Effect of bromocriptine-QR (a quick-release formulation of bromocriptine mesylate) on major adverse cardiovascular events in type 2 diabetes subjects

J Michael Gaziano, Anthony H Cincotta, Aaron Vinik, Lawrence Blonde, Nancy Bohannon, Richard Scranton, J Michael Gaziano, Anthony H Cincotta, Aaron Vinik, Lawrence Blonde, Nancy Bohannon, Richard Scranton

Abstract

Background: Bromocriptine-QR (a quick-release formulation of bromocriptine mesylate), a dopamine D2 receptor agonist, is a US Food and Drug Administrration-approved treatment for type 2 diabetes mellitus (T2DM). A 3070-subject randomized trial demonstrated a significant, 40% reduction in relative risk among bromocriptine-QR-treated subjects in a prespecified composite cardiovascular (CV) end point that included ischemic-related (myocardial infarction and stroke) and nonischemic-related (hospitalization for unstable angina, congestive heart failure [CHF], or revascularization surgery) end points, but did not include cardiovascular death as a component of this composite. The present investigation was undertaken to more critically evaluate the impact of bromocriptine-QR on cardiovascular outcomes in this study subject population by (1) including CV death in the above-described original composite analysis and then stratifying this new analysis on the basis of multiple demographic subgroups and (2) analyzing the influence of this intervention on only the "hard" CV end points of myocardial infarction, stroke, and CV death (major adverse cardiovascular events [MACEs]).

Methods and results: Three thousand seventy T2DM subjects on stable doses of ≤2 antidiabetes medications (including insulin) with HbA1c ≤10.0 (average baseline HbA1c=7.0) were randomized 2:1 to bromocriptine-QR (1.6 to 4.8 mg/day) or placebo for a 52-week treatment period. Subjects with heart failure (New York Heart Classes I and II) and precedent myocardial infarction or revascularization surgery were allowed to participate in the trial. Study outcomes included time to first event for each of the 2 CV composite end points described above. The relative risk comparing bromocriptine-QR with the control for the cardiovascular outcomes was estimated as a hazard ratio with 95% confidence interval on the basis of Cox proportional hazards regression. The statistical significance of any between-group difference in the cumulative percentage of CV events over time (derived from a Kaplan-Meier curve) was determined by a log-rank test on the intention-to-treat population. Study subjects were in reasonable metabolic control, with an average baseline HbA1c of 7.0±1.1, blood pressure of 128/76±14/9, and total and LDL cholesterol of 179±42 and 98±32, respectively, with 88%, 77%, and 69% of subjects being treated with antidiabetic, antihypertensive, and antihyperlipidemic agents, respectively. Ninety-one percent of the expected person-year outcome ascertainment was obtained in this study. Respecting the CV-inclusive composite cardiovascular end point, there were 39 events (1.9%) among 2054 bromocriptine-QR-treated subjects versus 33 events (3.2%) among 1016 placebo subjects, yielding a significant, 39% reduction in relative risk in this end point with bromocriptine-QR exposure (P=0.0346; log-rank test) that was not influenced by age, sex, race, body mass index, duration of diabetes, or preexisting cardiovascular disease. In addition, regarding the MACE end point, there were 14 events (0.7%) among 2054 bromocriptine-QR-treated subjects and 15 events (1.5%) among 1016 placebo-treated subjects, yielding a significant, 52% reduction in relative risk in this end point with bromocriptine-QR exposure (P<0.05; log-rank test).

Conclusions: These findings reaffirm and extend the original observation of relative risk reduction in cardiovascular adverse events among type 2 diabetes subjects treated with bromocriptine-QR and suggest that further investigation into this impact of bromocriptine-QR is warranted.

Clinical trial registration: URL: https://ichgcp.net/clinical-trials-registry/NCT00377676" title="See in ClinicalTrials.gov">NCT00377676.

Keywords: Cycloset; bromocriptine; circadian rhythm; diabetes mellitus type 2; infarction.

