Most treatment guidelines, including those from the American Diabetes Association/European Association for the Study of Diabetes and the International Diabetes Federation, suggest metformin be used as the first-line therapy after diet and exercise. This recommendation is based on the considerable body of evidence that has accumulated over the last 30 years, but it is also supported on clinical grounds based on metformin’s affordability and tolerability. As such, metformin is the most commonly used oral antihyperglycemic agent in the U.S. However, based on the release of newer agents over the recent past, some have suggested that the modern approach to disease management should be based upon identification of its etiology and correcting the underlying biological disturbances. That is, we should use interventions that normalize or at least ameliorate the recognized derangements in physiology that drive the clinical manifestation of disease, in this circumstance, hyperglycemia. Thus, it is argued that therapeutic interventions that target glycemia but do not correct the underlying pathogenic disturbances are unlikely to result in a sustained benefit on the disease process. In our field, there is an evolving debate regarding the suggested first step in diabetes management and a call for a new paradigm. Given the current controversy, we provide a Point-Counterpoint debate on this issue. In the point narrative below that precedes the counterpoint narrative, Drs. Abdul-Ghani and DeFronzo provide their argument that a treatment approach for type 2 diabetes based upon correcting the underlying pathophysiological abnormalities responsible for the development of hyperglycemia provides the best therapeutic strategy. Such an approach requires a change in the recommendation for first-line therapy from metformin to a GLP-1 receptor agonist. In the counterpoint narrative that follows Drs. Abdul-Ghani and DeFronzo’s contribution, Dr. Inzucchi argues that, based on the medical community’s extensive experience and the drug’s demonstrated efficacy, safety, low cost, and cardiovascular benefits, metformin should remain the “foundation therapy” for all patients with type 2 diabetes, barring contraindications.

—William T. Cefalu

Chief Scientific, Medical & Mission Officer, American Diabetes Association

The modern approach to disease management is based upon identification of its etiology and correcting the underlying pathophysiological disturbances with interventions that ameliorate/normalize known defects responsible for the clinical manifestation of the disease, i.e., hyperglycemia. Therapeutic interventions that simply target hyperglycemia but do not correct the underlying pathogenic disturbances are unlikely to result in a sustained reduction in HbA1c. It is well established that type 2 diabetes (T2D) is a complex metabolic/cardiovascular disorder with at least eight distinct pathophysiological disturbances, referred to as the Ominous Octet (1). Hyperglycemia is a manifestation of these eight pathophysiological abnormalities. Nonetheless, the current recommended approach in T2D management still focuses on lowering the plasma glucose concentration rather than correcting the underlying metabolic abnormalities that cause the hyperglycemia (24). Therefore, it is not surprising that current therapeutic guidelines (24) do not result in a sustained HbA1c reduction (58). In this Point-Counterpoint, we argue that it is time to apply the modern concepts of clinical practice to diabetes management and base therapy on pathophysiology. Thereby, GLP-1 receptor agonists (GLP-1 RAs), which 1) correct six of the eight components of the Ominous Octet, 2) prevent/reverse the progressive β-cell failure and rise in HbA1c, and 3) lower cardiovascular risk in T2D independent of their glucose-lowering ability (9,10), should replace metformin as the recommended first-line therapy in newly diagnosed T2D patients.

The etiology of T2D is complex and involves multiple pathophysiological disturbances involving multiple organs (1) (Fig. 1). Insulin resistance in skeletal muscle, liver, and adipocytes (1115) and β-cell dysfunction (1622) remain the major core defects responsible for the development and progression of hyperglycemia. Insulin resistance is also associated with multiple metabolic abnormalities, e.g., hypertension, dyslipidemia, endothelial dysfunction, procoagulant state, inflammation, and visceral obesity, which collectively are known as the insulin resistance (metabolic) syndrome (2325). Each individual component of the insulin resistance syndrome, as well as the basic molecular etiology of the insulin resistance (25), is causally related to the development of atherosclerotic cardiovascular disease (CVD) and contributes to the increased risk for CVD in T2D patients.

Figure 1

GLP-1 RAs correct six components of the Ominous Octet, whereas metformin corrects only one component.

Figure 1

GLP-1 RAs correct six components of the Ominous Octet, whereas metformin corrects only one component.

Close modal

Because progressive β-cell failure is the principal factor responsible for the development and progression of hyperglycemia in T2D patients (1,1619), only therapies that halt/reverse the progressive β-cell failure will be effective in lowering and maintaining HbA1c at the target level, and ideally this should be accomplished without increasing the risk of hypoglycemia.

In addition to insulin resistance and β-cell dysfunction, impaired incretin effect in T2D plays a major role in the progression of β-cell failure and hyperglycemia (26,27). Further, T2D patients have elevated fasting plasma glucagon levels that fail to suppress normally after a meal and enhanced hepatic sensitivity to glucagon (28,29), in part due to resistance to GLP-1 (26,30); these pathophysiological abnormalities can be reversed with GLP-1 RA therapy (26,31).

