evidence-based practice use of incretin-based therapy in ......, 0.01 vs controls). 43 in another...

19
CLINICAL FOCUS: DIABETES AND CONCOMITANT DISORDERS © Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260 1 ResearchSHARE ® : www.research-share.com • Permissions: [email protected] • Reprints: [email protected] Warning: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries. Evidence-Based Practice Use of Incretin-Based Therapy in the Natural History of Diabetes Stanley Schwartz, MD Affiliate, Main Line Health System, Ardmore, PA; Emeritus, Clinical Associate Professor of Medicine, University of Pennsylvania, Philadelphia, PA Correspondence: Stanley Schwartz, MD, 233 E Lancaster Ave, Ardmore, PA 19003. Tel: 610-642-6800 Fax: 610-642-6850 E-mail: [email protected] DOI: Abstract: The incretin class of anti-hyperglycemic agents, including glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-inhibitors, is an important addition to the therapeutic armamentarium for the management of appropriate patients with type 2 diabetes mellitus as an adjunct to diet and exercise and/or with the agents metformin, sulfonylureas, thiazolidinediones, or any combination thereof. More recently, US Food and Drug Administration (FDA)-approved indications for incretins were expanded to include use with basal insulin. This review article takes an evidence-based practice approach in discussing the importance of aggressive treatment for diabetes, the principles of incretin physiology and pathophysiology, use of glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors, and patient types and contexts where incretin therapy has been found beneficial, from metabolic syndrome to overt diabetes. Keywords: diabetes; dipeptidyl peptidase-4 inhibitors; glucagon-like peptide-1 receptor ago- nists; incretin Introduction The incretin class of anti-hyperglycemic agents is an important addition to the therapeutic armamentarium for the care of patients with type 2 diabetes mellitus (T2DM). Incretin- based therapies have been available since 2005 with the initial launch of glucagon-like peptide-1 (GLP-1) receptor agonists, followed by the launch of the dipeptidyl pepti- dase-4 (DPP-4) inhibitors. Although these agents were initially approved as an adjunct to diet and exercise and/or metformin, sulfonylureas, thiazolidinediones (TZDs), or any therapeutic combination of these agents in patients with T2DM, indications for incretin therapy have been recently expanded to include their use with basal insulin. This review takes a broad view of the use of GLP-1 receptor agonists and DPP-4 inhibitors based on the concept of evidence-based practice, the philosophy that takes the principles of evidence-based medicine and combines them with patient- based experience, clinical expertise, expert opinions, and existing guidelines to treat any individual patient in the optimal fashion. 1,2 The review provides a rationale for the need to treat patients aggressively, discusses principles of incretin physiology and pathophysiology, the incretin medications themselves (similarities and differences), and patient types and contexts where incretin therapy has proven to be useful, from metabolic syndrome (MetS) to overt diabetes. Materials and Methods The author’s clinical experience and expertise were used as the foundation for the article, supplemented by supportive literature related to the topic. Evidence-based SCHWARTZ_proof

Upload: others

Post on 20-Sep-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

C L I N I C A L F O C U S : D I A B E T E S A N D C O N C O M I TA N T D I S O R D E R S

© Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260 1ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

Evidence-Based Practice Use of Incretin-Based Therapy in the Natural History of Diabetes

Stanley Schwartz, MD

Affiliate, Main Line Health System, Ardmore, PA; Emeritus, Clinical Associate Professor of Medicine, University of Pennsylvania, Philadelphia, PA

Correspondence: Stanley Schwartz, MD,233 E Lancaster Ave,Ardmore, PA 19003.Tel: 610-642-6800Fax: 610-642-6850E-mail: [email protected]

DOI:

Abstract: The incretin class of anti-hyperglycemic agents, including glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-inhibitors, is an important addition to the therapeutic armamentarium for the management of appropriate patients with type 2 diabetes mellitus as an adjunct to diet and exercise and/or with the agents metformin, sulfonylureas, thiazolidinediones, or any combination thereof. More recently, US Food and Drug Administration (FDA)-approved indications for incretins were expanded to include use with basal insulin. This review article takes an evidence-based practice approach in discussing the importance of aggressive treatment for diabetes, the principles of incretin physiology and pathophysiology, use of glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors, and patient types and contexts where incretin therapy has been found beneficial, from metabolic syndrome to overt diabetes.

Keywords: diabetes; dipeptidyl peptidase-4 inhibitors; glucagon-like peptide-1 receptor ago-nists; incretin

IntroductionThe incretin class of anti-hyperglycemic agents is an important addition to the therapeutic armamentarium for the care of patients with type 2 diabetes mellitus (T2DM). Incretin-based therapies have been available since 2005 with the initial launch of glucagon-like peptide-1 (GLP-1) receptor agonists, followed by the launch of the dipeptidyl pepti-dase-4 (DPP-4) inhibitors. Although these agents were initially approved as an adjunct to diet and exercise and/or metformin, sulfonylureas, thiazolidinediones (TZDs), or any therapeutic combination of these agents in patients with T2DM, indications for incretin therapy have been recently expanded to include their use with basal insulin.

This review takes a broad view of the use of GLP-1 receptor agonists and DPP-4 inhibitors based on the concept of evidence-based practice, the philosophy that takes the principles of evidence-based medicine and combines them with patient-based experience, clinical expertise, expert opinions, and existing guidelines to treat any individual patient in the optimal fashion.1,2 The review provides a rationale for the need to treat patients aggressively, discusses principles of incretin physiology and pathophysiology, the incretin medications themselves (similarities and differences), and patient types and contexts where incretin therapy has proven to be useful, from metabolic syndrome (MetS) to overt diabetes.

Materials and MethodsThe author’s clinical experience and expertise were used as the foundation for the article, supplemented by supportive literature related to the topic. Evidence-based

SCHWARTZ_proof

Page 2: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

Stanley Schwartz

2 © Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]

Warning: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

guidelines and an evidence-based practice approach are used for the recommendations.

Treatment RationaleToday’s diabetes epidemic correlates directly with the obe-sity epidemic in the United States and worldwide. Estimates from 2010 indicated that 25.6 million (11.3%) US adults have diabetes, with an additional 79 million (35%) with prediabetes.3 The hyperglycemia of diabetes leads directly to complications that can be present well before a patient is diagnosed with overt diabetes. Both hyperglycemic spikes (ie, variability) and continuous glycemic elevations result in acute and chronic toxicity, which leads to both micro- and macrovascular complications.4–7 The presence of hypergly-cemia, especially previously unrecognized hyperglycemia, correlates directly with adverse patient outcomes in the hospital setting.8

Data from meta-analyses of large prospective, random-ized trials show that in patients with T2DM, both early and late in the course of disease, aggressive glycemic control can reduce both micro- and macrovascular complications.9 Confusion has arisen from apparent negative results in the Action to Control Cardiovascular Risk in Diabetes (ACCORD) study,10 the Action in Diabetes and Vascular Dis-ease: Preterax and Diamicron Modified Release Controlled Evaluation (ADVANCE) trial,11 and the Veterans Affairs Diabetes Trial (VADT).12 In the ACCORD trial (N = 10 251), intensive therapy (target glycated hemoglobin [HbA1c] level , 6.0%) increased patient mortality and did not signifi-cantly reduce major cardiovascular (CV) events compared with standard therapy (target HbA1c 7.0% to 7.9%). Patients who were older, with longer duration of diabetes, and with existing CV comorbidities had undue hypoglycemia and weight gain—results arguably accounting for the inability to reduce CV outcomes and increased patient mortality with aggressive treatment to lower HbA1c levels , 6.0% using hypoglycemia agents.10 It should be no surprise given that patient weight gain increases risk of CV disease and mortal-ity, and that weight loss reduces cardiometabolic risk factors in patients with T2DM.13,14 Additionally, hypoglycemia has many negative CV consequences.15–26 In the ADVANCE trial (N = 11 140), intensive glucose control, defined as the use of gliclazide plus other drugs to achieve HbA1c level , 6.5%, resulted in a 10% relative reduction in the combined outcome of major macro- and microvascular events, primarily as a consequence of a 21% relative reduction in nephropathy, compared with standard glucose control. There were no significant differences between types of glucose control used

on patient major macrovascular events, mortality from CV causes, or mortality from any cause. Severe hypoglycemia was uncommon, but was more prevalent in the intensive-control group compared with the standard-control group.11 In the VADT trial (N = 1791), intensive therapy (absolute reduction of 1.5% in HbA1c level compared with standard therapy group) in patients with poorly controlled T2DM had no significant effect on rates of major CV events, mortality, or microvascular complications.12 Thus, aggressive treatment is warranted, but agents that cause hypoglycemia and weight gain should be avoided, which is the logic for use of the new incretin classes of agents, which are either weight neutral or result in weight loss for patients, with no undue hypoglycemic risk in the absence of concurrent hypoglycemic agents.

Natural History of T2DMType 2 diabetes mellitus is a chronic, progressive disorder that occurs in genetically susceptible individuals exposed to an environmental trigger favoring insulin resistance. The disease can be exacerbated by factors including obesity, mental illness, sleep disorders, exposure to second-hand smoke, and lack of physical activity. Prediabetes (defined by the presence of impaired fasting glucose level or impaired glucose tolerance) reflects patient risk for future development of diabetes, as well as CV disease, and is associated with obesity, dyslipidemia, and hypertension.27 Early intensifica-tion of T2DM management has been proven to minimize patient risk of developing long-term micro- and macrovas-cular complications. In addition to the core pathophysiologic defects in T2DM of insulin resistance in muscle and the liver, and abnormal β-cell function, other mechanisms of hyperg-lycemia include accelerated lipolysis of fat cells, decreased response to intestinal incretin hormones, hyperglucagonemia of the α-cell, increased glucose reabsorption in the kidney, and insulin resistance in the brain.28

The Role of Incretin-Based TherapiesThe potential of incretin therapy is observed through gluco-regulatory and other effects in the patient. Glucoregulatory effects include improvement in the incretin effect (by defi-nition) and the function of both α- and β-cells (especially a glucose-dependent release of insulin and suppression of glucagon) with both DPP-4 inhibitors and GLP-1 recep-tor agonists, as well as in reduction of insulin resistance in fat and muscle via direct and indirect effects on the liver. Additional actions and benefits of incretin therapy include reducing hepatic glucose output and weight reduction with

SCHWARTZ_proof

Page 3: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

Incretin-Based Therapy in Patients With Diabetes

© Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260 3ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

GLP-1 receptor agonists via inducing hypothalamic satiety and slowing the fast gastric emptying observed in patients with diabetes.

Glucoregulatory Incretin EffectsThe incretin effect was initially described in 1964.29 It was recognized that the β-cell secreted 4 times more insulin when glucose was given to metabolically normal subjects orally rather than intravenously. The phenomenon, termed the incretin effect (due to gut-derived hormones secreted in response to nutrient ingestion that potentiate insulin secretion from β-cells), was found to be diminished in patients with diabetes.30 The hormone with the most therapeutic poten-tial was identified as GLP-1.31,32 Secretion of GLP-1 may be reduced in patients with diabetes, but the most recent work suggests that GLP-1 resistance also exists and can be overcome by pharmacologic levels of the hormone.33,34 The key effects of GLP-1 are to produce glucose-dependent secretion of insulin (increasing both first- and second-phase insulin secretion) and suppress glucagon secretion with nutrient ingestion (with a low risk of hypoglycemia); it has been shown to normalize plasma glucose levels in patients with T2DM.35–38 Moreover, GLP-1 also has been shown to minimize suppression of insulin induced by therapeutic use of steroids and calmodulin inhibitors.39,40

Additional Actions and Benefits of IncretinsThe actions of GLP-1 extend beyond the glucoregulatory effects outlined. Additional benefits include slowing of the fast gastric emptying that exists in patients with T2DM, sup-pressing appetite in the hypothalamus, and multiple beneficial CV effects, as shown in clinical and preclinical studies.41,42 The reported CV effects included reduced total and low-density lipoprotein cholesterol (LDL-C), apolipoprotein B, and triglyceride levels; increased high-density lipoprotein cholesterol (HDL-C); reduced high-sensitivity C-reactive protein; lowered systolic and diastolic blood pressure; improved endothelial function in patients with T2DM who had established coronary heart disease (CHD); improved left ventricular (LV) function in patients with acute myocardial infarction (MI); enhanced sodium excretion;43 improved cardiomyocyte viability after ischemia-reperfusion injury; improved systolic function; peripheral vasodilation; and reduced vascular inflammation.41,44–49

Patients with acute MI with or without diabetes who received a 72-hour infusion of GLP-1 added to standard ther-apy after successful primary angioplasty showed significant

improvements in LV ejection fraction (LVEF; from 29 ± 2% to 39 ± 2%; P , 0.01 vs controls).46 In addition, GLP-1 infu-sion peri-MI reduced infarct size.46 Improvements in LVEF also were observed after MI in patients with chronic heart failure with or without diabetes who received GLP-1 infu-sion for 5 weeks in addition to standard therapy (from 21 ± 3% to 27 ± 3%; P , 0.01 vs controls).43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1 initiated 12 hours before coronary artery bypass graft surgery reduced the need for vasopressors and anti-arrhythmic agents, and provided significantly better glycemic control pre- and perioperatively (95 ± 3 vs 140 ± 10 mg/dL; P # 0.02 vs controls).50 In addition, GLP-1 recipients required 45% less insulin than controls to achieve the same glycemic control during the postoperative period.

