. Ehrenkranz J.R., Lewis N.G., Kahn C.R., &, Roth J. Phlorizin: a review. Diabetes Metab. Res. Rev. 2005;21:31–38.
    1. Kellett G.L., &, Helliwell P.A. The diffusive component of intestinal glucose absorption is mediated by the glucose-induced recruitment of GLUT2 to the brush-border membrane. Biochem. J. 2000;350 Pt 1:155–162.
    1. Fan H.T., Morishima S., Kida H., &, Okada Y. Phloretin differentially inhibits volume-sensitive and cyclic AMP-activated, but not Ca-activated, Cl(-) channels. Br. J. Pharmacol. 2001;133:1096–1106.
    1. De Jonge P.C., Wieringa T., Van Putten J.P., Krans H.M., &, Van Dam K. Phloretin - an uncoupler and an inhibitor of mitochondrial oxidative phosphorylation. Biochim. Biophys. Acta. 1983;722:219–225.
    1. Wright E.M., Loo D.D., &, Hirayama B.A. Biology of human sodium glucose transporters. Physiol. Rev. 2011;91:733–794.
    1. White J. Efficacy and safety of incretin based therapies: clinical trial data. J. Am. Pharm. Assoc. 2003;49 suppl. 1:S30–S40.
    1. Horton E.S., Silberman C., Davis K.L., &, Berria R. Weight loss, glycemic control, and changes in cardiovascular biomarkers in patients with type 2 diabetes receiving incretin therapies or insulin in a large cohort database. Diabetes Care. 2010;33:1759–1765.
    1. US Food and Drug Administration, Center for Drug Evaluation and Research (CDER) Guidance for industry: diabetes mellitus—evaluating cardiovascular risk in new antidiabetic therapies to treat type 2 diabetes, December 2008 < < >. Accessed 3 May 2011.
    1. Buse J.B., American Heart Association; American Diabetes Association et al. Primary prevention of cardiovascular diseases in people with diabetes mellitus: a scientific statement from the American Heart Association and the American Diabetes Association. Diabetes Care. 2007;30:162–172.
    1. Cockcroft D.W., &, Gault M.H. Prediction of creatinine clearance from serum creatinine. Nephron. 1976;16:31–41.
    1. Matthews D.R., Hosker J.P., Rudenski A.S., Naylor B.A., Treacher D.F., &, Turner R.C. Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia. 1985;28:412–419.

Source: PubMed

3
Sottoscrivi