Activation of GLP-1 receptors in the β-cell amplifies glucose-stimulated insulin secretion but only under conditions of hyperglycemia (26,32), whereas in the α-cell, GLP-1 suppresses glucagon secretion, leading to correction of postmeal hyperglycemia in T2D (26,27,30). GLP-1 RAs improve β-cell function by enhancing β-cell responsiveness to glucose, i.e., improving β-cell glucose sensitivity; this beneficial effect on the β-cell can be observed within 8 h after a single injection of the GLP-1 RA (liraglutide) (33), is maintained at 3 months (semaglutide) (34), and persists for at least 3 years (exenatide) (35). Thus, GLP-1 RAs produce a rapid and durable reduction in HbA1c with low risk of hypoglycemia (36,37).

GLP-1 also exerts multiple nonglycemic actions, all of which improve metabolic control in T2D patients (Table 1), including 1) delayed gastric emptying, which slows the absorption of ingested glucose (32), 2) appetite suppression, which promotes weight loss (26,38,39), 3) reduction of hepatic and visceral fat content (40), making them an attractive intervention to prevent/reverse nonalcoholic fatty liver disease/nonalcoholic steatohepatitis (41), and 4) prevention of diabetic nephropathy in Liraglutide Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results—A Long Term Evaluation (LEADER) (42). Recent evidence suggests that gut stimulation of GLP-1 secretion by the L cells is an important mechanism via which metformin suppresses hepatic glucose production (43,44).

Table 1

Metabolic actions of GLP-1 RAs

• Pancreas 
 Potentiate glucose-mediated insulin secretion 
 Preserve β-cell function/reverse β-cell failure 
 Inhibit glucagon secretion in a glucose-dependent fashion 
• Cardiovascular system 
 Reduce MACE 
 Reduce systolic blood pressure 
 Reduce pulmonary capillary wedge pressure 
 Increase myocardial salvage following myocardial infarction 
 Improve endothelial dysfunction 
• GI 
 Slow gastric emptying 
 Inhibit hepatic glucose production 
 Decrease liver fat content 
 Decrease visceral fat 
• Central nervous system 
 Suppress appetite 
• Kidney 
 Preserve renal function 
 Increase sodium excretion 
• General 
 Promote weight loss 
• Pancreas 
 Potentiate glucose-mediated insulin secretion 
 Preserve β-cell function/reverse β-cell failure 
 Inhibit glucagon secretion in a glucose-dependent fashion 
• Cardiovascular system 
 Reduce MACE 
 Reduce systolic blood pressure 
 Reduce pulmonary capillary wedge pressure 
 Increase myocardial salvage following myocardial infarction 
 Improve endothelial dysfunction 
• GI 
 Slow gastric emptying 
 Inhibit hepatic glucose production 
 Decrease liver fat content 
 Decrease visceral fat 
• Central nervous system 
 Suppress appetite 
• Kidney 
 Preserve renal function 
 Increase sodium excretion 
• General 
 Promote weight loss 

CVD is the leading cause of death in T2D patients (45), accounting for ∼80% of mortality, and T2D is best viewed as a cardiometabolic disorder (25,46). Thus, reducing CVD risk is a high priority in T2D management, and reduction in blood pressure and correction of diabetic dyslipidemia are essential components of diabetes management. Considerable evidence documents that hyperglycemia is a weak risk factor for cardiovascular complications and improving glucose control has little benefit on macrovascular disease risk (UK Prospective Diabetes Study [UKPDS], Action to Control Cardiovascular Risk in Diabetes [ACCORD], Action in Diabetes and Vascular Disease: Preterax and Diamicron MR Controlled Evaluation [ADVANCE], Veterans Affairs Diabetes Trial [VADT]), especially when it is well established. Numerous clinical trials have demonstrated that antidiabetes agents that reduce plasma glucose without altering other cardiovascular risk factors fail to reduce CVD risk in T2D patients. Conversely, antidiabetes medications that in addition to lowering the plasma glucose concentration also improve cardiovascular risk factors, e.g., GLP-1 RAs (9,10), pioglitazone (47,48), and SGLT2 inhibitors (49,50), significantly reduce cardiovascular events in T2D with established CVD. Thus, these agents should be favored over agents that lower plasma glucose but have no effect on cardiovascular risk factors or CVD, e.g., sulfonylureas (51,52), DPP-4 inhibitors (5355), and insulin (56) (Fig. 2). GLP-1 RAs consistently have been shown to reduce many CVD risk factors (Table 2) (25,34,5760). Thus, it is not surprising that two large, prospective, randomized, double-blind, placebo-controlled trials (9,10) have demonstrated that liraglutide and semaglutide significantly lower the incidence of 3-point MACE (major adverse cardiovascular events), which includes nonfatal myocardial infarction, nonfatal stroke, and cardiovascular death, by 13% and 24%, respectively, in T2D patients with existing CVD. Of note, despite the high CVD risk in the patient populations in both LEADER and Trial to Evaluate Cardiovascular and Other Long-term Outcomes With Semaglutide in Subjects With Type 2 Diabetes (SUSTAIN-6), all cardiovascular risk factors, including blood pressure and LDL cholesterol, were well controlled at baseline, consistent with the high number of patients receiving statins, ACE inhibitors, angiotensin receptor blockers, and aspirin therapy. Addition of the GLP-1 RA to the patients’ antidiabetes treatment resulted in only modest reductions in HbA1c (0.4%) and systolic blood pressure (∼2–3 mmHg) in LEADER and SUSTAIN-6; these glucose- and blood pressure–lowering effects are quite modest and unlikely to explain the CVD benefit of liraglutide and semaglutide. This suggests the GLP-1 RAs may have a direct beneficial action to slow the atherosclerotic process, independent of their effect to reduce glycemia and improve traditional cardiovascular risk factors (59).