Incretin-Based TherapiesAttempts to use GLP-1 as a therapeutic agent in patients with T2DM have been limited by its short half-life in circulation,51 which is largely due to its inactivation by the enzyme DPP-4.41 These findings led to the development of incretin mimetics: GLP-1 analogs that are resistant to DPP-4 degradation, and DPP-4 inhibitors, agents that inhibit the proteolytic cleavage and inactivation of native GLP-1.41,51 The DPP-4 inhibitors increase concentration of endogenous GLP-1 approximately 2- to 4-fold, whereas GLP-1 receptor agonists, via their homology to native GLP-1, increase its effect 8- to 10-fold,52 with resultant different effects of the 2 classes of agents. Orally administered DPP-4 inhibitors improve the incretin effect and glucose-dependent secretion of insulin, as well as the suppression of glucagon with nutrient ingestion,52–54 which results in a low risk of hypoglycemia.55–58 In addition to these benefits, injectable GLP-1 receptor agonists decrease caloric intake and result in patient weight loss by slowing down the fast gastric emptying observed in patients with T2DM and by suppressing appetite through effect on the hypothalamus.52–54

The primary principle in choosing the right class of incretin is matching the right drug with the right patient and vice versa. The choice of agent should be based on individualized comorbidities, compliance issues, and patient administration preference – injection (once or twice daily, or once weekly; needle size) or oral dosing – and all of the practical issues surrounding clinical judgment, along with use of a patient-centric approach, from matching the patient’s needs with regard to extent of reduction in HbA1c level and weight, to other effects achieved by the different incretin drug classes.27,59,60

SCHWARTZ_proof

Page 4: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

Stanley Schwartz

4 © Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]

Warning: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

Table 1 summarizes key studies and reductions in patient HbA1c levels per US Food and Drug Administration (FDA)-approved agents currently available, including the DPP-4 inhibitors linagliptin,61–66 saxagliptin,67–72 sitaglip-tin,55,73–76 and alogliptin77–81; and the GLP-1 receptor agonists exenatide twice daily,82–87 exenatide once weekly,88–92 and liraglutide.93–98 As Bloomgarden et al99 noted for antihyper-glycemic agents in general, the higher the baseline HbA1c level, the greater the reduction in HbA1c level when incretins are used.100 An interesting exception occurs when sodium-glucose cotransporter-2 (SGLT-2) inhibitors are added to both incretin classes of agents. Monotherapy efficacy is preserved completely with regard to patient glycemic benefit and weight loss,101 presumably because incretins reduce the compensatory increase in hepatic glucose production induced by SGLT-2 inhibition.102 The incretin therapies are effective when added to existing regimens, such as metformin and/or pioglitazone; and although incretin therapies have proven effective when added on to sulfonylureas, the combination can be associated with hypoglycemia – thus, incretins are used to discontinue sulfonylureas and glinides in patients, a major focus of our evidence-based practice approach in the use of incretins.

The weight neutrality of the DPP-4 inhibitors and the weight loss associated with the GLP-1 receptor agonists are important considerations for the approach. The DPP-4 inhibi-tors are weight neutral. Very importantly, the GLP-1 receptor agonists are associated with mean weight reductions whether used as monotherapy or in combination therapy,103 an effect that does not appear due to the nausea that some patients experience on GLP-1 receptor agonist therapy.95,104 The weight-loss effect is durable if the agent is continued.91,105

Tolerability and Notable Adverse EventsHypoglycemia is infrequent and severe hypoglycemia is rare when using DPP-4 inhibitors and GLP-1 receptor agonists,91,106 in contrast to sulfonylureas.55–58

Cardiovascular safety is a major focus of the evidence-based practice approach in the use of incretins. All the incretins have met the less stringent CV safety requirements instituted by the FDA in 2008.107 Saxagliptin and alogliptin have demonstrated CV safety in large prospective, random-ized, controlled trials reported in 2013108,109 and have met more stringent FDA criteria, while CV outcomes trials are in progress for other incretin agents. A meta-analysis of data with DPP-4 inhibitors110 suggested a modest reduction in CV events. The mechanism for the CV effects of DPP-4 inhibi-tors through modulation of endothelial progenitor cells,

the inflammatory pathway, and the ischemic response, was reviewed by Fadini and Avogaro.111 Sitagliptin has been shown to have cardioprotective effects on ischemic segments and mitigated post-ischemic stunning in patients undergoing stress testing.112 A retrospective database analysis of claims during a 4-year period showed that patients treated with exenatide twice daily were 19% less likely to have a CV event than patients treated with other glucose-lowering agents.113

The most common adverse effects of DPP-4 inhibitors are upper respiratory tract infections, nasopharyngitis, and headache.114–117 The most common adverse effects of GLP-1 receptor agonists include nausea, vomiting, diarrhea, head-ache, and injection site reactions.118–120

In 3 placebo-controlled, 30-week clinical trials of exenatide twice daily as an add-on to metformin and/or sulfonylurea, the most common adverse events occurring in patients receiv-ing exenatide (N = 963) compared with placebo (N = 483) were nausea (44% vs 18%), vomiting (13% vs 4%), and diarrhea (13% vs 6%).118 Of note, in a 2008, 24-week study of exenatide twice daily as monotherapy showed the rate of nausea to be 3% with exenatide 5 mcg (2 of 77 patients) and 13% with exenatide 10 mcg (10 of 78 patients).86 The lower nausea rates may have occurred because patients learned how to use exenatide and to stop eating when full. Moreover, the incidence of nausea with exenatide once weekly 2 mg (9% of 461 patients) was nearly one-third that with liraglutide 1.8 mg daily (21% of 450 patients) in a 26-week open-label study.89 Nausea decreases over time with both exenatide and liraglutide use.121–125 Some patients on GLP-1 receptor agonists have undue hypothalamic sensitivity resulting in nausea, even at low doses. It can account for the 1% dropout rates in many studies,86,89,118,121–125 but may be treated with metoclopramide or ondansetron.126

In patients exposed to GLP-1 receptor agonists, slower gastric emptying seems to be the cause of gastrointestinal upset, nausea, and even vomiting, especially if the patient continues eating after the first sense of fullness, has a high-fiber intake, consumes high-fat meals, or eats too quickly. Thus, patients should be taught that they will lose their appetite between meals and should not eat out of habit, to stop eating at the first sense of fullness, eat slowly, and avoid high-fat and very high-fiber meals. To reduce the risk of nausea, physicians may advise patients to slow the titration of exenatide twice daily118 and liraglutide,119 and administer exenatide twice daily closer to the start of a meal.118

With regard to renal safety with use of incretin-based therapies, there is no evidence of direct nephrotoxicity,114,118,119,127,128 but worsening renal function

SCHWARTZ_proof

Page 5: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

Incretin-Based Therapy in Patients With Diabetes

© Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260 5ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

Table 1. Reduction in HbA1c Levels in Key Monotherapy and Combination-Therapy Studies With GLP-1 Receptor Agonists and DPP-4 Inhibitors

Agent Study

N Duration, Weeksa Treatments Baseline HbA1c Level, %

HbA1c Change, (- or +)%

P Value vs Placebob

AlogliptinDeFronzo et al, 200877 329 26 Alogliptin 25 mg QD 7.9 -0.59 , 0.001

Placebo -0.02 –Pratley et al, 200979 500 26 Alogliptin 25 mg QD 8.1 -0.53 , 0.001

Placebo 8.2 +0.01 –Nauck et al, 200980 527 26 (add-on to metformin) Alogliptin 12.5 mg QD 7.9 -0.6 , 0.001

Alogliptin 25 mg QD 7.9 -0.6 , 0.001Placebo 8.0 -0.1 –

Pratley et al, 200978 493 26 (add-on to pioglitazone) Alogliptin 25 mg QD 8.0 -0.80 , 0.001Placebo 8.0 -0.19 –

Rosenstock et al, 201081 655 26 Alogliptin 25 mg QD 8.80 -0.96 –Pioglitazone 30 mg QD 8.76 -1.15 –

Alogliptin 25 mg QD + pioglitazone 30 mg QD

8.80 -1.71 , 0.05 vs each component alone

LinagliptinDel Prato et al, 201161 503 24 Linagliptin 5 mg QD 8.00 -0.44 , 0.0001

Placebo 8.00 +0.25 –Haak et al, 201264 791 24 Linagliptin 5 mg QD 8.7 -0.5 , 0.0001

Metformin 500 mg BID 8.7 -0.6 , 0.0001Metformin 1000 mg BID 8.5 -1.1 , 0.0001Linagliptin 2.5 mg BID + metformin 500 mg BID

8.7 -1.2 , 0.0001 vs placebo; metformin 500 mg alone

Linagliptin 2.5 mg BID + metformin 1000 mg BID

8.7 -1.6 , 0.0001 vs placebo; metformin 1000 mg alone

Placebo 8.7 +0.1 –Gomis et al, 201163 389 24 Pioglitazone 30 mg QD +

linagliptin 5 mg QD8.60 -1.06 , 0.0001

Pioglitazone 30 mg QD + placebo

8.58 -0.56 –

Taskinen et al, 201166 700 24 (add-on to metformin) Linagliptin 5 mg QD 8.09 -0.49 , 0.0001Placebo 8.02 +0.15 –

Gallwitz et al, 201262 1551 2 years (add-on to metformin)

Linagliptin 5 mg QD 7.7 -0.16 0.0004 vs glimepirideGlimepiride 1–4 mg QD 7.7 -0.36 –

Owens et al, 201165 1055 24 (add-on to metformin + sulfonylurea)

Linagliptin 5 mg QD 8.2 -0.72 , 0.0001Placebo 8.1 -0.10 –

SaxagliptinRosenstock et al, 200971 401 24 Saxagliptin 2.5 mg QD 7.9 -0.43 , 0.0001

Saxagliptin 5 mg QD 8.0 -0.46 , 0.0001Placebo 7.9 +0.19 –

Jadzinsky et al, 200970 1306 24 Saxagliptin 5 mg QD + metformin 250–1000 mg BID

9.4 -2.5 , 0.0001 vs metformin alone

Metformin 250–1000 mg BID + placebo

9.4 -2.0 –

DeFronzo et al, 200968 743 24 (add-on to metformin) Saxagliptin 2.5 mg QD 8.1 -0.59 , 0.0001Saxagliptin 5 mg QD 8.1 -0.69 , 0.0001Placebo 8.1 +0.13 –

Scheen et al, 201072 801 18 (add-on to metformin) Saxagliptin 5 mg QD 7.7 -0.52 NRSitagliptin 100 mg QD 7.7 -0.62 NR

(Continued)

SCHWARTZ_proof

Page 6: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

Stanley Schwartz

6 © Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]

Warning: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

Table 1. (Continued)

Agent Study

N Duration, Weeksa Treatments Baseline HbA1c Level, %

HbA1c Change, (- or +)%

P Value vs Placebob

Chacra et al, 200967 768 24 Saxagliptin 2.5 mg QD + glyburide 3.75 mg BID

8.4 -0.54 , 0.0001 vs glyburide alone

Saxagliptin 5 mg + glyburide 3.75 mg BID

8.5 -0.64 , 0.0001 vs glyburide alone

Glyburide 5–7.5 mg BID 8.4 +0.08 –Hollander et al, 200969 565 24 (add-on to TZD) Saxagliptin 2.5 mg QD 8.3 -0.66 0.0007

Saxagliptin 5 mg QD 8.4 -0.94 , 0.0001Placebo 8.2 -0.30 –

SitagliptinNauck et al, 200755 1172 52 (add-on to metformin) Sitagliptin 100 mg QD 7.5 -0.67 NR

Glipizide 5–20 mg/dc 7.5 -0.67 NRGoldstein et al, 200775 1091 24 Sitagliptin 100 mg QD 8.87 -0.66 , 0.001

Metformin 500 mg BID 8.90 -0.82 , 0.001Metformin 1000 mg BID 8.68 -1.13 , 0.001Sitagliptin 50 mg BID + metformin 500 mg BID

8.79 -1.40 , 0.001 vs placebo; vs each component alone

Sitagliptin 50 mg BID + metformin 1000 mg BID

8.76 -1.90 , 0.001 vs placebo; vs each component alone

Placebo 8.68 +0.17 –Charbonnel et al, 200673

701 24 (add-on to metformin) Sitagliptin 100 mg QD 7.96 -0.67 , 0.001Placebo 8.03 -0.02 –

Vilsbøll et al, 201076 641 24 (add-on to insulin) Sitagliptin 100 mg QD 8.7 -0.6 , 0.001Placebo 8.6 0.0 –

Dobs et al, 201374 278 54 (add-on to rosiglitazone + metformin)

Sitagliptin 100 mg QD 8.8 -1.1 , 0.001Placebo 8.7 -0.3 –

Exenatide Twice DailyMoretto et al, 200886 232 24 Exenatide 5 mcg BID 7.9 -0.7 0.003

Exenatide 10 mcg BID 7.8 -0.9 , 0.001Placebo 7.8 -0.2 –

DeFronzo et al, 200583 336 30 (add-on to metformin) Exenatide 5 mcg BID 8.3 -0.40 , 0.001Exenatide 10 mcg BID 8.2 -0.78 , 0.001Placebo 8.2 +0.08 –

Buse et al, 200482 377 30 (add-on to sulfonylurea) Exenatide 5 mcg BID 8.5 -0.46 , 0.001Exenatide 10 mcg BID 8.6 -0.86 , 0.001Placebo 8.7 +0.12 –

