7
Is It Time to Change the Type 2 Diabetes Treatment Paradigm? Yes! GLP-1 RAs Should Replace Metformin in the Type 2 Diabetes Algorithm Diabetes Care 2017;40:11211127 | https://doi.org/10.2337/dc16-2368 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 rst-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 metformins 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 identication 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 benet on the disease process. In our eld, there is an evolving debate regarding the suggested rst step in diabetes management and a call for a new paradigm. Given the current con- troversy, we provide a Point-Counterpoint debate on this issue. In the point narrative below that precedes the counterpoint narrative, Drs. Abdul-Ghani and DeFronzo pro- vide their argument that a treatment approach for type 2 diabetes based upon correct- ing 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 rst-line therapy from metformin to a GLP-1 re- ceptor agonist. In the counterpoint narrative that follows Drs. Abdul-Ghani and DeFronzos contribution, Dr. Inzucchi argues that, based on the medical communitys extensive experience and the drugs demonstrated efcacy, safety, low cost, and cardiovascular benets, metformin should remain the foundation therapyfor all patients with type 2 diabetes, barring contraindications. dWilliam T. Cefalu Chief Scientic, Medical & Mission Ofcer, American Diabetes Association The modern approach to disease management is based upon identication of its etiology and correcting the underlying pathophysiological disturbances with interven- tions that ameliorate/normalize known defects responsible for the clinical manifesta- tion of the disease, i.e., hyperglycemia. Therapeutic interventions that simply target hyperglycemia but do not correct the underlying pathogenic disturbances are unlikely 1 Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX 2 Diabetes Research, Academic Health System, Hamad General Hospital, Doha, Qatar Corresponding author: Muhammad Abdul- Ghani, [email protected]. M.A.-G. and R.A.D. contributed equally to this article. © 2017 by the American Diabetes Association. Readers may use this article as long as the work is properly cited, the use is educational and not for prot, and the work is not altered. More infor- mation is available at http://www.diabetesjournals .org/content/license. See accompanying article, p. 1128. Muhammad Abdul-Ghani 1,2 and Ralph A. DeFronzo 1 Diabetes Care Volume 40, August 2017 1121 POINT-COUNTERPOINT

Is It Time to Change the Type 2 Diabetes Treatment ......care.diabetesjournals.org Abdul-Ghani and DeFronzo 1123 major core defects in T2D patients, i.e., b-cell dysfunction and muscle

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

  • Is It Time to Change the Type 2Diabetes Treatment Paradigm?Yes! GLP-1 RAs Should ReplaceMetformin in the Type 2 DiabetesAlgorithmDiabetes Care 2017;40:1121–1127 | https://doi.org/10.2337/dc16-2368

    Most treatment guidelines, including those from theAmericanDiabetes Association/European Association for the Study of Diabetes and the International DiabetesFederation, suggestmetformin be used as thefirst-line therapy after diet and exercise.This recommendation is based on the considerable body of evidence that hasaccumulatedover the last 30 years, but it is also supportedon clinical groundsbasedonmetformin’s affordability and tolerability. As such, metformin is the most commonlyused oral antihyperglycemic agent in the U.S. However, based on the release ofnewer agents over the recent past, some have suggested that the modern approachto disease management should be based upon identification of its etiology andcorrecting the underlying biological disturbances. That is, we should use interventionsthat normalize or at least ameliorate the recognized derangements in physiology thatdrive the clinical manifestation of disease, in this circumstance, hyperglycemia. Thus, itis argued that therapeutic interventions that target glycemia but do not correct theunderlying pathogenic disturbances are unlikely to result in a sustained benefit on thedisease process. In our field, there is an evolving debate regarding the suggested firststep in diabetes management and a call for a new paradigm. Given the current con-troversy, we provide a Point-Counterpoint debate on this issue. In the point narrativebelow that precedes the counterpoint narrative, Drs. Abdul-Ghani and DeFronzo pro-vide their argument that a treatment approach for type 2diabetes basedupon correct-ing the underlying pathophysiological abnormalities responsible for the developmentof hyperglycemia provides the best therapeutic strategy. Such an approach requires achange in the recommendation for first-line therapy from metformin to a GLP-1 re-ceptor agonist. In the counterpoint narrative that follows Drs. Abdul-Ghani andDeFronzo’s contribution, Dr. Inzucchi argues that, based on the medical community’sextensive experience and the drug’s demonstrated efficacy, safety, low cost, andcardiovascular benefits, metformin should remain the “foundation therapy” for allpatients with type 2 diabetes, barring contraindications.

    dWilliam T. CefaluChief Scientific, Medical & Mission Officer, American Diabetes Association

    The modern approach to disease management is based upon identification of itsetiology and correcting the underlying pathophysiological disturbances with interven-tions that ameliorate/normalize known defects responsible for the clinical manifesta-tion of the disease, i.e., hyperglycemia. Therapeutic interventions that simply targethyperglycemia but do not correct the underlying pathogenic disturbances are unlikely

    1Division of Diabetes, University of Texas HealthScience Center at San Antonio, San Antonio, TX2Diabetes Research, Academic Health System,Hamad General Hospital, Doha, Qatar

    Corresponding author: Muhammad Abdul-Ghani, [email protected].