Figure 2

Not all antidiabetes agents are equal in their ability to reduce cardiovascular risk.

Figure 2

Not all antidiabetes agents are equal in their ability to reduce cardiovascular risk.

Close modal
Table 2

Benefits of GLP-1 RAs far outweigh those of metformin

GLP-1 RAsMetformin
Pathophysiological defects in T2D (see Fig. 1Corrects six of the defects Corrects only one of the defects 
Glucose-lowering efficacy Strong Strong 
Durability of HbA1c reduction Strong None 
Weight loss 3–4 kg 1–2 kg 
Blood pressure ∼2–3 mmHg reduction Neutral 
Lipid profile Lowers triglycerides, increases HDL cholesterol Neutral 
Cardiovascular protection (MACE) Reduction by 13–26% Neutral 
Renal protection Reduction by 22% Neutral 
Tolerability ∼10–15% GI side effects ∼10–15% GI side effects 
Dosing Weekly subcutaneous injection Once to twice daily oral administration 
Cost High Low 
GLP-1 RAsMetformin
Pathophysiological defects in T2D (see Fig. 1Corrects six of the defects Corrects only one of the defects 
Glucose-lowering efficacy Strong Strong 
Durability of HbA1c reduction Strong None 
Weight loss 3–4 kg 1–2 kg 
Blood pressure ∼2–3 mmHg reduction Neutral 
Lipid profile Lowers triglycerides, increases HDL cholesterol Neutral 
Cardiovascular protection (MACE) Reduction by 13–26% Neutral 
Renal protection Reduction by 22% Neutral 
Tolerability ∼10–15% GI side effects ∼10–15% GI side effects 
Dosing Weekly subcutaneous injection Once to twice daily oral administration 
Cost High Low 

The above review demonstrates that GLP-1 RAs directly and/or indirectly correct/improve six of the eight pathophysiological defects responsible for hyperglycemia in T2D, improve cardiovascular risk factors, and reduce MACE in two large, well-designed, prospective cardiovascular intervention trials.

GLP-1 RAs correct six members of the Ominous Octet, whereas the only known action of metformin is to inhibit hepatic glucose production (61) (Fig. 1 and Table 2). Contrary to common belief, metformin is not an insulin sensitizer in muscle or adipocytes (6163) in the absence of weight loss, which is a frequent occurrence in patients treated with the biguanide (Fig. 3A). Consistent with this, following intravenous administration of 11C-metformin, none of the biguanide can be detected in muscle (64). Most importantly, and in direct contrast to the GLP-1 RAs, metformin lacks any effect on β-cell function (61,62) (Fig. 3B), which is the primary pathophysiological disturbance responsible for progressive hyperglycemia in T2D patients (1). This is most graphically demonstrated in the UKPDS (65) and A Diabetes Outcome Progression Trial (ADOPT) (5) (Fig. 3D) in which, after an initial decline during the first year, HbA1c rose progressively because of progressive β-cell failure. This stands in marked contrast to the GLP-1 RAs, which exert a potent protective effect on the β-cell that persists for at least 3 years (34). Because the GLP-1 RAs cause significant weight loss, they also improve insulin sensitivity in muscle. Thus, GLP-1 RAs, but not metformin, correct the two major core defects in T2D patients, i.e., β-cell dysfunction and muscle insulin resistance. The major mechanism of action of metformin to reduce glycemia is inhibition of hepatic gluconeogenesis (61,62) (Fig. 3C), whereas the GLP-1 RAs also effectively reduce hepatic glucose production but by multiple other mechanisms, i.e., inhibition of glucagon secretion, stimulation of insulin secretion, direct effect on the liver, and depletion of liver fat (26,27,30,59,6668).