Zinman et al, 200787 233 16 (add-on to a TZD ± metformin)

Exenatide 10 mcg BID 7.89 -0.89 , 0.001Placebo 7.91 +0.09 –

Kendall et al, 200585 733 30 (add-on to metformin + sulfonylurea)

Exenatide 5 mcg BID 8.5 -0.55 , 0.0001Exenatide 10 mcg BID 8.5 -0.77 , 0.0001Placebo 8.5 +0.23 –

Heine et al, 200584 551 26 (add-on to metformin + sulfonylurea)

Exenatide 10 mcg BID 8.2 -1.1 NRTitrated insulin glargine QD

8.3 -1.1 NR

Exenatide Once WeeklyRussell-Jones et al, 201292

820 26 Exenatide 2 mg QW 8.5 -1.53 –Metformin 2000 mg/dc -1.48 0.620 vs exenatidePioglitazone 45 mg/dc -1.63 0.328 vs exenatideSitagliptin 100 mg/dc -1.15 , 0.001 vs exenatide

Bergenstal et al, 201088 491 26 (add-on to metformin) Exenatide 2 mg QW 8.6 -1.5 –Sitagliptin 100 mg QD 8.5 -0.9 , 0.0001 vs exenatidePioglitazone 45 mg QD 8.5 -1.2 0.0165 vs exenatide

(Continued)

SCHWARTZ_proof

Page 7: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

Incretin-Based Therapy in Patients With Diabetes

© Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260 7ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

Table 1. (Continued)

Agent Study

N Duration, Weeksa Treatments Baseline HbA1c Level, %

HbA1c Change, (- or +)%

P Value vs Placebob

Diamant et al, 201090 456 26 (add-on to metformin ± sulfonylurea)

Exenatide 2 mg QW 8.3 -1.5 0.017 vs insulin glargineTitrated insulin glargine QD

8.3 -1.3 –

Drucker et al, 200891 295 30 Exenatide 2 mg QW 8.3 -1.9 0.0023 vs exenatide BIDExenatide 10 mcg BID 8.3 -1.5 –

Buse et al, 201389 911 26 (add-on to oral agents) Exenatide 2 mg QW 8.5 -1.28 –Liraglutide 1.8 mg QD 8.4 -1.48 0.02 vs exenatide

LiraglutideGarber et al, 200993 746 52 Liraglutide 1.2 mg QD 8.3 -0.84 0.0014 vs glimepiride

Liraglutide 1.8 mg QD 8.3 -1.14 , 0.0001 vs glimepirideGlimepiride 8 mg QD 8.4 -0.51 –

Nauck et al, 200995 1087 26 (add-on to metformin) Liraglutide 1.2 mg QD 8.3 -1.0 NRLiraglutide 1.8 mg QD 8.4 -1.0 NRGlimepiride 4 mg QD 8.4 -1.0 NRPlacebo 8.4 +0.1 NR

Pratley et al, 201096 658 26 (add-on to metformin) Liraglutide 1.2 mg QD 8.4 -1.24 , 0.0001 vs sitagliptinLiraglutide 1.8 mg QD 8.4 -1.50 , 0.0001 vs sitagliptinSitagliptin 100 mg QD 8.5 -0.90 –

Marre et al, 200994 1041 26 (add-on to glimepiride) Liraglutide 1.2 mg QD 8.5 -1.1 , 0.0001 vs placebo; vs rosiglitazone

Liraglutide 1.8 mg QD 8.5 -1.1 , 0.0001 vs placebo; vs rosiglitazone

Rosiglitazone 4 mg QD 8.4 -0.4 , 0.0001Placebo 8.4 +0.2 –

Zinman et al, 200998 533 26 (add-on to metformin + rosiglitazone)

Liraglutide 1.2 mg QD 8.5 -1.5 , 0.0001Liraglutide 1.8 mg QD 8.6 -1.5 , 0.0001Placebo 8.4 -0.5 –

Russell-Jones et al, 200997

576 26 (add-on to metformin and glimepiride)

Liraglutide 1.8 mg QD 8.3 -1.33 , 0.0001 vs placebo; 0.0015 vs insulin glargine

Insulin glargine 8.2 -1.09 , 0.0001Placebo 8.3 -0.24 –

aMeasured in weeks unless otherwise noted. bCompared with placebo unless otherwise noted. cFrequency of dosing not reported. Abbreviations: BID, twice daily; DPP-4, dipeptidyl peptidase-4; GLP-1, glucagon-like peptide-1; HbA1c, glycated hemoglobin; NR, not reported; QD, once daily; QW, once weekly; TZD, thiazolidinedione.

has been reported in some individuals taking exenatide, liraglutide, or sitagliptin.114–116,118–120,129,130 Exenatide should not be used in patients with severe renal impairment or end-stage renal disease, and should be used with caution in patients with renal transplantation,118,120 while liraglutide should be used with caution in patients with renal impair-ment.119 Assessment of renal function is recommended prior to initiating sitagliptin and alogliptin and periodically thereafter; dose adjustment is recommended in patients with moderate-to-severe renal insufficiency and in patients with end-stage renal disease.114,117 No dose adjustments are indicated for patients with renal impairment taking linagliptin.116 With saxagliptin therapy, the dose should be limited to 2.5 mg based on renal function, and assessment

is recommended before initiation of saxagliptin and periodically thereafter.115

Both longer duration formulations of GLP-1 receptor agonists carry black-box warnings for the risk of medullary thyroid cancer118,119; however, this was observed in rodents and not humans, and 3 studies have shown no GLP-1 recep-tors on human C-cells.131–133 Nevertheless, in accordance with prescribing information, a personal or family history of medullary thyroid cancer or multiple endocrine neoplasia syndrome type 2 in patients should be considered contraindi-cations for liraglutide and exenatide once weekly use based on the findings in rodents.119,120 Routine calcitonin and/or ultrasound testing is not recommended, but referral to an endocrinologist is recommended for patients with thyroid

SCHWARTZ_proof

Page 8: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

Stanley Schwartz

8 © Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]

Warning: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

nodules or elevated serum calcitonin levels identified for other reasons.

Pancreatitis has been reported with use of DPP-4 inhibi-tors and GLP-1 receptor agonists, and warnings exist on all incretin labels – specifically, they should not be prescribed to patients with a history of pancreatitis. Incretin-based therapies should be discontinued if signs and symptoms arise, and in this regard, it is important to educate patients on the signs and symptoms of pancreatitis. Therapy should not be resumed if pancreatitis is confirmed.114–120,134,135

Current evidence, however, does not support a causal rela-tionship between incretin-based therapies and pancreatitis. A large database analysis of patients taking exenatide twice daily and sitagliptin showed no increased risk compared with patients taking metformin or glyburide.136 Although there was a higher incidence of pancreatitis in liraglutide-treated patients than in those on an active comparator, it was smaller than expected given that patients with diabetes have a 1.5- to 3-fold higher risk of developing pancreatitis than nondiabetic patients.119,137 Furthermore, in the Saxagliptin Assessment of Vascular Outcomes Recorded in Patients with Diabetes Mel-litus (SAVOR) study, pancreatitis occurred infrequently, and the incidence of acute or chronic pancreatitis was similar in patients receiving saxagliptin or placebo.109 A recent review of the data by the FDA and European Medicines Agency (EMA) found no undue risk of pancreatitis or pancreatic cancer, and recommended continued scrutiny and mainte-nance of current warnings.138

Clinical Applications for Incretin-Based TherapiesBased on the characteristics of GLP-1 and the incretin drug classes discussed, it becomes evident how and for which clinical applications one might use incretin-based therapies across the natural history of the MetS and T2DM, and even for many patients with type 1 diabetes mellitus (T1DM) and latent autoimmune diabetes in adults (LADA)139 (Figure 1).

Clinical Study DataIncretin therapies have proved effective in treating late-postprandial hypoglycemia in patients at risk of developing or who already have MetS due to loss of or decreased first-phase insulin secretion, resulting in slightly higher 1-hour postpran-dial blood glucose levels and higher initial insulin levels. As a result of loss of first-phase insulin release, a disproportion of blood glucose/insulin concentration 4 to 5 hours after eating is believed to result in hypoglycemia, with classic catechol

symptoms, and occasionally, in neuroglycopenic symptoms. These patients are usually put on a no-concentrated-sweets diet, but some patients still have symptoms. Almeda-Valdes et al140 showed that patients with postprandial hypoglycemia treated with sitagliptin had significant improvement due to increased first-phase insulin release. In clinical experience, the same effect has been observed with the DPP-4 inhibitors and GLP-1 receptor agonists.

Next along the MetS treatment continuum are the GLP-1 receptor agonists, which generally reduce hypoglycemia (although concomitant use of insulin or insulin secretagogues may increase risk of hypoglycemia) and also promote weight loss. In a meta-analysis of 8 studies conducted in adults with T2DM, significant weight loss was observed with GLP-1 receptor agonist therapy versus comparators (weighted mean difference of –2.37 kg [95% CI, –3.95 to –0.78]), with a more pronounced difference when exenatide was compared with insulin (2 studies: –4.76 kg [95% CI, –6.03 to –3.49]).141 Weight loss with use of exenatide twice daily has been shown to be durable, with 84% of patients (182 of 217 patients) experiencing weight loss over 3 years.142 Treatment with exenatide twice daily for 16 weeks resulted in 77% of patients (229 of 299 patients) with MetS losing an average of 6% of their body weight.143 In a group of obese nondiabetic patients, 64% of the 186 patients receiving 2.4 or 3 mg of liraglutide once daily lost . 5% of their body weight over 1 year, and . 85% maintained this level of weight loss at 2 years.144

In a pilot study by Armato et al,145 56% of 25 patients with impaired fasting plasma glucose levels and 59% of 22 patients with impaired glucose tolerance returned to normal glucose tolerance when treated with exenatide twice daily plus pioglitazone and metformin. In the Diabetes Prevention Program Outcomes Study (N = 1990), the incidence of overt diabetes in patients with prediabetes was reduced 50% to only 5% per year; and, in those patients who had achieved normal glucose regulation, the incidence of future overt diabetes was only 3% per year.146

Both DPP-4 inhibitors and GLP-1 receptor agonists have demonstrated durability of glycemic control over 2 to 3 years.56,142,147–151 With exenatide, results are likely due to improved β-cell function, as evidenced by increased C-peptide secretion in patients during hyperglycemic clamp studies fol-lowing 1 year of therapy with exenatide twice daily.152 The results can be contrasted to the marked monotherapy failure rates observed with sulfonylureas after 1 year of use.153–158

Many commentators contrast the low cost of individual doses of sulfonylureas with the higher cost of incretin

SCHWARTZ_proof

Page 9: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

Incretin-Based Therapy in Patients With Diabetes

© Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260 9ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

classes of drugs, however, focusing solely on drug costs fails to recognize the greater expenses associated with older medications for increased patient emergency room visits, hospitalizations, mortality, under-recognized hypoglycemic unawareness, lifestyle restrictions, and self-monitoring of blood glucose testing, as well as diminished quality of life, worry for spouse, friends, and coworkers, fear of hypoglyce-mia leading to inadequate glycemic control, and severe hypo-glycemia increasing the risk of dementia.159–161 In addition, given increased apoptosis (death) of β-cells with sulfonylurea use, patients will need more expensive drugs in 1 to 3 years, but with the disadvantage of having lost β-cell mass. In fact, various comparisons of sulfonylureas with incretin therapies have shown incretins to be cost-effective.162–168

Guideline RecommendationsThe 2012 ADA guidelines note DPP-4 inhibitors and GLP-1 receptor agonists as therapeutic options,59 but sulfonylureas maintain a prominent role in the treatment paradigm, even as the guidelines advocate the importance of avoiding hypo-glycemia and weight gain in patients with T2DM. There is concern regarding greater patient mortality associated with higher exposure to sulfonylureas169 and acute coronary events170 shown in large drug database studies,170–172 which may be contributed to by the loss of ischemic precondition-ing observed with use of most sulfonylureas.173 In contrast,

the incretin classes of therapy have shown CV safety and possible CV benefits, as discussed.

Early combination therapy of the incretin classes with other antidiabetic agents is supported, given the efficacy of such regimens and the known tissue damage caused by the mechanisms of complications of diabetes that can accrue with less aggressive treatment.174 In addition, combination therapy is supported by complementary mechanisms of action. There is a synergy with co-administration of DPP-4 inhibitors and metformin,66,68,175,176 as metformin increases endogenous active GLP-1 levels.177 In addition, there is a particular syn-ergy with co-administration of DPP-4 inhibitors and GLP-1 receptor agonists with SGLT-2 inhibitors,101 as the increase in hepatic glucose production induced by SGLT-2 inhibitors is counteracted by reduced hepatic glucose production with use of DPP-4 inhibitors and GLP-1 receptor agonists.178

Recent data presented by Abdul-Ghani et al179 dem-onstrated that early, triple antidiabetic therapy with met-formin, pioglitazone, and exenatide twice daily, targeting the core metabolic defects responsible for hyperglycemia (insulin resistance and β-cell dysfunction) is more effica-cious and better tolerated than therapy aimed simply at lowering patient plasma glucose concentration (escalating metformin, followed by sequential addition of glipizide, and then basal insulin), without correcting the underlying pathophysiologic disturbances present in T2DM, such as

Figure 1. Role of incretins in the continuum of DM, based on the author’s clinical experience.