Figures

Figure 1.
Figure 1.
Subject disposition. ITT indicates intention-to-treat.
Figure 2.
Figure 2.
Time to composite end point of cardiovascular serious adverse events including nonfatal myocardial infarction (excluding silent MI), stroke, hospitalization for angina or hospitalization for heart failure, coronary revascularization, and cardiovascular death. SAEs indicates serious adverse events; MI, myocardial infarction.
Figure 3.
Figure 3.
Time to composite end point of nonfatal myocardial infarction (excluding silent MI) or stroke or cardiovascular death (MACE) in ITT population for bromocriptine-QR and placebo groups. SAEs indicates serious adverse events; MI, myocardial infarction; and ITT, intention-to-treat.

References

    1. Cincotta AH, Meier AH, Cincotta M., Jr Bromocriptine improves glycaemic control and serum lipid profile in obese type 2 diabetic subjects: a new approach in the treatment of diabetes. Expert Opin Investig Drugs. 1999;8:1683-1707.
    1. Scranton R, Cincotta A. Bromocriptine – unique formulation of a dopamine agonist for the treatment of type 2 diabetes. Expert Opin Pharmacother. 2010;11:269-279.
    1. Cycloset. 2010Tiverton, RI: VeroScience LLC; [package insert]
    1. Scranton RE, Gaziano JM, Rutty D, Ezrokhi M, Cincotta A. A randomized, double-blind, placebo-controlled trial to assess safety and tolerability during treatment of type 2 diabetes with usual diabetes therapy and either Cycloset or placebo. BMC Endocr Disord. 2007;7:3.
    1. Gaziano JM, Cincotta AH, O'Connor CM, Ezrokhi M, Rutty D, Ma ZJ, Scranton RE. Randomized clinical trial of quick-release bromocriptine among patients with type 2 diabetes on overall safety and cardiovascular outcomes. Diabetes Care. 2010;33:1503-1508.
    1. American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care. 2004;27suppl 1S5-S10.
    1. American Diabetes Association. Standards of medical care in diabetes. Diabetes Care. 2004;27suppl 1S15-S35.
    1. Action to Control Cardiovascular Risk in Diabetes Study Group. Gerstein HC, Miller ME, Byington RP, Goff DC, Jr, Bigger JT, Buse JB, Cushman WC, Genuth S, Ismail-Beigi F, Grimm RH, Jr, Probstfield JL, Simons-Morton DG, Friedewald WT. Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med. 2008;358:2545-2559.
    1. Patel A, MacMahon S, Chalmers J, Neal B, Woodward M, Billot L, Harrap S, Poulter N, Marre M, Cooper M, Glasziou P, Grobbee DE, Hamet P, Heller S, Liu LS, Mancia G, Mogensen CE, Pan CY, Rodgers A, Williams BADVANCE Collaborative Group. Effects of a fixed combination of perindopril and indapamide on macrovascular and microvascular outcomes in patients with type 2 diabetes mellitus (the ADVANCE trial): a randomised controlled trial. Lancet. 2007;370:829-840.
    1. Cincotta AH.Hansen B, Shafrir E. Hypothalamic role in insulin resistance and insulin resistance syndrome. Frontiers in Animal Diabetes Research. 2002London: Taylor and Francis; 271-312.
    1. Liang Y, Cincotta AH. Increased responsiveness to the hyperglycemic, hyperglucagonemic and hyperinsulinemic effects of circulating norepinephrine in ob/ob mice. Int J Obes Relat Metab Disord. 2001;25:698-704.
    1. Arner P. Catecholamine-induced lipolysis in obesity. Int J Obes Relat Metab Disord. 1999;23suppl 110-13.