Figure 3

Effect of metformin on glycemic control, insulin secretion, and insulin sensitivity in T2D. A: Metformin does not improve muscle insulin sensitivity (measured with euglycemic insulin clamp) in T2D individuals (n = 20) in the absence of weight loss (72). B: Metformin has no effect on β-cell function in T2D individuals (n = 14) (measured with an oral glucose tolerance test [OGTT] and hyperglycemic clamp) (73). C: The primary effect via which metformin reduces the HbA1c in T2D is related to the suppression of hepatic glucose production (HGP) via inhibition of gluconeogenesis (72). FPG, fasting plasma glucose. D: Effect of metformin on HbA1c. Because metformin does not affect muscle insulin sensitivity or β-cell function, following an initial decline after metformin administration, the HbA1c rises progressively in T2D patients (5,6,74). KPNW, Kaiser Permanente Northwest.

Figure 3

Effect of metformin on glycemic control, insulin secretion, and insulin sensitivity in T2D. A: Metformin does not improve muscle insulin sensitivity (measured with euglycemic insulin clamp) in T2D individuals (n = 20) in the absence of weight loss (72). B: Metformin has no effect on β-cell function in T2D individuals (n = 14) (measured with an oral glucose tolerance test [OGTT] and hyperglycemic clamp) (73). C: The primary effect via which metformin reduces the HbA1c in T2D is related to the suppression of hepatic glucose production (HGP) via inhibition of gluconeogenesis (72). FPG, fasting plasma glucose. D: Effect of metformin on HbA1c. Because metformin does not affect muscle insulin sensitivity or β-cell function, following an initial decline after metformin administration, the HbA1c rises progressively in T2D patients (5,6,74). KPNW, Kaiser Permanente Northwest.

Close modal

Although the UKPDS demonstrated that metformin caused a reduction in cardiovascular events in T2D patients (65), the patient population consisted of a small number of obese T2D subjects (n = 342); these results, by today’s standards, would never be accepted as evidence for a cardiovascular benefit of the biguanide. Moreover, a beneficial effect on cardiovascular events was not observed in other clinical studies with metformin, i.e., the ADOPT study (5), which included twice the number of patients as the UKPDS (n = 818). To the contrary, subjects receiving metformin in ADOPT experienced more cardiovascular events than subjects receiving glyburide, although this difference was not statistically significant. This emphasizes the problem of interpreting results from studies that are markedly underpowered to detect clinically significant differences in cardiac event rates. Conversely, the CVD benefit of GLP-1 RAs has conclusively been demonstrated in two very large, prospective cardiovascular intervention trials, LEADER and SUSTAIN-6 (9,10).

With regard to safety, both metformin and GLP-1 RAs are associated with gastrointestinal (GI) adverse events. Approximately 15–20% of T2D patients do not tolerate metformin because of GI side effects (66). The incidence of GI side effects with the long-acting GLP-1 RAs is similar to that of metformin. Further, and unlike those with metformin, the GI side effects usually are mild to moderate, waning over the first 4–6 weeks of initiating therapy. The percentage of patients who discontinue long-acting GLP-1 RAs because of GI side effects is significantly lower than that of metformin (67). Some postmarketing reports have suggested an increased risk of acute pancreatitis with GLP-1 RA use. However, three large, prospective, double-blind, placebo-controlled clinical trials including ∼20,000 patients followed for 2–4 years have demonstrated no increased risk of acute pancreatitis with GLP-1 RA use (9,10,68).

Metformin is administered orally versus via injection for GLP-1 RAs. However, intermediate-acting metformin requires multiple daily dosing, whereas two long-acting GLP-1 RAs (exenatide and dulaglutide) are available as weekly injections and a third (semaglutide) is under review by the U.S. Food and Drug Administration. A subcutaneously implanted osmotic mini pump that continuously delivers exenatide for 6 months is expected to be approved within the next year (69), and an oral formulation of the GLP-1 RA semaglutide is in phase 3 trials (70) and is anticipated to be available within 3–4 years. These modern delivery methods will improve patient compliance for GLP-1 RAs versus metformin.

Lastly, metformin is generic and inexpensive, whereas GLP-1 RAs are still under patent and, therefore, expensive. However, most large health care plans have at least one GLP-1 RA on formulary with a modest copay. Moreover, liraglutide (Victoza) is expected to become generic by the end of 2017, and this should significantly reduce its cost. A cost-effective analysis is beyond the scope of this discussion, and the appropriate long-term, clinical, real-world studies to perform such an analysis are not available. However, a recent cost analysis for the treatment of T2D patients in the U.S. by the American Diabetes Association (71) demonstrated that only a small portion (12%) of the cost of T2D is due to the direct cost of antihyperglycemic medications. The vast majority of the cost of diabetes care is related to the development of diabetic vascular complications, with CVD disease contributing 50% of that cost, and two recent studies, LEADER and SUSTAIN-6, have demonstrated that GLP-1 RAs decrease cardiovascular events. Further, the cost of medications to treat the complications of diabetes is 50% greater than the cost of antihyperglycemic medications. It remains to be determined whether, on a long-term basis, the use of GLP-1 RAs, which in addition to causing a durable reduction in the plasma glucose concentration (thereby decreasing microvascular complications) also reduce cardiovascular events, will be cost-effective.