Abbreviations: AACE, American Academy of Clinical Endocrinologists; DM, diabetes mellitus; DPP-4, dipeptidyl peptidase-4; GLP-1 RA, glucagon-like peptide-1 receptor agonist; hypo, hypoglycemia; LADA, latent autoimmune diabetes in adults; PIO, pioglitazone; SU, sulfonylurea.

SCHWARTZ_proof

Page 10: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

Stanley Schwartz

10 © Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]

Warning: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

step-by-step addition to metformin of sulfonylureas and insulin.

Additional concerns with regard to the 2012 ADA guide-lines were recently published.180 The guidelines recommend less aggressive HbA1c level goals in patients who are older, have a longer duration of diabetes, and have important comor-bidities59 (based upon the ACCORD, VADT, and ADVANCE trials, in which aggressive sulfonylurea and insulin use was associated with hypoglycemia and weight-gain complica-tions). The guidelines now recognized that there are 9 classes of antidiabetic agents, including the DPP-4 inhibitors and GLP-1 receptor agonists, which are unlikely to cause hypo-glycemia and, in my clinical opinion, allow clinicians to be as aggressive in these patients as in those who are younger, have a shorter duration of diabetes, and have no significant comorbidities.

The 2013 American Association of Clinical Endocri-nologists/American College of Endocrinology (AACE/ACE) pharmacologic guidelines update,60 encompassing the broader care of patients with MetS through overt diabetes, recommends early use of combination therapy, consisting of metformin as the cornerstone of dual therapy for most patients (when metformin is contraindicated, TZDs may be used) and a GLP-1 receptor agonist or DPP-4 inhibitor. Additional combinations include metformin plus a TZD, an SGLT-2 inhibitor, basal insulin, colesevelam, bromocrip-tine-QR, or an alpha-glucosidase inhibitor. The insulin secretagogues (sulfonylureas or glinides) can be used in combination with metformin as a last dual therapy treatment option. Furthermore, the guidelines recommend choosing the right drug for the right patient and, most importantly, with regard to avoidance of hypoglycemia and weight gain in patients, the early use of DPP-4 inhibitors and GLP-1 receptor agonists.

Clinical Practice ExperienceIn my clinical practice experience, incorporation of additional principles into the diabetes-management approach maximizes control of as many mechanisms of hyperglycemia as possible, especially combining agents that address fasting and post-prandial blood glucose levels, β-cell secretion, and insulin resistance.28 Measures include use of 3 to 4 non-insulin agents before considering insulin; not using sulfonylureas/glinides; incorporating the 2013 AACE/ACE first-tier/second-tier prin-ciple of using the most effective agents, ideally metformin, GLP-1 receptor agonists, DPP-4 inhibitors, alpha-glucosidase inhibitors, and now, SGLT-2 inhibitors. For patients who cannot take $ 1 of these agents, using second-line agents,

such as pioglitazone, bromocriptine-QR, or colesevelam;60 combining agents that target or improve β-cell function, directly or indirectly, such as incretins/SGLT-2 inhibitors, pioglitazone; agents that improve insulin resistance, such as pioglitazone/metformin/bromocriptine-QR; and using insulin only if the patient is following a no-concentrated-sweets diet (otherwise the patient is positioned for weight gain and hypo-glycemia). Patients are kept on non-insulin treatments when they start basal insulin. If the patient is already on insulin and is willing to follow a no-concentrated-sweets diet, the current insulin dose can be decreased ∼25% for agreeing to follow the diet; it can also be decreased an additional ∼25% if hypoglycemic episodes are occurring. The recalculated dose can be decreased ∼25% if an SGLT-2 inhibitor is added and ∼25% if a GLP-1 receptor agonist is added to the therapeutic regimen. Thus, when patients are on , 40 units of insulin daily, without a restrictive diet and are experiencing hypo-glycemic episodes, insulin treatment can be discontinued, as a no-concentrated-sweets diet is started along with a GLP-1 receptor agonist and/or SGLT-2 inhibitor. For patients on greater amounts of insulin, the same proportionate reductions in insulin and down-titrations are recommended initially; and as the patient loses weight, insulin therapy can be further decreased, and potentially stopped in many patients.181–185

Weight reduction is an important clinical outcome in disease management. Incretin therapies are used in patients with MetS, as well as in those with diabetes.144,186,187 Treat-ments include using SGLT-2 inhibitors with GLP-1 recep-tor agonists to maximize weight loss;101 using incretins before pioglitazone to prevent weight gain87,165; using SGLT-2 inhibitors with pioglitazone to reduce edema-related weight (personal observation); using GLP-1 receptor agonists as the preferred therapy over DPP-4 inhibitors in the “right patient”; using GLP-1 receptor agonists always before initiating insulin unless the patient is “sick” (ie, symptomatic);84,188,189 avoiding insulin if the patient is not following a no-concentrated-sweets diet; and keeping the patient on incretin/SGLT-2 inhibitor when adding basal insulin. Only ∼10% of patients need bolus insulins if non-insulin therapies are being administered as basal insulin therapy is initiated. Only analog insulins should be used per the AACE recommendations.60 The combination of incretin therapy with basal insulin is preferred, as 60% to 87% of patients on such a regimen achieve the ADA goal of HbA1c level , 7%.181,190–192 Use DPP-4 inhibitors when reductions of , 1.0% in HbA1c level are needed and use GLP-1 receptor agonists when reductions $ 1.0% are needed (patients may benefit from possible weight loss).183,193–195 Exenatide twice

SCHWARTZ_proof

Page 11: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

Incretin-Based Therapy in Patients With Diabetes

© Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260 11ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

Table 2. Reductions in Patient Body Weight in Key Monotherapy and Combination Therapy Studies With GLP-1 Receptor Agonists

Agent Study

N Duration, Weeks Treatments Baseline Body Weight, kg

Body Weight Change, (+ or -) kg

P Value (vs placeboa)

Exenatide Twice DailyMoretto et al, 200886 232 24 Exenatide 5 mcg BID 85 -2.8 0.004

Exenatide 10 mcg BID 86 -3.1 , 0.001Placebo 86 -1.4 –

DeFronzo et al, 200583 336 30 (add-on to metformin) Exenatide 5 mcg BID 100 -1.6 , 0.001Exenatide 10 mcg BID 101 -2.8 , 0.001Placebo 100 -0.3 –

Buse et al, 200482 377 30 (add-on to sulfonylurea) Exenatide 5 mcg BID 95 -0.9 NSExenatide 10 mcg BID 95 -1.6 , 0.05Placebo 99 -0.6 –

Zinman et al, 200787 233 16 (add-on to a TZD ± metformin)

Exenatide 10 mcg BID 97.5 -1.75 , 0.001Placebo 96.9 -0.24 –

Kendall et al, 200585 733 30 (add-on to metformin + sulfonylurea)

Exenatide 5 mcg BID 97 -1.6 # 0.001Exenatide 10 mcg BID 98 -1.6 # 0.001Placebo 99 -0.9 –

Heine et al, 200584 551 26 (add-on to metformin + sulfonylurea)

Exenatide 10 mcg BID 87.5 -2.3 , 0.0001 vs insulin glargineTitrated insulin glargine QD 88.3 +1.8 –

Exenatide Once WeeklyRussell-Jones et al, 201292 820 26 Exenatide 2 mg QW 87.0 -2.0 –

Metformin 2000 mg/db -2.0 0.892 vs exenatidePioglitazone 45 mg/db +1.5 , 0.001 vs exenatideSitagliptin 100 mg/db -0.8 , 0.001 vs exenatide

Bergenstal et al, 201088 491 26 (add-on to metformin) Exenatide 2 mg QW 89 -2.3 –Sitagliptin 100 mg QD 87 -0.8 0.0002 vs exenatidePioglitazone 45 mg QD 88 +2.8 , 0.0001 vs exenatide

Diamant et al, 201090 456 26 (add-on to metformin ± sulfonylurea)

Exenatide 2 mg QW 91.2 -2.6 , 0.0001 vs insulin glargineTitrated insulin glargine QD 90.6 +1.4 –

Drucker et al, 200891 295 30 Exenatide 2 mg QW 102 -3.7 0.89 vs exenatide BIDExenatide 10 mcg BID 102 -3.6 –

Buse et al, 201389 911 26 (add-on to oral agents) Exenatide 2 mg QW 90.9 -2.68 –Liraglutide 1.8 mg QD 91.1 -3.57 0.0005 vs exenatide

LiraglutideNauck et al, 200995 1087 26 (add-on to metformin) Liraglutide 1.2 mg QD NR -2.6 # 0.01 vs placebo; , 0.0001

vs glimepirideLiraglutide 1.8 mg QD NR -2.8 # 0.01 vs placebo; , 0.0001

vs glimepirideGlimepiride 4 mg QD NR +1.0 –Placebo NR -1.5 –

Pratley et al, 201096 658 26 (add-on to metformin) Liraglutide 1.2 mg QD 93.7 -2.86 , 0.0001 vs sitagliptinLiraglutide 1.8 mg QD 94.6 -3.38 , 0.0001 vs sitagliptinSitagliptin 100 mg QD 93.1 -0.96 –

Marre et al, 200994 1041 26 (add-on to glimepiride) Liraglutide 1.2 mg QD 80.0 +0.3 , 0.0001 vs rosiglitazoneLiraglutide 1.8 mg QD 83.0 -0.2 , 0.0001 vs rosiglitazoneRosiglitazone 4 mg QD 80.6 +2.1 –Placebo 81.9 -0.1 –

Zinman et al, 200998 533 26 (add-on to metformin + rosiglitazone)

Liraglutide 1.2 mg QD NR -1.0 , 0.0001Liraglutide 1.8 mg QD NR -2.0 , 0.0001Placebo NR +0.6 –

(Continued)

SCHWARTZ_proof

Page 12: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

Stanley Schwartz

12 © Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]

Warning: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

daily is approved for use with insulin glargine; however, exenatide once weekly is not. Liraglutide is approved for use with basal insulin analogs.

If an incretin is added to a patient already on insulin, it is likely that insulin doses will need to be lowered to avoid hypoglycemia. If needed, insulin doses can be up-titrated. When adding exenatide to insulin therapy, lower the basal insulin dose 10% and bolus insulin dose 50%. When adding liraglutide to insulin therapy, lower basal and bolus insulin doses 25% each. With the addition of exenatide once weekly, maximal effect is achieved over ∼6 weeks, which reflects the delayed effect of the extended-release formulation; thus, it will be necessary to down-titrate insulin doses gradually as glucose numbers drop over time. Sitagliptin, saxagliptin, linagliptin, and alogliptin are approved for concurrent use with insulin. When adding a DPP-4 inhibitor to basal insu-lin, reduce patient basal insulin dose 10%, and reduce bolus insulin dose 30%.

Of great interest, when GLP-1 receptor agonists are added to insulin therapy in patients with T1DM, they have also been found efficacious. Once insulin doses are adjusted, incretins reduce the dawn effect and reduce variability, brittleness, and rates of hypoglycemia simply due to the reduction in glucagon secretion. Moreover, some studies have shown improvement in HbA1c level and weight reduction in patients with T1DM.196–198 Similar effects are observed with the use of DPP-4 inhibitors.199,200 Treatment with a GLP-1 receptor agonist or DPP-4 inhibitor may be implemented in patients with T1DM depending on the need for weight reduction. A special case seems to be efficacy of incretins in treating patients with LADA, based on findings from a study of the DPP-4 inhibitor sitagliptin.139

The use of incretins is advocated in cases of stress and steroid diabetes, especially for patients in the hospital and those with new-onset diabetes after transplantation, to

exploit the benefits in reducing hypoglycemia caused by the reduction in insulin secretion resulting from use of steroids and calmodulin inhibitors.39,40,201

Additionally, incretins are effective in treating patients undergoing bariatric surgery. A GLP-1 receptor agonist is used preoperatively. In some patients, these agents may obvi-ate the need for surgery, especially when combined with an SGLT-2 inhibitor; however, in others, any weight loss prior to bariatric surgery makes the procedure easier. In patients undergoing bariatric surgery, incretins may be initiated post-operatively to avoid hypoglycemia, engendered by the use of sulfonylureas and insulin, as the patients will have rapid drops in plasma glucose initially and then have rapid weight loss on discharge. Incretins are also used in those who develop late-postprandial hypoglycemia after bariatric surgery, reminiscent of the same syndrome seen prior to developing prediabetes or diabetes (restoring first-phase insulin release).202

Lastly, with the advent of 9 antihyperglycemic agents that do not cause hypoglycemia, patient treatment goals for glyce-mic control and HbA1c level have changed. The ADA goal of HbA1c level , 7%, and even the AACE goal of HbA1c level # 6.5%, were previously generated to avoid hyperglycemia. Now clinicians can aim for the lowest HbA1c level possible without undue hypoglycemia, even in the elderly (in whom the appropriate caution was applied by the ADA in 2013, given the results of the ACCORD, VADT, and ADVANCE trials on the aggressive use of sulfonylureas and insulin). It seems very appropriate to do so given the greater survival in patients aged . 75 years with low HbA1c levels,203 as long as antihyperglycemic agents that do not cause hypoglycemia, such as the incretins, are used.