    1. Cincotta AH, Luo S, Zhang Y, Liang Y, Bina KG, Jetton TL, Scislowski PW. Chronic infusion of norepinephrine into the VMH of normal rats induces the obese glucose-intolerant state. Am J Physiol Regul Integr Comp Physiol. 2000;278:R435-R444.
    1. Cusi K. The role of adipose tissue and lipotoxicity in the pathogenesis of type 2 diabetes. Curr Diab Rep. 2010;10:306-315.
    1. Cherrington AD. Control of glucose uptake and release by the liver in vivo. Diabetes. 1999;48:1198-1214.
    1. Dicostanzo CA, Dardevet DP, Neal DW, Lautz M, Allen E, Snead W, Cherrington AD. Role of the hepatic sympathetic nerves in the regulation of net hepatic glucose uptake and the mediation of the portal glucose signal. Am J Physiol. 2006;290:E9-E16.
    1. Nielsen MF, Caumo A, Chandramouli V, Schumann WC, Cobelli C, Landau BR, Vilstrup H, Rizza RA, Schmitz O. Impaired basal glucose effectiveness but unaltered fasting glucose release and gluconeogenesis during short-term hypercortisolemia in healthy subjects. Am J Physiol Endocrinol Metab. 2004;286:E102-E110.
    1. Goldstein RE, Rossetti L, Palmer BA, Liu R, Massillon D, Scott M, Neal D, Williams P, Peeler B, Cherrington AD. Effects of fasting and glucocorticoids on hepatic gluconeogenesis assessed using two independent methods in vivo. Am J Physiol Endocrinol Metab. 2002;283:E946-E957.
    1. Grassi G, Bombelli M, Seravalle G, Dell'Oro R, Quarti-Trevano F. Diurnal blood pressure variation and sympathetic activity. Hypertens Res. 2010;33:381-385.
    1. Tsuji H, Larson MG, Venditti FJ, Jr, Manders ES, Evans JC, Feldman CL, Levy D. Impact of reduced heart rate variability on risk factors for cardiac events: the Framingham Heart Study. Circulation. 1992;94:2850-2855.
    1. Kannel WB, Kanel C, Paffenbarger RS, Cupples LA. Heart rate and cardiovascular mortality: the Framingham Study. Am Heart J. 1987;113:1489-1494.
    1. Huikuri HV, Jokinen V, Syvänne M, Nieminen MS, Airaksinen KE, Ikäheimo MJ, Koistinen JM, Kauma H, Kesäniemi AY, Majahalme S, Niemelä KO, Frick MH. Heart rate variability and progression of coronary atherosclerosis. Arterioscler Thromb Vasc Biol. 1999;19:1979-1985.
    1. Vinik AI, Maser RE, Ziegler D. Autonomic imbalance: prophet of doom or scope for hope?. Diabet Med. 2011;28:643-651.
    1. Yu S-M, Tsai S-Y, Guh J-H, Lo F-H, Teng C-M, Ou J-T. Mechanisms of catecholamine-induced proliferation of vascular smooth muscle cells. Circulation. 1996;94:547-554.
    1. Kukerja RS, Datta BN, Chakra-Vari RN. Catecholamine-induced aggravation of aortic and coronary atherosclerosis in monkeys. Atherosclerosis. 1981;40:291-298.
    1. Beere PA, Glagov S, Zarins CK. Retarding effect of lowered heart rate on coronary atherosclerosis. Science. 1984;226:180-182.
    1. Ramchandra R, Barrett CJ, Malpas SC. Nitric oxide and sympathetic nerve activity in the control of blood pressure. Clin Exp Pharmacol Physiol. 2005;32:440-446.
    1. Palatini P. Sympathetic overactivity in hypertension: a risk factor for cardiovascular disease. Curr Hypertens Rep. 2001;3suppl 1S3-S9.
    1. Fagerholm V, Haaparanta M, Scheinin M. Alpha2-adrenoceptor regulation of blood glucose homeostasis. Basic Clin Pharmacol Toxicol. 2011;108:365-370.