In summary, the currently available clinical and scientific evidence (Table 2) is overwhelmingly in favor of the use of GLP-1 RAs over metformin as first-line therapy in newly diagnosed T2D patients.

Funding. M.A.-G. receives funding from the National Institutes of Health (DK 097554-01) and the Qatar Foundation (National Priorities Research Program 4-248-3-076), and R.A.D. receives funding from the National Institutes of Health (DK 024092-42). R.A.D.’s salary is, in part, supported by the South Texas Veterans Health Care System, Audie L. Murphy Division.

Duality of Interest. R.A.D. is on the advisory board of AstraZeneca, Janssen, Novo Nordisk, Boehringer Ingelheim, and Intarcia. R.A.D. has received grants from AstraZeneca, Janssen, and Boehringer Ingelheim. R.A.D. is a member of the speakers’ bureau for AstraZeneca and Novo Nordisk. No other potential conflicts of interest relevant to this article were reported.

See accompanying article, p. 1128.

1.
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
:
773
795
2.
Inzucchi
SE
,
Bergenstal
RM
,
Buse
JB
, et al
.
Management of hyperglycemia in type 2 diabetes, 2015: a patient-centered approach: update to a position statement of the American Diabetes Association and the European Association for the Study of Diabetes
.
Diabetes Care
2015
;
38
:
140
149
3.
Garber
AJ
,
Abrahamson
MJ
,
Barzilay
JI
, et al.;
American Association of Clinical Endocrinologists (AACE); American College of Endocrinology (ACE)
.
Consensus statement of the American Association of Clinical Endocrinology and American College of Endocrinology on the comprehensive type 2 diabetes algorithm–2015 executive summary
.
Endocr Pract
2015
;
21
:
1403
1414
4.
International Diabetes Federation
Clinical Guidelines Task Force
.
Global Guideline for Type 2 Diabetes
.
Brussels, Belgium
,
International Diabetes Federation
,
2012
, p.
44
46
5.
Kahn
SE
,
Haffner
SM
,
Heise
MA
, et al.;
ADOPT Study Group
.
Glycemic durability of rosiglitazone, metformin, or glyburide monotherapy
.
N Engl J Med
2006
;
355
:
2427
2443
6.
United Kingdom Prospective Diabetes Study (UKPDS). 13: relative efficacy of randomly allocated diet, sulphonylurea, insulin, or metformin in patients with newly diagnosed non-insulin dependent diabetes followed for three years
.
BMJ
1995
;
310
:
83
88
7.
Abdul-Ghani
MA
,
Puckett
C
,
Triplitt
C
, et al
.
Initial combination therapy with metformin, pioglitazone and exenatide is more effective than sequential add-on therapy in subjects with new-onset diabetes. Results from the Efficacy and Durability of Initial Combination Therapy for Type 2 Diabetes (EDICT): a randomized trial
.
Diabetes Obes Metab
2015
;
17
:
268
275
8.
Abdul-Ghani
M
,
Mujahid
O
,
Mujahid
A
, et al
.
Combination therapy with exenatide plus pioglitazone versus basal/bolus insulin in patients with poorly controlled type 2 diabetes on sulfonylurea plus metformin: the QATAR Study
.
Diabetes Care
2017
;
40
:
325
331
9.
Marso
SP
,
Daniels
GH
,
Brown-Frandsen
K
, et al.;
LEADER Steering Committee
;
LEADER Trial Investigators
.
Liraglutide and cardiovascular outcomes in type 2 diabetes
.
N Engl J Med
2016
;
375
:
311
322
10.
Marso
SP
,
Bain
SC
,
Consoli
A
, et al.;
SUSTAIN-6 Investigators
.
Semaglutide and cardiovascular outcomes in patients with type 2 diabetes
.
N Engl J Med
2016
;
375
:
1834
1844
11.
Abdul-Ghani
MA
,
DeFronzo
RA
.
Pathogenesis of insulin resistance in skeletal muscle
.
J Biomed Biotechnol
2010
;
2010
:
476279
12.
Groop
LC
,
Widén
E
,
Ferrannini
E
.
Insulin resistance and insulin deficiency in the pathogenesis of type 2 (non-insulin-dependent) diabetes mellitus: errors of metabolism or of methods?
Diabetologia
1993
;
36
:
1326
1331
13.
Shalata
A
,
Jazmawi
W
,
Aslan
O
, et al
.
Early metabolic defects in Arab subjects with strong family history of type 2 diabetes
.
J Endocrinol Invest
2013
;
36
:
417
421