ConclusionsIn summary, patients with MetS and T2DM are treated aggressively because of the epidemic of obesity and

Table 2. (Continued)

Agent Study

N Duration, Weeks Treatments Baseline Body Weight, kg

Body Weight Change, (+ or -) kg

P Value (vs placeboa)

Russell-Jones et al, 200997 576 26 (add-on to metformin and glimepiride)

Liraglutide 1.8 mg QD 85.5 -1.8a 0.0001 vs placebo; , 0.0001 vs insulin glargine

Insulin glargine 85.0 +1.6 –Placebo 85.7 -0.42 –

aCompared with placebo unless otherwise noted. bFrequency of dosing not reported. Abbreviations: BID, twice daily; GLP-1, glucagon-like peptide-1; NR, not reported; NS, not significant; QD, once daily; QW, once weekly; TZD, thiazolidinedione.

SCHWARTZ_proof

Page 13: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

Incretin-Based Therapy in Patients With Diabetes

© Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260 13ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

diabetes, and the potential for adverse patient outcomes. The physiology and pathophysiology of the incretin agents have been reviewed, and it has been noted that incretin agents, which normalize and correct incretin pathophysiology, have pleotropic effects in reducing weight gain, causing weight loss, and reducing the frequency of hypoglycemia in patients; use of incretin therapy will likely also have a benefit on CV outcomes. The right drug for the right patient should be chosen, but incretin agents, in general, can be therapeutically applied in patients across the whole continuum of the natural history of MetS and diabetes.

Finally, in seeking an approach to diabetes and glycemic control that aims to reduce patient blood sugar levels as low as possible, as early as possible, for as long as possible, as safely as possible, and especially, in as rational a manner as possible – the incretin classes of agents now allow us to do so.

AcknowledgmentsSushma Soni and Lisa M. Klumpp Callan, PhD, CMPP, of inScience Communications, Springer Healthcare, provided medical writing support funded by AstraZeneca and Bristol-Myers Squibb Company.

Conflict of Interest StatementStanley Schwartz, MD, has participated in speakers bureaus for Boehringer Ingelheim/Eli Lilly and Company, Merck and Co., Inc., Novo Nordisk Inc., sanofi-aventis U.S., Takeda Pharmaceuticals North America, Inc., Janssen Pharmaceu-ticals, Inc., Santarus, Inc., Amylin Pharmaceuticals, LLC, Bristol-Myers Squibb Company, and AstraZeneca; and advisory boards for Gilead Sciences, Inc., Eli Lilly and Company, Merck and Co., Inc., Novo Nordisk, Inc., Takeda Pharmaceuticals North America, Inc., AstraZeneca, Bristol-Myers Squibb Company, Amylin Pharmaceuticals, LLC, Santarus, Inc., Boehringer Ingelheim, Eli Lilly and Company, and Janssen Pharmaceuticals, Inc.

4. Brownlee M. Advanced products of nonenzymatic glycosylation and the pathogenesis of diabetic complications. In: Rifkin H, Porte D Jr, eds. Ellenberg and Rifkin’s Diabetes Mellitus: Theory and Practice. 4th ed. New York, NY: Elsevier Science Publishers; 1990: 279–291.

5. Ceriello A. Postprandial hyperglycemia and diabetes complications: is it time to treat? Diabetes. 2005;54(1):1–7.

6. Giacco F, Brownlee M. Oxidative stress and diabetic complications. Circ Res. 2010;107(9):1058–1070.

7. Hermanides J, Vriesendorp TM, Bosman RJ, Zandstra DF, Hoekstra JB, Devries JH. Glucose variability is associated with intensive care unit mortality. Crit Care Med. 2010;38(3):838–842.

8. Umpierrez GE, Isaacs SD, Bazargan N, You X, Thaler LM, Kitabchi AE. Hyperglycemia: an independent marker of in-hospital mortality in patients with undiagnosed diabetes. J Clin Endocrinol Metab. 2002;87(3):978–982.

9. Ray KK, Seshasai SR, Wijesuriya S, et 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. 2009;373(9677):1765–1772.

10. Action to Control Cardiovascular Risk in Diabetes Study Group, Gerstein HC, Miller ME, Byington RP, et al. Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med. 2008;358(24):2545–2559.

11. ADVANCE Collaborative Group, Patel A, MacMahon S, Chalmers J, et al. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med. 2008;358(24):2560–2572.

12. Duckworth W, Abraira C, Moritz T, et al; VADT Investigators. Glucose control and vascular complications in veterans with type 2 diabetes. N Engl J Med. 2009;360(2):129–139.

13. Look AHEAD Research Group, Pi-Sunyer X, Blackburn G, et al. Reduction in weight and cardiovascular disease risk factors in indi-viduals with type 2 diabetes: one-year results of the look AHEAD trial. Diabetes Care. 2007;30(6):1374–1383.

14. Calle EE, Thun MJ, Petrelli JM, Rodriguez C, Heath CW Jr. Body-mass index and mortality in a prospective cohort of U.S. adults. N Engl J Med. 1999;341(15):1097–1105.

15. Lindstrom T, Jorfeldt L, Tegler L, Arnqvist HJ. Hypoglycaemia and cardiac arrhythmias in patients with type 2 diabetes mellitus. Diabet Med. 1992;9(6):536–541.

16. Nielsen JB, Graff C, Pietersen A, et al. J-shaped association between QTc interval duration and the risk of atrial fibrillation: results from the Copenhagen ECG study. J Am Coll Cardiol. 2013;61(25):2557–2564.

17. Gruden G, Barutta F, Chaturvedi N, et al. Severe hypoglyce-mia and cardiovascular disease incidence in type 1 diabetes: the EURODIAB Prospective Complications Study. Diabetes Care. 2012;35(7):1598–1604.

18. Gill GV, Woodward A, Casson IF, Weston PJ. Cardiac arrhythmia and nocturnal hypoglycaemia in type 1 diabetes--the ‘dead in bed’ syndrome revisited. Diabetologia. 2009;52(1):42–45.

19. Heller SR. Abnormalities of the electrocardiogram during hypoglycae-mia: the cause of the dead in bed syndrome? Int J Clin Pract Suppl. 2002;(129):27–32.

20. Due-Andersen R, Hoi-Hansen T, Larroude CE, et al. Cardiac repo-larization during hypoglycaemia in type 1 diabetes: impact of basal renin-angiotensin system activity. Europace. 2008;10(7):860–867.

21. Desouza C, Salazar H, Cheong B, Murgo J, Fonseca V. Association of hypoglycemia and cardiac ischemia: a study based on continuous monitoring. Diabetes Care. 2003;26(5):1485–1489.

22. Tanenberg RJ, Newton CA, Drake AJ. Confirmation of hypoglycemia in the “dead-in-bed” syndrome, as captured by a retrospective continu-ous glucose monitoring system. Endocr Pract. 2010;16(2):244–248.

23. Zoungas S, Patel A, Chalmers J, et al; ADVANCE Collaborative Group. Severe hypoglycemia and risks of vascular events and death. N Engl J Med. 2010;363(15):1410–1418.

24. Robinson RT, Harris ND, Ireland RH, Lee S, Newman C, Heller SR. Mechanisms of abnormal cardiac repolarization during insulin-induced hypoglycemia. Diabetes. 2003;52(6):1469–1474.

References 1. Duggal R, Menkes DB. Evidence-based medicine in practice. Int J

Clin Pract. 2011;65(6):639–644. 2. Sniderman AD, LaChapelle KJ, Rachon NA, Furberg CD. The neces-

sity for clinical reasoning in the era of evidence-based medicine. Mayo Clin Proc. 2013;88(10):1108–1114.

3. Centers for Disease Control and Prevention. National Diabetes Fact Sheet: national estimates and general information on diabetes and prediabetes in the United States, 2011. Atlanta, GA: US Department of Health and Human Services, Centers for Disease Control and Prevention, 2011.

SCHWARTZ_proof

Page 14: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

Stanley Schwartz

14 © Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]

Warning: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

25. Christensen TF, Randlov J, Kristensen LE, Eldrup E, Hejlesen OK, Struijk JJ. QT Measurement and heart rate correction during hypoglycemia: is there a bias? Cardiol Res Pract. 2010;2010: 961290.

26. Tremblay A, Pinsard D, Coveney S, et al. Counterregulatory response to insulin-induced hypoglycemia in trained and nontrained humans. Metabolism. 1990;39(11):1138–1143.

27. American Diabetes Association. Standards of medical care in diabetes—2014. Diabetes Care. 2014;37(Suppl 1):S14-S80.

28. DeFronzo RA. Banting Lecture. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes mellitus. Diabetes. 2009;58(4):773–795.

29. Elrick H, Stimmler L, Hlad CJ Jr, Arai Y. Plasma insulin response to oral and intravenous glucose administration. J Clin Endocrinol Metab. 1964;24:1076–1082.

30. Nauck M, Stockmann F, Ebert R, Creutzfeldt W. Reduced incretin effect in type 2 (non-insulin-dependent) diabetes. Diabetologia. 1986;29(1):46–52.

31. Holst JJ, Orskov C. The incretin approach for diabetes treatment: modulation of islet hormone release by GLP-1 agonism. Diabetes. 2004;53(Suppl 3):S197–S204.

32. Meier JJ, Nauck MA. Glucagon-like peptide 1(GLP-1) in biology and pathology. Diabetes Metab Res Rev. 2005;21(2):91–117.

33. Hojberg PV, Vilsboll T, Rabol R, et al. Four weeks of near-normalisation of blood glucose improves the insulin response to glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide in patients with type 2 diabetes. Diabetologia. 2009;52(2):199–207.

34. Vilsboll T, Krarup T, Madsbad S, Holst JJ. Defective amplification of the late phase insulin response to glucose by GIP in obese Type II diabetic patients. Diabetologia. 2002;45(8):1111–1119.

35. Aronoff SL, Berkowitz K, Shreiner B, Want L. Glucose metabolism and regulation: beyond insulin and glucagon. Diabetes Spectr. 2004;17(3):183–190.

36. Vilsboll T, Knop FK, Krarup T, et al. The pathophysiology of dia-betes involves a defective amplification of the late-phase insulin response to glucose by glucose-dependent insulinotropic poly-peptide, regardless of etiology and phenotype. J Clin Endocrinol Metab. 2003;88(10):4897–4903.

37. Rachman J, Barrow BA, Levy JC, Turner RC. Near-normalisation of diurnal glucose concentrations by continuous administration of glucagon-like peptide-1 (GLP-1) in subjects with NIDDM. Diabetologia. 1997;40(2):205–211.

38. Nauck MA, Kleine N, Orskov C, Holst JJ, Willms B, Creutzfeldt W. Normalization of fasting hyperglycaemia by exogenous glucagon-like peptide 1 (7–36 amide) in type 2 (non-insulin-dependent) diabetic patients. Diabetologia. 1993;36(8):741–744.

39. Schwartz S, DeFronzo RA. Is incretin-based therapy ready for the care of hospitalized patients with type 2 diabetes?: The time has come for GLP-1 receptor agonists! Diabetes Care. 2013;36(7):2107–2111.

40. Zhu ZQ, Wang D, Xiang D, Yuan YX, Wang Y. Calcium/calmodulin-dependent serine protein kinase is involved in exendin-4-induced insulin secretion in INS-1 cells. Metabolism. 2014;63(1):120–126.

41. Drucker DJ. The biology of incretin hormones. Cell Metab. 2006;3(3):153–165.

42. Sasaki S, Inoguchi T. The role of oxidative stress in the pathogen-esis of diabetic vascular complications. Diabetes Metab J. 2012;36(4):255–261.

43. Sokos GG, Nikolaidis LA, Mankad S, Elahi D, Shannon RP. Glucagon-like peptide-1 infusion improves left ventricular ejection fraction and functional status in patients with chronic heart failure. J Card Fail. 2006;12(9):694–699.

44. Mafong DD, Henry RR. Exenatide as a treatment for diabetes and obesity: implications for cardiovascular risk reduction. Curr Atheroscler Rep. 2008;10(1):55–60.

45. Gutzwiller JP, Tschopp S, Bock A, et al. Glucagon-like peptide 1 induces natriuresis in healthy subjects and in insulin-resistant obese men. J Clin Endocrinol Metab. 2004;89(6):3055–3061.

46. Nikolaidis LA, Mankad S, Sokos GG, et al. Effects of glucagon-like peptide-1 in patients with acute myocardial infarction and left ventricular dysfunction after successful reperfusion. Circulation. 2004;109(8):962–965.

47. Jax T. Treatment of patients with diabetes with GLP-1 analogues or DPP-4- inhibitors: a hot topic for cardiologists? Clin Res Cardiol. 2009;98(2):75–79.

48. Nystrom T, Gonon AT, Sjoholm A, Pernow J. Glucagon-like peptide-1 relaxes rat conduit arteries via an endothelium-independent mecha-nism. Regul Pept. 2005;125(1–3):173–177.

49. Sulistio M, Carothers C, Mangat M, Lujan M, Oliveros R, Chilton R. GLP-1 agonist-based therapies: an emerging new class of antidiabetic drug with potential cardioprotective effects. Curr Atheroscler Rep. 2009;11(2):93–99.

50. Sokos GG, Bolukoglu H, German J, et al. Effect of glucagon-like peptide-1 (GLP-1) on glycemic control and left ventricular function in patients undergoing coronary artery bypass grafting. Am J Cardiol. 2007;100(5):824–829.

51. Toft-Nielsen MB, Damholt MB, Madsbad S, et al. Determinants of the impaired secretion of glucagon-like peptide-1 in type 2 diabetic patients. J Clin Endocrinol Metab. 2001;86(8):3717–3723.

52. DeFronzo RA, Okerson T, Viswanathan P, Guan X, Holcombe JH, MacConell L. Effects of exenatide versus sitagliptin on postpran-dial glucose, insulin and glucagon secretion, gastric emptying, and caloric intake: a randomized, cross-over study. Curr Med Res Opin. 2008;24(10):2943–2952.