    M.A.-G. and R.A.D. contributed equally to thisarticle.

    © 2017 by the American Diabetes Association.Readers may use this article as long as the workis properly cited, the use is educational and notfor profit, and thework is not altered.More infor-mation is available at http://www.diabetesjournals.org/content/license.

    See accompanying article, p. 1128.

    Muhammad Abdul-Ghani1,2 and

    Ralph A. DeFronzo1

    Diabetes Care Volume 40, August 2017 1121

    POINT-C

    OUNTER

    POINT

    https://doi.org/10.2337/dc16-2368http://crossmark.crossref.org/dialog/?doi=10.2337/dc16-2368&domain=pdf&date_stamp=2017-07-01mailto:[email protected]://www.diabetesjournals.org/content/licensehttp://www.diabetesjournals.org/content/license

  • to result in a sustained reduction inHbA1c. It is well established that type 2diabetes (T2D) is a complex metabolic/cardiovascular disorderwith at least eightdistinct pathophysiological disturbances,referred to as the Ominous Octet (1). Hy-perglycemia is a manifestation of theseeight pathophysiological abnormalities.Nonetheless, the current recommendedapproach in T2D management still fo-cuses on lowering the plasma glucoseconcentration rather than correcting theunderlying metabolic abnormalities thatcause the hyperglycemia (2–4). There-fore, it is not surprising that current ther-apeutic guidelines (2–4) do not result in asustained HbA1c reduction (5–8). In thisPoint-Counterpoint, we argue that it istime to apply the modern concepts ofclinical practice to diabetes managementand base therapy on pathophysiology.Thereby, GLP-1 receptor agonists (GLP-1RAs), which 1) correct six of the eightcomponents of the Ominous Octet, 2)prevent/reverse the progressive b-cellfailure and rise in HbA1c, and 3) lowercardiovascular risk in T2D independentof their glucose-lowering ability (9,10),should replace metformin as the recom-mended first-line therapy in newly diag-nosed T2D patients.

    PATHOPHYSIOLOGY OF T2D

    Theetiology of T2D is complex and involvesmultiple pathophysiological disturbancesinvolving multiple organs (1) (Fig. 1).

    Insulin resistance in skeletal muscle, liver,and adipocytes (11–15) and b-cell dys-function (16–22) remain the major coredefects responsible for the developmentand progression of hyperglycemia. Insulinresistance is also associated withmultiplemetabolic abnormalities, e.g., hyperten-sion, dyslipidemia, endothelial dysfunc-tion, procoagulant state, inflammation,and visceral obesity, which collectivelyare known as the insulin resistance (met-abolic) syndrome (23–25). Each individualcomponent of the insulin resistance syn-drome, as well as the basic molecularetiology of the insulin resistance (25), iscausally related to the development ofatherosclerotic cardiovascular disease(CVD) and contributes to the increasedrisk for CVD in T2D patients.

    Because progressive b-cell failure isthe principal factor responsible for thedevelopment and progression of hyper-glycemia in T2D patients (1,16–19), onlytherapies that halt/reverse the progres-sive b-cell failure will be effective in low-ering and maintaining HbA1c at the targetlevel, and ideally this should be ac-complished without increasing the riskof hypoglycemia.

    In addition to insulin resistance andb-cell dysfunction, impaired incretineffect in T2D plays a major role in theprogression of b-cell failure and hyper-glycemia (26,27). Further, T2D patientshave elevated fasting plasma glucagonlevels that fail to suppress normally after

    a meal and enhanced hepatic sensitivityto glucagon (28,29), in part due to resis-tance to GLP-1 (26,30); these pathophys-iological abnormalities can be reversedwith GLP-1 RA therapy (26,31).

    BIOLOGICAL ACTIONSOF GLP-1 RAs

    Activation of GLP-1 receptors in theb-cellamplifies glucose-stimulated insulin se-cretion but only under conditions of hy-perglycemia (26,32), whereas in thea-cell, GLP-1 suppresses glucagon secre-tion, leading to correction of postmealhyperglycemia in T2D (26,27,30). GLP-1RAs improve b-cell function by enhancingb-cell responsiveness to glucose, i.e., im-proving b-cell glucose sensitivity; thisbeneficial effect on the b-cell can be ob-served within 8 h after a single injectionof the GLP-1 RA (liraglutide) (33), is main-tained at 3 months (semaglutide) (34),and persists for at least 3 years (exenatide)(35). Thus, GLP-1 RAs produce a rapid anddurable reduction in HbA1c with low riskof hypoglycemia (36,37).

    GLP-1 also exerts multiple nonglyce-mic actions, all of which improve meta-bolic control in T2D patients (Table 1),including 1) delayed gastric emptying,which slows the absorption of ingestedglucose (32), 2) appetite suppression,which promotes weight loss (26,38,39),3) reduction of hepatic and visceral fatcontent (40), making them an attractiveintervention to prevent/reverse non-alcoholic fatty liver disease/nonalcoholicsteatohepatitis (41), and 4) preventionof diabetic nephropathy in Liraglutide Ef-fect and Action in Diabetes: Evaluation ofCardiovascular Outcome ResultsdA LongTerm Evaluation (LEADER) (42). Recent ev-idence suggests that gut stimulationof GLP-1 secretion by the L cells is an im-portant mechanism via which metforminsuppresses hepatic glucose production(43,44).