14.
Reaven
GM
.
Relationships among insulin resistance, type 2 diabetes, essential hypertension, and cardiovascular disease: similarities and differences
.
J Clin Hypertens (Greenwich)
2011
;
13
:
238
243
15.
Kashyap
S
,
Belfort
R
,
Gastaldelli
A
, et al
.
A sustained increase in plasma free fatty acids impairs insulin secretion in nondiabetic subjects genetically predisposed to develop type 2 diabetes
.
Diabetes
2003
;
52
:
2461
2474
16.
Kahn
SE
,
Cooper
ME
,
Del Prato
S
.
Pathophysiology and treatment of type 2 diabetes: perspectives on the past, present, and future
.
Lancet
2014
;
383
:
1068
1083
17.
Alejandro
EU
,
Gregg
B
,
Blandino-Rosano
M
,
Cras-Méneur
C
,
Bernal-Mizrachi
E
.
Natural history of β-cell adaptation and failure in type 2 diabetes
.
Mol Aspects Med
2015
;
42
:
19
41
18.
Halban
PA
,
Polonsky
KS
,
Bowden
DW
, et al
.
β-Cell failure in type 2 diabetes: postulated mechanisms and prospects for prevention and treatment
.
Diabetes Care
2014
;
37
:
1751
1758
19.
Saad
MF
,
Knowler
WC
,
Pettitt
DJ
,
Nelson
RG
,
Mott
DM
,
Bennett
PH
.
The natural history of impaired glucose tolerance in the Pima Indians
.
N Engl J Med
1988
;
319
:
1500
1506
20.
Ferrannini
E
,
Gastaldelli
A
,
Miyazaki
Y
,
Matsuda
M
,
Mari
A
,
DeFronzo
RA
. β
-Cell function in subjects spanning the range from normal glucose tolerance to overt diabetes: a new analysis
.
J Clin Endocrinol Metab
2005
;
90
:
493
500
21.
Ferrannini
E
,
Mari
A
.
β-Cell function in type 2 diabetes
.
Metabolism
2014
;
63
:
1217
1227
22.
Polonsky
KS
.
Lilly Lecture 1994. The β-cell in diabetes: from molecular genetics to clinical research
.
Diabetes
1995
;
44
:
705
717
23.
Jornayvaz
FR
,
Samuel
VT
,
Shulman
GI
.
The role of muscle insulin resistance in the pathogenesis of atherogenic dyslipidemia and nonalcoholic fatty liver disease associated with the metabolic syndrome
.
Annu Rev Nutr
2010
;
30
:
273
290
24.
Reaven
GM
.
Insulin resistance: the link between obesity and cardiovascular disease
.
Med Clin North Am
2011
;
95
:
875
892
25.
DeFronzo
RA
.
Insulin resistance, lipotoxicity, type 2 diabetes and atherosclerosis: the missing links. The Claude Bernard Lecture 2009
.
Diabetologia
2010
;
53
:
1270
1287
26.
Nauck
MA
,
Meier
JJ
.
The incretin effect in healthy individuals and those with type 2 diabetes: physiology, pathophysiology, and response to therapeutic interventions
.
Lancet Diabetes Endocrinol
2016
;
4
:
525
536
27.
Holst
JJ
,
Gribble
F
,
Horowitz
M
,
Rayner
CK
.
Roles of the gut in glucose homeostasis
.
Diabetes Care
2016
;
39
:
884
892
28.
Matsuda
M
,
DeFronzo
RA
,
Glass
L
, et al
.
Glucagon dose-response curve for hepatic glucose production and glucose disposal in type 2 diabetic patients and normal individuals
.
Metabolism
2002
;
51
:
1111
1119
29.
Knop
FK
,
Vilsbøll
T
,
Madsbad
S
,
Holst
JJ
,
Krarup
T
.
Inappropriate suppression of glucagon during OGTT but not during isoglycaemic i.v. glucose infusion contributes to the reduced incretin effect in type 2 diabetes mellitus
.
Diabetologia
2007
;
50
:
797
805
30.
Sandoval
DA
,
D’Alessio
DA
.
Physiology of proglucagon peptides: role of glucagon and GLP-1 in health and disease
.
Physiol Rev
2015
;
95
:
513
548
31.
Gamble
JM
,
Clarke
A
,
Myers
KJ
, et al
.
Incretin-based medications for type 2 diabetes: an overview of reviews
.
Diabetes Obes Metab
2015
;
17
:
649
658
32.
Umapathysivam
MM
,
Lee
MY
,
Jones
KL
, et al
.
Comparative effects of prolonged and intermittent stimulation of the glucagon-like peptide 1 receptor on gastric emptying and glycemia
.
Diabetes
2014
;
63
:
785
790
33.
Chang
AM
,
Jakobsen
G
,
Sturis
J
, et al
.
The GLP-1 derivative NN2211 restores β-cell sensitivity to glucose in type 2 diabetic patients after a single dose
.
Diabetes
2003
;
52
:
1786
1791
34.
Kapitza
C
,
Dahl
K
,
Jacobsen
JB
,
Axelsen
MB
,
Flint
A
.
The effects of semaglutide on β-cell function in subjects with type 2 diabetes (Abstract)