53. Holst JJ, Deacon CF, Vilsboll T, Krarup T, Madsbad S. Glucagon-like peptide-1, glucose homeostasis and diabetes. Trends Mol Med. 2008;14(4):161–168.

54. Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 dia-betes. Lancet. 2006;368(9548):1696–1705.

55. Nauck MA, Meininger G, Sheng D, Terranella L, Stein PP; Sitagliptin Study 024 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, compared with the sulfonylurea, glipizide, in patients with type 2 diabetes inadequately controlled on metformin alone: a randomized, double-blind, non-inferiority trial. Diabetes Obes Metab. 2007;9(2):194–205.

56. Seck T, Nauck M, Sheng D, et al; Sitagliptin Study 024 Group. Safety and efficacy of treatment with sitagliptin or glipizide in patients with type 2 diabetes inadequately controlled on metformin: a 2-year study. Int J Clin Pract. 2010;64(5):562–576.

57. Ferrannini E, Fonseca V, Zinman B, et al. Fifty-two-week efficacy and safety of vildagliptin vs. glimepiride in patients with type 2 diabetes mellitus inadequately controlled on metformin monotherapy. Diabetes Obes Metab. 2009;11(2):157–166.

58. Filozof C, Gautier JF. A comparison of efficacy and safety of vildaglip-tin and gliclazide in combination with metformin in patients with Type 2 diabetes inadequately controlled with metformin alone: a 52-week, randomized study. Diabet Med. 2010;27(3):318–326.

59. Inzucchi SE, Bergenstal RM, Buse JB, et al. Management of hyper-glycemia in type 2 diabetes: a patient-centered approach. Position statement of the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care. 2012;35(6):1364–1379.

60. Garber AJ, Abrahamson MJ, Barzilay JI, et al; American Association of Clinical Endocrinologists. AACE comprehensive diabetes manage-ment algorithm 2013. Endocr Pract. 2013;19(2):327–336.

61. Del Prato S, Barnett AH, Huisman H, Neubacher D, Woerle HJ, Dugi KA. Effect of linagliptin monotherapy on glycaemic control and markers of β-cell function in patients with inadequately con-trolled type 2 diabetes: a randomized controlled trial. Diabetes Obes Metab. 2011;13(3):258–267.

62. Gallwitz B, Rosenstock J, Rauch T, et al. 2-year efficacy and safety of linagliptin compared with glimepiride in patients with type 2 diabetes inadequately controlled on metformin: a randomised, double-blind, non-inferiority trial. Lancet. 2012;380(9840):475–483.

SCHWARTZ_proof

Page 15: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

Incretin-Based Therapy in Patients With Diabetes

© Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260 15ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

63. Gomis R, Espadero RM, Jones R, Woerle HJ, Dugi KA. Efficacy and safety of initial combination therapy with linagliptin and pio-glitazone in patients with inadequately controlled type 2 diabetes: a randomized, double-blind, placebo-controlled study. Diabetes Obes Metab. 2011;13(7):653–661.

64. Haak T, Meinicke T, Jones R, Weber S, von Eynatten M, Woerle HJ. Initial combination of linagliptin and metformin improves glycaemic control in type 2 diabetes: a randomized, double-blind, placebo-controlled study. Diabetes Obes Metab. 2012;14(6):565–574.

65. Owens DR, Swallow R, Dugi KA, Woerle HJ. Efficacy and safety of linagliptin in persons with type 2 diabetes inadequately controlled by a combination of metformin and sulphonylurea: a 24-week randomized study. Diabet Med. 2011;28(11):1352–1361.

66. Taskinen MR, Rosenstock J, Tamminen I, et al. Safety and efficacy of linagliptin as add-on therapy to metformin in patients with type 2 diabetes: a randomized, double-blind, placebo-controlled study. Diabetes Obes Metab. 2011;13(1):65–74.

67. Chacra AR, Tan GH, Apanovitch A, Ravichandran S, List J, Chen R; CV181–040 Investigators. Saxagliptin added to a submaximal dose of sulphonylurea improves glycaemic control compared with uptitra-tion of sulphonylurea in patients with type 2 diabetes: a randomised controlled trial. Int J Clin Pract. 2009;63(9):1395–1406.

68. DeFronzo RA, Hissa MN, Garber AJ, et al; Saxagliptin 014 Study Group. The efficacy and safety of saxagliptin when added to metformin therapy in patients with inadequately controlled type 2 diabetes with metformin alone. Diabetes Care. 2009;32(9):1649–1655.

69. Hollander P, Li J, Allen E, Chen R; CV181–013 Investigators. Saxagliptin added to a thiazolidinedione improves glycemic con-trol in patients with type 2 diabetes and inadequate control on thiazolidinedione alone. J Clin Endocrinol Metab. 2009;94(12): 4810–4819.

70. Jadzinsky M, Pfutzner A, Paz-Pacheco E, et al; CV181–039 Inves-tigators. Saxagliptin given in combination with metformin as initial therapy improves glycaemic control in patients with type 2 diabetes compared with either monotherapy: a randomized controlled trial. Diabetes Obes Metab. 2009;11(6):611–622.

71. Rosenstock J, Aguilar-Salinas C, Klein E, Nepal S, List J, Chen R; CV181–011 Study Investigators. Effect of saxagliptin monotherapy in treatment-naive patients with type 2 diabetes. Curr Med Res Opin. 2009;25(10):2401–2411.

72. Scheen AJ, Charpentier G, Ostgren CJ, Hellqvist A, Gause-Nilsson I. Efficacy and safety of saxagliptin in combination with metformin compared with sitagliptin in combination with metformin in adult patients with type 2 diabetes mellitus. Diabetes Metab Res Rev. 2010;26(7):540–549.

73. Charbonnel B, Karasik A, Liu J, Wu M, Meininger G, Sitagliptin Study Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing metformin therapy in patients with type 2 diabetes inadequately controlled with metformin alone. Diabetes Care. 2006;29(12):2638–2643.

74. Dobs AS, Goldstein BJ, Aschner P, et al. Efficacy and safety of sita-gliptin added to ongoing metformin and rosiglitazone combination therapy in a randomized placebo-controlled 54-week trial in patients with type 2 diabetes. J Diabetes. 2013;5(1):68–79.

75. Goldstein BJ, Feinglos MN, Lunceford JK, Johnson J, Williams-Herman DE; Sitagliptin 036 Study Group. Effect of initial combination therapy with sitagliptin, a dipeptidyl peptidase-4 inhibitor, and met-formin on glycemic control in patients with type 2 diabetes. Diabetes Care. 2007;30(8):1979–1987.

76. Vilsboll T, Rosenstock J, Yki-Jarvinen H, et al. Efficacy and safety of sitagliptin when added to insulin therapy in patients with type 2 diabetes. Diabetes Obes Metab. 2010;12(2):167–177.

77. DeFronzo RA, Fleck PR, Wilson CA, Mekki Q; Alogliptin Study 010 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor alogliptin in patients with type 2 diabetes and inadequate glycemic control: a randomized, double-blind, placebo-controlled study. Diabetes Care. 2008;31(12):2315–2317.

78. Pratley RE, Reusch JE, Fleck PR, Wilson CA, Mekki Q; Alogliptin Study 009 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor alogliptin added to pioglitazone in patients with type 2 diabetes: a randomized, double-blind, placebo-controlled study. Curr Med Res Opin. 2009;25(10):2361–2371.

79. Pratley RE, Kipnes MS, Fleck PR, Wilson C, Mekki Q; Alo-gliptin Study 007 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor alogliptin in patients with type 2 diabetes inadequately controlled by glyburide monotherapy. Diabetes Obes Metab. 2009;11(2):167–176.

80. Nauck MA, Ellis GC, Fleck PR, Wilson CA, Mekki Q; Alogliptin Study 008 Group. Efficacy and safety of adding the dipeptidyl peptidase-4 inhibitor alogliptin to metformin therapy in patients with type 2 diabetes inadequately controlled with metformin monotherapy: a multicentre, randomised, double-blind, placebo-controlled study. Int J Clin Pract. 2009;63(1):46–55.

81. Rosenstock J, Inzucchi SE, Seufert J, Fleck PR, Wilson CA, Mekki Q. Initial combination therapy with alogliptin and piogli-tazone in drug-naive patients with type 2 diabetes. Diabetes Care. 2010;33(11):2406–2408.

82. Buse JB, Henry RR, Han J, et al; Exenatide-113 Clinical Study Group. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in sulfonylurea-treated patients with type 2 diabetes. Diabetes Care. 2004;27(11):2628–2635.

83. DeFronzo RA, Ratner RE, Han J, Kim DD, Fineman MS, Baron AD. Effects of exenatide (exendin-4) on glycemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetes. Diabetes Care. 2005;28(5):1092–1100.

84. Heine RJ, Van Gaal LF, Johns D, et al. Exenatide versus insulin glargine in patients with suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med. 2005;143(8):559–569.

85. Kendall DM, Riddle MC, Rosenstock J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in patients with type 2 diabetes treated with metformin and a sulfonylurea. Diabetes Care. 2005;28(5):1083–1091.

86. Moretto TJ, Milton DR, Ridge TD, et al. Efficacy and tolerability of exenatide monotherapy over 24 weeks in antidiabetic drug-naive patients with type 2 diabetes: a randomized, double-blind, placebo-controlled, parallel-group study. Clin Ther. 2008;30(8):1448–1460.

87. Zinman B, Hoogwerf BJ, Duran Garcia S, et al. The effect of adding exenatide to a thiazolidinedione in suboptimally controlled type 2 dia-betes: a randomized trial. Ann Intern Med. 2007;146(7):477–485.

88. Bergenstal RM, Wysham C, Macconell L, et al; DURATION-2 Study Group. Efficacy and safety of exenatide once weekly versus sitagliptin or pioglitazone as an adjunct to metformin for treatment of type 2 diabetes (DURATION-2): a randomised trial. Lancet. 2010;376(9739):431–439.

89. Buse JB, Nauck M, Forst T, et al. Exenatide once weekly versus lira-glutide once daily in patients with type 2 diabetes (DURATION-6): a randomised, open-label study. Lancet. 2013;381(9861):117–124.

90. Diamant M, Van Gaal L, Stranks S, et al. Once weekly exenatide compared with insulin glargine titrated to target in patients with type 2 diabetes (DURATION-3): an open-label randomised trial. Lancet. 2010;375(9733):2234–2243.

91. Drucker DJ, Buse JB, Taylor K, et al; DURATION-1 Study Group. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label, non-inferiority study. Lancet. 2008;372(9645):1240–1250.

92. Russell-Jones D, Cuddihy RM, Hanefeld M, et al; DURATION-4 Study Group. Efficacy and safety of exenatide once weekly versus metformin, pioglitazone, and sitagliptin used as monotherapy in drug-naive patients with type 2 diabetes (DURATION-4): a 26-week double-blind study. Diabetes Care. 2012;35(2):252–258.

93. Garber A, Henry R, Ratner R, et al; LEAD-3 (Mono) study Group. Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a randomised, 52-week, phase III, double-blind, parallel-treatment trial. Lancet. 2009;373(9662):473–481.

SCHWARTZ_proof

Page 16: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

Stanley Schwartz

16 © Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]

Warning: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

94. Marre M, Shaw J, Brandle M, et al; LEAD-1 SU Study Group. Liraglutide, a once-daily human GLP-1 analogue, added to a sul-phonylurea over 26 weeks produces greater improvements in gly-caemic and weight control compared with adding rosiglitazone or placebo in subjects with Type 2 diabetes (LEAD-1 SU). Diabet Med. 2009;26(3):268–278.

95. Nauck M, Frid A, Hermansen K, et al; LEAD-2 Study Group. Effi-cacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (liraglutide effect and action in diabetes)-2 study. Diabetes Care. 2009;32(1):84–90.

96. Pratley RE, Nauck M, Bailey T, et al; 1860-LIRA-DPP-4 Study Group. Liraglutide versus sitagliptin for patients with type 2 diabe-tes who did not have adequate glycaemic control with metformin: a 26-week, randomised, parallel-group, open-label trial. Lancet. 2010;375(9724):1447–1456.

97. Russell-Jones D, Vaag A, Schmitz O, et al; Liraglutide Effect and Action in Diabetes 5 (LEAD-5) met+SU Study Group. Liraglutide vs insulin glargine and placebo in combination with metformin and sulfonylurea therapy in type 2 diabetes mellitus (LEAD-5 met+SU): a randomised controlled trial. Diabetologia. 2009;52(10): 2046–2055.

98. Zinman B, Gerich J, Buse JB, et al; LEAD-4 Study Investigators. Efficacy and safety of the human glucagon-like peptide-1 analog liraglutide in combination with metformin and thiazolidinedione in patients with type 2 diabetes (LEAD-4 Met+TZD). Diabetes Care. 2009;32(7):1224–1230.

99. Bloomgarden ZT, Dodis R, Viscoli CM, Holmboe ES, Inzucchi SE. Lower baseline glycemia reduces apparent oral agent glucose-lowering efficacy: a meta-regression analysis. Diabetes Care. 2006;29(9):2137–2139.

100. Bailey T, Pratley R, Buse J, Furber S, Hartvig H, Schmidt W. Compari-son of incretin-based therapies: liraglutide offers greater improvements in A1c than sitagliptin or exenatide across baseline A1c categories [abstract 2252-PO]. Diabetes. 2011;60(Suppl 1):A599.