    1. Bernstein IM, Damron D, Schonberg AL, Shapiro R. The relationship of plasma volume, sympathetic tone, and proinflammatory cytokines in young healthy nonpregnant women. Reprod Sci. 2009;16:980-985.
    1. Palatini P. Elevated heart rate as a predictor of increased cardiovascular morbidity. J Hypertens Suppl. 1999;17:S3-S10.
    1. Schulz E, Gori T, Munzel T. Oxidative stress and endothelial dysfunction in hypertension. Hypertens Res. 2011;34:665-673.
    1. Gayen JR, Zhang K, Ramachandra Rao SP, Mahata M, Chen Y, Kim HS, Naviaux RK, Sharma K, Mahata SK, O'Connor DT. Role of reactive oxygen species in hyperadrenergic hypertension: biochemical, physiological, and pharmacological evidence from targeted ablation of the chromogranin a (Chga) gene. Circ Cardiovasc Genet. 2010;3:414-425.
    1. Rahman S, Rahman T, Ismail A, Rashid ARA. Diabetes-associated macrovasculopathy: pathophysiology and pathogenesis. Diabetes Obes Metab. 2007;9:767-780.
    1. Perin PC, Maule S, Quadri R. Sympathetic nervous system, diabetes, and hypertension. Clin Exp Hypertens. 2001;23:45-55.
    1. Masuo K, Rakugi H, Ogihara T, Esler MD, Lambert GW. Cardiovascular and renal complications of type 2 diabetes in obesity: role of sympathetic nerve activity and insulin resistance. Curr Diabetes Rev. 2010;6:58-67.
    1. Conde SV, Nunes da Silva T, Gonzalez C, Mota Carmo M, Monteiro EC, Guarino MP. Chronic caffeine intake decreases circulating catecholamines and prevents diet-induced insulin resistance and hypertension in rats. Br J Nutr. 2012;107:86-95.
    1. Lambert EA, Lambert GW. Stress and its role in sympathetic nervous system activation in hypertension and the metabolic syndrome. Curr Hypertens Rep. 2011;13:244-248.
    1. Lambert GW, Straznicky NE, Lambert EA, Dixon JB, Schlaich MP. Sympathetic nervous activation in obesity and the metabolic syndrome–causes, consequences and therapeutic implications. Pharmacol Ther. 2010;126:159-172.
    1. Ezrokhi M, Trubitsyna Y, Luo S, Cincotta AH. Timed dopamine agonist treatment ameliorates both vascular nitrosative/oxidative stress pathology and aortic stiffness in arteriosclerotic, hypertensive SHR rats (Abstract). Diabetes. 2010;59suppl 1A67
    1. Whaley-Connell A, McCullough PA, Sowers JR. The role of oxidative stress in the metabolic syndrome. Rev Cardiovasc Med. 2011;12:21-29.
    1. Cincotta AH, Gaziano JM, Ezrokhi M, Scranton RE. Cycloset (quick-release bromocriptine mesylate), a novel centrally acting treatment for type 2 diabetes (Abstract). Diabetologia. 2008;51suppl 1S22
    1. Scranton RE, Farwell W, Ezrokhi M, Gaziano JM, Cincotta AH. Quick release bromocriptine (Cycloset®) a novel treatment for type 2 diabetes also demonstrates improvements in blood pressure (Abstract). Can J Diabetes. 2009;3:235
    1. Sowers JR. Dopaminergic control of circadian norepinephrine levels in patients with essential hypertension. J Clin Endocrinol Metab. 1981;53:1133-1137.
    1. Sowers JR, Golub MS, Berger ME, Whitfield LA. Dopaminergic modulation of pressor and hormonal responses in essential hypertension. Hypertension. 1982;4:424-430.
    1. Kolloch R, Kobayashi K, DeQuattro V. Dopaminergic control of sympathetic tone and blood pressure: evidence on primary hypertension. Hypertension. 1980;2:390-394.

Source: PubMed

3
Abonner