.
Diabetes
2016
;
65
(
Suppl. 1
):
A262
35.
Bunck
MC
,
Cornér
A
,
Eliasson
B
, et al
.
Effects of exenatide on measures of β-cell function after 3 years in metformin-treated patients with type 2 diabetes
.
Diabetes Care
2011
;
34
:
2041
2047
36.
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
:
275
286
37.
Diamant
M
,
Van Gaal
L
,
Guerci
B
, et al
.
Exenatide once weekly versus insulin glargine for type 2 diabetes (DURATION-3): 3-year results of an open-label randomised trial
.
Lancet Diabetes Endocrinol
2014
;
2
:
464
473
38.
Eldor
R
,
Daniele
G
,
Huerta
C
, et al
.
Discordance between central (brain) and pancreatic action of exenatide in lean and obese subjects
.
Diabetes Care
2016
;
39
:
1804
1810
39.
van Bloemendaal
L
,
Ten Kulve
JS
,
la Fleur
SE
,
Ijzerman
RG
,
Diamant
M
.
Effects of glucagon-like peptide 1 on appetite and body weight: focus on the CNS
.
J Endocrinol
2014
;
221
:
T1
T16
40.
Armstrong
MJ
,
Hull
D
,
Guo
K
, et al
.
Glucagon-like peptide 1 decreases lipotoxicity in non-alcoholic steatohepatitis
.
J Hepatol
2016
;
64
:
399
408
41.
Cusi
K
.
Treatment of patients with type 2 diabetes and non-alcoholic fatty liver disease: current approaches and future directions
.
Diabetologia
2016
;
59
:
1112
1120
42.
Mann
JF
,
Brown Frandsen
K
,
Daniels
G
, et al
.
Liraglutide and renal outcomes in type 2 diabetes: results of the LEADER trial (Abstract). J Am Soc Nephrol 2016;27(Abstract Edition):1B
43.
Buse
JB
,
DeFronzo
RA
,
Rosenstock
J
, et al
.
The primary glucose-lowering effect of metformin resides in the gut, not the circulation: results from short-term pharmacokinetic and 12-week dose-ranging studies
.
Diabetes Care
2016
;
39
:
198
205
44.
DeFronzo
RA
,
Buse
JB
,
Kim
T
, et al
.
Once-daily delayed-release metformin lowers plasma glucose and enhances fasting and postprandial GLP-1 and PYY: results from two randomised trials
.
Diabetologia
2016
;
59
:
1645
1654
45.
Sarwar
N
,
Gao
P
,
Seshasai
SR
, et al.;
Emerging Risk Factors Collaboration
.
Diabetes mellitus, fasting blood glucose concentration, and risk of vascular disease: a collaborative meta-analysis of 102 prospective studies
.
Lancet
2010
;
375
:
2215
2222
46.
Di Angelantonio
E
,
Kaptoge
S
,
Wormser
D
, et al.;
Emerging Risk Factors Collaboration
.
Association of cardiometabolic multimorbidity with mortality [published correction appears in JAMA 205;314:1179]
.
JAMA
2015
;
314
:
52
60
47.
Dormandy
JA
,
Charbonnel
B
,
Eckland
DJ
, et al.;
PROactive Investigators
.
Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macroVascular Events): a randomised controlled trial
.
Lancet
2005
;
366
:
1279
1289
48.
Kernan
WN
,
Viscoli
CM
,
Furie
KL
, et al.;
IRIS Trial Investigators
.
Pioglitazone after ischemic stroke or transient ischemic attack
.
N Engl J Med
2016
;
374
:
1321
1331
49.
Zinman
B
,
Wanner
C
,
Lachin
JM
, et al.;
EMPA-REG OUTCOME Investigators
.
Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes
.
N Engl J Med
2015
;
373
:
2117
2128
50.
Wu
JH
,
Foote
C
,
Blomster
J
, et al
.
Effects of sodium-glucose cotransporter-2 inhibitors on cardiovascular events, death, and major safety outcomes in adults with type 2 diabetes: a systematic review and meta-analysis
.
Lancet Diabetes Endocrinol
2016
;
4
:
411
419
51.
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
:
169
174
52.
Evans
JM
,
Ogston
SA
,
Emslie-Smith
A
,
Morris
AD
.
Risk of mortality and adverse cardiovascular outcomes in type 2 diabetes: a comparison of patients treated with sulfonylureas and metformin
.
Diabetologia
2006
;
49
:
930
936
53.
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
:
1317
1326
54.
White
WB
,
Cannon
CP
,
Heller
SR
, et al.;
EXAMINE Investigators
.
Alogliptin after acute coronary syndrome in patients with type 2 diabetes
.
N Engl J Med
2013
;
369
:
1327
1335
55.
Green
JB
,
Bethel
MA
,
Armstrong
PW
, et al.;