101. Wysham CH, Woo VC, Mathieu C, et al. Canagliflozin (CANA) added on to dipeptidyl peptidase-4 inhibitors (DPP-4i) or glucagon-like peptide-1 (GLP-1) agonists with or without other antihyperglycemic agents (AHAs) in type 2 diabetes mellitus (T2DM) [abstract 1080-P]. Diabetes. 2013;62(Suppl 1):A279.

102. Merovci A, Solis-Herrera C, Daniele G, et al. Dapagliflozin improves muscle insulin sensitivity but enhances endogenous glucose produc-tion. J Clin Invest. 2014;124(2):509–514.

103. Vilsboll T, Christensen M, Junker AE, Knop FK, Gluud LL. Effects of glucagon-like peptide-1 receptor agonists on weight loss: system-atic review and meta-analyses of randomised controlled trials. BMJ. 2012;344:d7771.

104. Maggs D, Kim D, Holcombe J, et al. Exenatide-induced reductions in A1C and body weight in long-term trials are not explained by gas-trointestinal side effects [abstract 485-P]. Diabetes. 2005;54(Suppl 1):A120.

105. Buse JB, Drucker DJ, Taylor KL, et al; DURATION-1 Study Group. DURATION-1: exenatide once weekly produces sustained glycemic control and weight loss over 52 weeks. Diabetes Care. 2010;33(6):1255–1261.

106. Buse JB, Rosenstock J, Sesti G, et al; LEAD-6 Study Group. Lira-glutide once a day versus exenatide twice a day for type 2 diabetes: a 26-week randomised, parallel-group, multinational, open-label trial (LEAD-6). Lancet. 2009;374(9683):39–47.

107. US Department of Health and Human Services. Guidance for Industry: Diabetes Mellitus — Evaluating Cardiovascular Risk in New Antidi-abetic Therapies to Treat Type 2 Diabetes. Silver Spring, MD: Food and Drug Administration, Center for Drug Evaluation and Research (CDER); 2008.

108. White WB, Pratley R, Fleck P, et al. Cardiovascular safety of the dipetidyl peptidase-4 inhibitor alogliptin in type 2 diabetes mellitus. Diabetes Obes Metab. 2013;15(7):668–673.

109. Scirica BM, Bhatt DL, Braunwald E, et al; SAVOR-TIMI 53 Steering Committee and Investigators. Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N Engl J Med. 2013;369(14):1317–1326.

110. Patil HR, Al Badarin FJ, Al Shami HA, et al. Meta-analysis of effect of dipeptidyl peptidase-4 inhibitors on cardiovascular risk in type 2 diabetes mellitus. Am J Cardiol. 2012;110(6):826–833.

111. Fadini GP, Avogaro A. Cardiovascular effects of DPP-4 inhibition: beyond GLP-1. Vascul Pharmacol. 2011;55(1–3):10–16.

112. Read PA, Khan FZ, Heck PM, Hoole SP, Dutka DP. DPP-4 inhibition by sitagliptin improves the myocardial response to dobutamine stress and mitigates stunning in a pilot study of patients with coronary artery disease. Circ Cardiovasc Imaging. 2010;3(2):195–201.

113. Best JH, Hoogwerf BJ, Herman WH, et al. Risk of cardiovascular disease events in patients with type 2 diabetes prescribed the glucagon-like peptide 1 (GLP-1) receptor agonist exenatide twice daily or other glucose-lowering therapies: a retrospective analysis of the LifeLink database. Diabetes Care. 2011;34(1):90–95.

114. Januvia [prescribing information]. Whitehouse Station, NJ: Merck and Co., Inc.; 2010.

115. Onglyza [prescribing information]. Princeton, NJ: Bristol-Myers Squibb Company/ Wilmington, DE: AstraZeneca Pharmaceuticals LP; 2013.

116. Tradjenta [prescribing information]. Ridgefield, CT: Boehringer Ingelheim Pharmaceuticals Inc./Indianapolis, IN: Eli Lilly and Company; 2013.

117. Nesina [prescribing information]. Deerfield, IL: Takeda Pharmaceu-ticals America Inc.; 2013.

118. Byetta [prescribing information]. Princeton, NJ: Bristol-Myers Squibb Company/ Wilmington, DE: AstraZeneca Pharmaceuticals LP.; 2013.

119. Victoza [prescribing information]. Plainsboro, NJ: Novo Nordisk Inc.; 2013.

120. Bydureon [prescribing information]. Wilmington, DE: AstraZeneca Pharmaceuticals LP.; 2014.

121. Ratner RE, Maggs D, Nielsen LL, et al. Long-term effects of exenatide therapy over 82 weeks on glycaemic control and weight in over-weight metformin-treated patients with type 2 diabetes mellitus. Diabetes Obes Metab. 2006;8(4):419–428.

122. Pratley R, Nauck M, Bailey T, et al; 1860-LIRA-DPP-4 Study Group. One year of liraglutide treatment offers sustained and more effective glycaemic control and weight reduction compared with sitagliptin, both in combination with metformin, in patients with type 2 diabetes: a randomised, parallel-group, open-label trial. Int J Clin Pract. 2011;65(4):397–407.

123. MacConell L, Brown C, Gurney K, Han J. Safety and tolerability of exenatide twice daily in patients with type 2 diabetes: integrated analysis of 5594 patients from 19 placebo-controlled and comparator- controlled clinical trials. Diabetes Metab Syndr Obes. 2012;5:29–41.

124. Buse JB, Klonoff DC, Nielsen LL, et al. Metabolic effects of two years of exenatide treatment on diabetes, obesity, and hepatic biomarkers in patients with type 2 diabetes: an interim analysis of data from the open-label, uncontrolled extension of three double-blind, placebo-controlled trials. Clin Ther. 2007;29(1):139–153.

125. Diamant M, Van Gaal L, Stranks S, et al. Safety and efficacy of once-weekly exenatide compared with insulin glargine titrated to target in patients with type 2 diabetes over 84 weeks. Diabetes Care. 2012;35(4):683–689.

126. Ellero C, Han J, Bhavsar S, et al. Prophylactic use of anti-emetic medications reduced nausea and vomiting associated with exenatide treatment: a retrospective analysis of an open-label, parallel-group, single-dose study in healthy subjects. Diabet Med. 2010;27(10): 1168–1173.

127. Pendergrass M, Fenton C, Haffner SM, Chen W. Exenatide and sitagliptin are not associated with increased risk of acute renal failure: a retrospective claims analysis. Diabetes Obes Metab. 2012;14(7):596–600.

SCHWARTZ_proof

Page 17: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

Incretin-Based Therapy in Patients With Diabetes

© Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260 17ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

128. Tuttle KR, Heilmann C, Hoogwerf BJ, Brown C, Anderson PW. Effects of exenatide on kidney function, adverse events, and clinical end points of kidney disease in type 2 diabetes. Am J Kidney Dis. 2013;62(2):396–398.

129. Johansen OE, Whitfield R. Exenatide may aggravate moderate diabetic renal impairment: a case report. Br J Clin Pharmacol. 2008;66(4):568–569.

130. Weise WJ, Sivanandy MS, Block CA, Comi RJ. Exenatide-associated ischemic renal failure. Diabetes Care. 2009;32(2):e22–e23.

131. Bjerre Knudsen L, Madsen LW, Andersen S, et al. Glucagon-like peptide-1 receptor agonists activate rodent thyroid C-cells caus-ing calcitonin release and C-cell proliferation. Endocrinology. 2010;151(4):1473–1486.

132. Kissner T, Dorau M, Perren A, Blank A, Heinrichs M, Dietert G. Comparison of GLP-1R expression in human C-cell thyroid tumor tissue and human thyroid tissue without C-cell pathology [abstract 1002-P]. Diabetes. 2013;62(Suppl 1):A257.

133. Schwahn U, Stengelin S, Werner U. Functional activity of lixisenatide and GLP-1 receptor expression in in-vitro C-cells of rat and human origin [abstract 1008-P]. Diabetes. 2013;62(Suppl 1):A259.

134. Ahmad SR, Swann J. Exenatide and rare adverse events. N Engl J Med. 2008;358(18):1970–1971; discussion 1971–1972.

135. Garg R, Chen W, Pendergrass M. Acute pancreatitis in type 2 diabetes treated with exenatide or sitagliptin: a retrospective obser-vational pharmacy claims analysis. Diabetes Care. 2010;33(11): 2349–2354.

136. Dore DD, Seeger JD, Arnold Chan K. Use of a claims-based active drug safety surveillance system to assess the risk of acute pancreatitis with exenatide or sitagliptin compared to metformin or glyburide. Curr Med Res Opin. 2009;25(4):1019–1027.

137. Ryder RE, Sen Gupta P, Thong KY; ABCD nationwide exenatide and liraglutide audit contributors. The Association of British Clinical Diabetologists nationwide exenatide and liraglutide audits suggest a low incidence of acute pancreatitis. Response to Robson. Incretins and pancreatitis--what happens next? A personal viewpoint. Diabet Med. 2013;30(12):1510–1511.

138. Egan AG, Blind E, Dunder K, et al. Pancreatic safety of incre-tin-based drugs—FDA and EMA assessment. N Engl J Med. 2014;370(9):794–797.

139. Zhao Y, Yang L, Xiang Y, et al. Dipeptidyl peptidase 4 inhibitor sita-gliptin maintains β-cell function in patients with recent-onset latent autoimmune diabetes in adults: one year prospective study. J Clin Endocrinol Metab. 2014;99(5):E876-E880.

140. Almeda-Valdes P, Cuevas-Ramos D, Brito-Cordova G, Gomez-Perez FJ. Treatment with sitagliptin phosphate in patients with symptomatic reactive hypoglycemia. A randomized, placebo-controlled, double-blinded, clinical trial [abstract SUN-209]. ENDO 2012, Houston, TX, June 23–26, 2012. Available from: http://www.abstracts2view.com/endo/view.php?nu = ENDO12L_SUN-209&terms = . Accessed January 28, 2014.

141. Amori RE, Lau J, Pittas AG. Efficacy and safety of incretin therapy in type 2 diabetes: systematic review and meta-analysis. JAMA. 2007;298(2):194–206.

142. Klonoff DC, Buse JB, Nielsen LL, et al. Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr Med Res Opin. 2008;24(1):275–286.

143. Bhushan R, Elkind-Hirsch KE, Bhushan M, Butler WJ, Duncan K, Marrioneaux O. Exenatide use in the management of metabolic syndrome: a retrospective database study. Endocr Pract. 2008;14(8):993–999.

144. Astrup A, Carraro R, Finer N, et al; NN8022–1807 Investigators. Safety, tolerability and sustained weight loss over 2 years with the once-daily human GLP-1 analog, liraglutide. Int J Obes (Lond). 2012;36(6):843–854.

145. Armato J, DeFronzo RA, Abdul-Ghani M, Ruby R. Successful treat-ment of prediabetes in clinical practice: targeting insulin resistance and beta-cell dysfunction. Endocr Pract. 2012;18(3):342–350.

146. Perreault L, Pan Q, Mather KJ, et al; Diabetes Prevention Program Research Group. Effect of regression from prediabetes to normal glucose regulation on long-term reduction in diabetes risk: results from the Diabetes Prevention Program Outcomes Study. Lancet. 2012;379(9833):2243–2251.

147. Chacra AR, Tan GH, Ravichandran S, List J, Chen R, CV181040 Investigators. Safety and efficacy of saxagliptin in combination with submaximal sulphonylurea versus up-titrated sulphonylurea over 76 weeks. Diab Vasc Dis Res. 2011;8(2):150–159.

148. Garber A, Henry RR, Ratner R, et al; LEAD-3 (Mono) Study Group. Liraglutide, a once-daily human glucagon-like peptide 1 analogue, provides sustained improvements in glycaemic control and weight for 2 years as monotherapy compared with glimepiride in patients with type 2 diabetes. Diabetes Obes Metab. 2011;13(4):348–356.

149. Williams-Herman D, Johnson J, Teng R, et al. Efficacy and safety of sitagliptin and metformin as initial combination therapy and as monotherapy over 2 years in patients with type 2 diabetes. Diabetes Obes Metab. 2010;12(5):442–451.

150. Goke B, Hershon K, Kerr D, et al. Efficacy and safety of vildagliptin monotherapy during 2-year treatment of drug-naive patients with type 2 diabetes: comparison with metformin. Horm Metab Res. 2008;40(12):892–895.

151. Gomis R, Owens DR, Taskinen MR, et al. Long-term safety and effi-cacy of linagliptin as monotherapy or in combination with other oral glucose-lowering agents in 2121 subjects with type 2 diabetes: up to 2 years exposure in 24-week phase III trials followed by a 78-week open-label extension. Int J Clin Pract. 2012;66(8):731–740.

152. Bunck MC, Diamant M, Corner A, et al. One-year treatment with exenatide improves beta-cell function, compared with insulin glargine, in metformin-treated type 2 diabetic patients: a randomized, controlled trial. Diabetes Care. 2009;32(5):762–768.

153. Hanefeld M, Pfutzner A, Forst T, Lubben G. Glycemic control and treatment failure with pioglitazone versus glibenclamide in type 2 diabetes mellitus: a 42-month, open-label, observational, primary care study. Curr Med Res Opin. 2006;22(6):1211–1215.

154. Kahn SE, Haffner SM, Heise MA, et al; ADOPT Study Group. Glyce-mic durability of rosiglitazone, metformin, or glyburide monotherapy. N Engl J Med. 2006;355(23):2427–2443.

155. Mazzone T, Meyer PM, Feinstein SB, et al. Effect of pioglita-zone compared with glimepiride on carotid intima-media thick-ness in type 2 diabetes: a randomized trial. JAMA. 2006;296(21): 2572–2581.

156. Nissen SE, Nicholls SJ, Wolski K, et al; PERISCOPE Investigators. Comparison of pioglitazone vs glimepiride on progression of coronary atherosclerosis in patients with type 2 diabetes: the PERISCOPE randomized controlled trial. JAMA. 2008;299(13):1561–1573.

157. Tan MH, Baksi A, Krahulec B, et al; GLAL Study Group. Com-parison of pioglitazone and gliclazide in sustaining glycemic con-trol over 2 years in patients with type 2 diabetes. Diabetes Care. 2005;28(3):544–550.

158. UK Prospective Diabetes Study (UKPDS) Group. Intensive blood-glucose control with sulphonylureas or insulin compared with con-ventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). Lancet. 1998;352(9131):837–853.

159. Williams SA, Shi L, Brenneman SK, Johnson JC, Wegner JC, Fonseca V. The burden of hypoglycemia on healthcare utilization, costs, and quality of life among type 2 diabetes mellitus patients. J Diabetes Complications. 2012;26(5):399–406.

160. Willis WD, Diago-Cabezudo JI, Madec-Hily A, Aslam A. Medi-cal resource use, disturbance of daily life and burden of hypo-glycemia in insulin-treated patients with diabetes: results from a European online survey. Expert Rev Pharmacoecon Outcomes Res. 2013;13(1):123–130.

161. Bron M, Marynchenko M, Yang H, Yu AP, Wu EQ. Hypoglyce-mia, treatment discontinuation, and costs in patients with type 2 diabetes mellitus on oral antidiabetic drugs. Postgrad Med. 2012;124(1):124–132.

SCHWARTZ_proof

Page 18: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

Stanley Schwartz

18 © Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]

Warning: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

162. Davies MJ, Chubb BD, Smith IC, Valentine WJ. Cost-utility analysis of liraglutide compared with sulphonylurea or sitagliptin, all as add-on to metformin monotherapy in Type 2 diabetes mellitus. Diabet Med. 2012;29(3):313–320.

163. Elgart JF, Caporale JE, Gonzalez L, Aiello E, Waschbusch M, Gagliardino JJ. Treatment of type 2 diabetes with saxagliptin: a pharma-coeconomic evaluation in Argentina. Health Econ Rev. 2013;3(1):11.

164. Minshall ME, Oglesby AK, Wintle ME, Valentine WJ, Roze S, Palmer AJ. Estimating the long-term cost-effectiveness of exenatide in the United States: an adjunctive treatment for type 2 diabetes mellitus. Value Health. 2008;11(1):22–33.

165. Schwartz S. Targeting the pathophysiology of type 2 diabetes: rationale for combination therapy with pioglitazone and exenatide. Curr Med Res Opin. 2008;24(11):3009–3022.

166. Sullivan SD, Alfonso-Cristancho R, Conner C, Hammer M, Blonde L. A simulation of the comparative long-term effectiveness of liraglutide and glimepiride monotherapies in patients with type 2 diabetes mel-litus. Pharmacotherapy. 2009;29(11):1280–1288.

167. Gao L, Zhao FL, Li SC. Cost-utility analysis of liraglutide versus glimepiride as add-on to metformin in type 2 diabetes patients in China. Int J Technol Assess Health Care. 2012;28(4):436–444.

168. Erhardt W, Bergenheim K, Duprat-Lomon I, McEwan P. Cost effective-ness of saxagliptin and metformin versus sulfonylurea and metformin in the treatment of type 2 diabetes mellitus in Germany: a Cardiff diabetes model analysis. Clin Drug Investig. 2012;32(3):189–202.

169. Simpson SH, Majumdar SR, Tsuyuki RT, Eurich DT, Johnson JA. Dose-response relation between sulfonylurea drugs and mortality in type 2 diabetes mellitus: a population-based cohort study. CMAJ. 2006;174(2):169–174.

170. Sanon S, Patel R, Eshelbrenner C, et al. Acute coronary syndrome in patients with diabetes mellitus: perspectives of an interventional cardiologist. Am J Cardiol. 2012;110(9 Suppl):13B–23B.

171. Margolis DJ, Hoffstad O, Strom BL. Association between serious ischemic cardiac outcomes and medications used to treat diabetes. Pharmacoepidemiol Drug Saf. 2008;17(8):753–759.

172. Tzoulaki I, Molokhia M, Curcin V, et al. Risk of cardiovascular disease and all cause mortality among patients with type 2 diabetes prescribed oral antidiabetes drugs: retrospective cohort study using UK general practice research database. BMJ. 2009;339:b4731. Available from: http://www.bmj.com/content/339/bmj.b4731.pdf%2Bhtml. Accessed January 28, 2014.

173. Brady PA, Terzic A. The sulfonylurea controversy: more questions from the heart. J Am Coll Cardiol. 1998;31(5):950–956.

174. Del Prato S, Felton AM, Munro N, et al; Global Partnership for Effec-tive Diabetes Management. Improving glucose management: ten steps to get more patients with type 2 diabetes to glycaemic goal. Int J Clin Pract. 2005;59(11):1345–1355.

175. Arechavaleta R, Seck T, Chen Y, et al. Efficacy and safety of treatment with sitagliptin or glimepiride in patients with type 2 diabetes inadequately controlled on metformin monotherapy: a randomized, double-blind, non-inferiority trial. Diabetes Obes Metab. 2011;13(2):160–168.

176. Wu D, Li L, Liu C. Efficacy and safety of dipeptidyl peptidase-4 inhibi-tors and metformin as initial combination therapy and as monotherapy in patients with type 2 diabetes mellitus: a meta-analysis. Diabetes Obes Metab. 2014;16(1):30–37.

177. Migoya EM, Bergeron R, Miller JL, et al. Dipeptidyl peptidase-4 inhib-itors administered in combination with metformin result in an additive increase in the plasma concentration of active GLP-1. Clin Pharmacol Ther. 2010;88(6):801–808.

178. Abdul-Ghani MA, Merovci A, Solis-Herrera C, et al. Plasma glucose reduction with the SGLT2 inhibitor, dapagliflozin, improves insulin sensitivity and insulin secretion in T2DM [abstract 242-OR]. Diabetes. 2013;62(Suppl 1):A62.

179. Abdul-Ghani MA, Puckett C, Adams J, Cersosimo E, Triplitt C, DeFronzo RA. Initial triple combination therapy is superior to stepwise add-on conventional therapy in newly diagnosed T2DM [abstract 72-OR]. Diabetes. 2013;62(Suppl 1):A18.

180. Schwartz SS. A practice-based approach to the 2012 position statement of the American Diabetes Association and the European Association for the Study of Diabetes. Curr Med Res Opin. 2013;29(7): 793–799.

181. Tzefos M, Olin JL. Glucagon-like peptide-1 analog and insulin com-bination therapy in the management of adults with type 2 diabetes mellitus. Ann Pharmacother. 2010;44(7–8):1294–1300.

182. Nayak UA, Govindan J, Baskar V, Kalupahana D, Singh BM. Exenatide therapy in insulin-treated type 2 diabetes and obesity. QJM. 2010;103(9):687–694.

183. Arnolds S, Dellweg S, Clair J, et al. Further improvement in post-prandial glucose control with addition of exenatide or sitagliptin to combination therapy with insulin glargine and metformin: a proof-of-concept study. Diabetes Care. 2010;33(7):1509–1515.

184. Govindan J, Healey B, Kalupahana DN, et al. Exenatide therapy in insulin treated patients with type 2 diabetes and obesity [abstract 884]. Diabetologia. 2008;51(suppl 1):S352–S353.

185. Schwartz SS. Do many folks with T2D REALLY need insulin? In: Draznin B, ed. Diabetes Case File: Practical Problems, Real Solutions. Alexandria, VA: American Diabetes Association, 2015; in press.

186. Gonzalez-Ortiz M, Martinez-Abundis E, Robles-Cervantes JA, Ramos-Zavala MG. Effect of exenatide on fat deposition and a metabolic profile in patients with metabolic syndrome. Metab Syndr Relat Disord. 2011;9(1):31–34.

187. Bhushan R, Elkind-Hirsch KE, Bhushan M, Butler WJ, Duncan K, Marrioneaux O. Improved glycemic control and reduction of cardio-metabolic risk factors in subjects with type 2 diabetes and metabolic syndrome treated with exenatide in a clinical practice setting. Diabetes Technol Ther. 2009;11(6):353–359.

188. Nauck MA, Duran S, Kim D, et al. A comparison of twice-daily exenatide and biphasic insulin aspart in patients with type 2 diabetes who were suboptimally controlled with sulfonylurea and metformin: a non-inferiority study. Diabetologia. 2007;50(2):259–267.

189. Barnett AH, Burger J, Johns D, et al. Tolerability and efficacy of exenatide and titrated insulin glargine in adult patients with type 2 diabetes previously uncontrolled with metformin or a sulfonylurea: a multinational, randomized, open-label, two-period, crossover non-inferiority trial. Clin Ther. 2007;29(11):2333–2348.

190. Buse JB, Bergenstal RM, Glass LC, et al. Use of twice-daily exenatide in Basal insulin-treated patients with type 2 diabetes: a randomized, controlled trial. Ann Intern Med. 2011;154(2):103–112.

191. Li CJ, Li J, Zhang QM, et al. Efficacy and safety comparison between liraglutide as add-on therapy to insulin and insulin dose-increase in Chinese subjects with poorly controlled type 2 diabetes and abdominal obesity. Cardiovasc Diabetol. 2012;11:142.

192. Mathieu C, Rodbard HW, Cariou B, et al; on behalf of the BEGIN: VICTOZA ADD-ON (NN1250–3948) Study Group. A comparison of adding liraglutide versus a single daily dose of insulin aspart to insulin degludec in subjects with type 2 diabetes (BEGIN: VICTOZA ADD-ON) [published online ahead of print January 20, 2014]. Dia-betes Obes Metab. 2014. http://onlinelibrary.wiley.com/doi/10.1111/dom.12262/pdf. Accessed March 31, 2014.

193. Balena R, Hensley IE, Miller S, Barnett AH. Combination therapy with GLP-1 receptor agonists and basal insulin: a systematic review of the literature. Diabetes Obes Metab. 2013;15(6):485–502.

194. Rizos EC, Ntzani EE, Papanas N, et al. Combination therapies of DPP4 inhibitors and GLP1 analogues with insulin in type 2 diabetic patients: a systematic review. Curr Vasc Pharmacol. 2013;11(6):992–1000.

195. Vora J, Bain SC, Damci T, et al. Incretin-based therapy in combina-tion with basal insulin: a promising tactic for the treatment of type 2 diabetes. Diabetes Metab. 2013;39(1):6–15.

196. Suen CS, Burn P. The potential of incretin-based therapies in type 1 diabetes. Drug Discov Today. 2012;17(1–2):89–95.

197. Varanasi A, Bellini N, Rawal D, et al. Liraglutide as additional treatment for type 1 diabetes. Eur J Endocrinol. 2011;165(1): 77–84.

SCHWARTZ_proof

Page 19: Evidence-Based Practice Use of Incretin-Based Therapy in ......, 0.01 vs controls). 43 In another study in patients with CHD and preserved LV function, a 48-hour infusion of GLP-1

Incretin-Based Therapy in Patients With Diabetes

© Postgraduate Medicine, Volume 126, Issue 3, May 2014, ISSN – 0032-5481, e-ISSN – 1941-9260 19ResearchSHARE®: www.research-share.com • Permissions: [email protected] • Reprints: [email protected]: No duplication rights exist for this journal. Only JTE Multimedia, LLC holds rights to this publication. Please contact the publisher directly with any queries.

198. Raman VS, Mason KJ, Rodriguez LM, et al. The role of adjunctive exenatide therapy in pediatric type 1 diabetes. Diabetes Care. 2010;33(6):1294–1296.

199. Ellis SL, Moser EG, Snell-Bergeon JK, Rodionova AS, Hazenfield RM, Garg SK. Effect of sitagliptin on glucose control in adult patients with Type 1 diabetes: a pilot, double-blind, randomized, crossover trial. Diabet Med. 2011;28(10):1176–1181.

200. Farngren J, Persson M, Schweizer A, Foley JE, Ahren B. Vildaglip-tin reduces glucagon during hyperglycemia and sustains glucagon counterregulation during hypoglycemia in type 1 diabetes. J Clin Endocrinol Metab. 2012;97(10):3799–3806.

201. Schwartz S, DeFronzo R. The use of non-insulin anti-diabetic agents to improve glycemia without hypoglycemia in the hospital setting: focus on incretins. Curr Diab Rep. 2014;13(3):466.

202. Abrahamsson N, Engstrom BE, Sundbom M, Karlsson FA. GLP1 analogs as treatment of postprandial hypoglycemia following gas-tric bypass surgery: a potential new indication? Eur J Endocrinol. 2013;169(6):885–889.

203. van Hateren KJ, Landman GW, Kleefstra N, et al. Glycaemic control and the risk of mortality in elderly type 2 diabetic patients (ZODIAC-20). Int J Clin Pract. 2011;65(4):415–419.

SCHWARTZ_proof