TECOS Study Group
.
Effect of sitagliptin on cardiovascular outcomes in type 2 Diabetes
.
N Engl J Med
2015
;
373
:
232
242
56.
Gerstein
HC
,
Bosch
J
,
Dagenais
GR
, et al.;
ORIGIN Trial Investigators
.
Basal insulin and cardiovascular and other outcomes in dysglycemia
.
N Engl J Med
2012
;
367
:
319
328
57.
Verge
D
,
López
X
.
Impact of GLP-1 and GLP-1 receptor agonists on cardiovascular risk factors in type 2 diabetes
.
Curr Diabetes Rev
2010
;
6
:
191
200
58.
Koska
J
,
Sands
M
,
Burciu
C
, et al
.
Exenatide protects against glucose- and lipid-induced endothelial dysfunction: evidence for direct vasodilation effect of GLP-1 receptor agonists in humans
.
Diabetes
2015
;
64
:
2624
2635
59.
Campbell
JE
,
Drucker
DJ
.
Pharmacology, physiology, and mechanisms of incretin hormone action
.
Cell Metab
2013
;
17
:
819
837
60.
Drucker
DJ
.
The cardiovascular biology of glucagon-like peptide-1
.
Cell Metab
2016
;
24
:
15
30
61.
DeFronzo
RA
,
Goodman
AM
;
The Multicenter Metformin Study Group
.
Efficacy of metformin in patients with non-insulin-dependent diabetes mellitus
.
N Engl J Med
1995
;
333
:
541
549
62.
Cusi
K
,
DeFronzo
RA
.
Metformin: a review of its metabolic effects
.
Diabetes Reviews
1998
;
6
:
89
131
63.
Natali
A
,
Ferrannini
E
.
Effects of metformin and thiazolidinediones on suppression of hepatic glucose production and stimulation of glucose uptake in type 2 diabetes: a systematic review
.
Diabetologia
2006
;
49
:
434
441
64.
Jensen
JB
,
Gormsen
LC
,
Sundelin
E
, et al
.
Organ-specific uptake and elimination of metformin can be determined in vivo in mice and humans by PET-imaging using a novel 11C-metformin tracer (Abstract)
.
Diabetes
2015
;
64
(
Suppl. 1
):
LB33
65.
UK Prospective Diabetes Study (UKPDS) Group
.
Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34)
.
Lancet
1998
;
352
:
854
865
66.
Hare
KJ
,
Knop
FK
,
Asmar
M
, et al
.
Preserved inhibitory potency of GLP-1 on glucagon secretion in type 2 diabetes mellitus
.
J Clin Endocrinol Metab
2009
;
94
:
4679
4687
67.
Jendle
J
,
Grunberger
G
,
Blevins
T
,
Giorgino
F
,
Hietpas
RT
,
Botros
FT
.
Efficacy and safety of dulaglutide in the treatment of type 2 diabetes: a comprehensive review of the dulaglutide clinical data focusing on the AWARD phase 3 clinical trial program
.
Diabetes Metab Res Rev
2016
;
32
:
776
790
68.
Pfeffer
MA
,
Claggett
B
,
Diaz
R
, et al.;
ELIXA Investigators
.
Lixisenatide in patients with type 2 diabetes and acute coronary syndrome
.
N Engl J Med
2015
;
373
:
2247
2257
69.
Henry
RR
,
Rosenstock
J
,
Logan
D
,
Alessi
T
,
Luskey
K
,
Baron
MA
.
Continuous subcutaneous delivery of exenatide via ITCA 650 leads to sustained glycemic control and weight loss for 48 weeks in metformin-treated subjects with type 2 diabetes
.
J Diabetes Complications
2014
;
28
:
393
398
70.
Novo Nordisk A/S. Efficacy and long-term safety of oral semaglutide versus sitagliptin in subjects with type 2 diabetes (PIONEER 3). In: ClinicalTrials.gov [Internet]. Bethesda, MD, National Library of Medicine, 2017. Available from https://clinicaltrials.gov/ct2/show/NCT2607865. NLM Identifier: NCT02607865. Accessed 18 May 2017
71.
American Diabetes Association
.
Economic costs of diabetes in the U.S. in 2012
.
Diabetes Care
2013
;
36
:
1033
1046
72.
Cusi
K
,
Consoli
A
,
DeFronzo
A
.
Metabolic effects of metformin on glucose and lactate metabolism in noninsulin-dependent diabetes mellitus
.
J Clin Endocrinol Metab
1996
;
81
:
4059
4067
73.
DeFronzo
RA
,
Barzilai
N
,
Simonson
DC
.
Mechanism of metformin action in obese and lean noninsulin-dependent diabetic subjects
.
J Clin Endocrinol Metab
1991
;
73
:
1294
1301
74.
Brown
JB
,
Conner
C
,
Nichols
GA
.
Secondary failure of metformin monotherapy in clinical practice
.
Diabetes Care
2010
;
33
:
501
506
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at http://www.diabetesjournals.org/content/license.