    GLP-1 RAs AND CVD

    CVD is the leading cause of death in T2Dpatients (45), accounting for ;80% ofmortality, and T2D is best viewed as acardiometabolic disorder (25,46). Thus,reducing CVD risk is a high priority inT2Dmanagement, and reduction in bloodpressure and correction of diabetic dysli-pidemia are essential components ofdiabetes management. Considerable evi-dence documents that hyperglycemiais a weak risk factor for cardiovascular

    Figure 1—GLP-1 RAs correct six components of the Ominous Octet, whereas metformin correctsonly one component.

    1122 Point Diabetes Care Volume 40, August 2017

  • complications and improving glucosecontrol has little benefit on macrovascu-lar disease risk (UK Prospective DiabetesStudy [UKPDS], Action to Control Cardio-vascular Risk in Diabetes [ACCORD], Ac-tion in Diabetes and Vascular Disease:Preterax and Diamicron MR ControlledEvaluation [ADVANCE], Veterans AffairsDiabetes Trial [VADT]), especially when

    it is well established. Numerous clinical tri-als have demonstrated that antidiabetesagents that reduceplasmaglucosewithoutaltering other cardiovascular risk factorsfail to reduce CVD risk in T2D patients.Conversely, antidiabetes medications thatin addition to lowering the plasma glu-cose concentration also improve car-diovascular risk factors, e.g., GLP-1 RAs(9,10), pioglitazone (47,48), and SGLT2inhibitors (49,50), significantly reducecardiovascular events in T2D with estab-lished CVD. Thus, these agents should befavored over agents that lower plasma glu-cose but have no effect on cardiovascularrisk factors or CVD, e.g., sulfonylureas(51,52), DPP-4 inhibitors (53–55), and in-sulin (56) (Fig. 2). GLP-1 RAs consistentlyhave been shown to reduce many CVDrisk factors (Table 2) (25,34,57–60).Thus, it is not surprising that two large,prospective, randomized, double-blind,placebo-controlled trials (9,10) havedem-onstrated that liraglutide and semaglu-tide significantly lower the incidence of3-pointMACE (major adverse cardiovascu-lar events), which includes nonfatal myo-cardial infarction, nonfatal stroke, andcardiovascular death, by 13% and 24%, re-spectively, in T2D patients with existingCVD. Of note, despite the high CVD riskin the patient populations in both LEADERand Trial to Evaluate Cardiovascular andOther Long-term Outcomes With Sema-glutide in Subjects With Type 2 Diabetes(SUSTAIN-6), all cardiovascular risk factors,including blood pressure and LDL choles-terol, were well controlled at baseline,

    consistent with the high number of pa-tients receiving statins, ACE inhibitors, an-giotensin receptor blockers, and aspirintherapy. Addition of the GLP-1 RA to thepatients’ antidiabetes treatment resultedin onlymodest reductions in HbA1c (0.4%)and systolic bloodpressure (;2–3mmHg)in LEADER and SUSTAIN-6; these glucose-and blood pressure–lowering effects arequite modest and unlikely to explain theCVD benefit of liraglutide and semaglu-tide. This suggests the GLP-1 RAs mayhave a direct beneficial action to slow theatherosclerotic process, independent oftheir effect to reduceglycemiaand improvetraditional cardiovascular risk factors (59).

    The above review demonstrates thatGLP-1RAsdirectly and/or indirectly correct/improve six of the eight pathophysiologicaldefects responsible for hyperglycemiain T2D, improve cardiovascular risk factors,and reduce MACE in two large, well-designed, prospective cardiovascular inter-vention trials.

    GLP-1 RAs, NOT METFORMIN,SHOULD BE THE FIRST-LINETHERAPY IN T2D

    GLP-1 RAs correct six members of theOminous Octet, whereas the only knownaction of metformin is to inhibit hepaticglucose production (61) (Fig. 1 and Table2). Contrary to common belief, metfor-min is not an insulin sensitizer in muscleor adipocytes (61–63) in the absence ofweight loss, which is a frequent occur-rence in patients treated with the bigua-nide (Fig. 3A). Consistent with this,following intravenous administration of11C-metformin, none of the biguanidecan be detected in muscle (64). Most im-portantly, and in direct contrast to theGLP-1 RAs, metformin lacks any effecton b-cell function (61,62) (Fig. 3B), whichis the primary pathophysiological distur-bance responsible for progressive hyper-glycemia in T2D patients (1). This is mostgraphically demonstrated in the UKPDS(65) and ADiabetesOutcome ProgressionTrial (ADOPT) (5) (Fig. 3D) in which, afteran initial decline during the first year,HbA1c rose progressively because of pro-gressive b-cell failure. This stands inmarked contrast to the GLP-1 RAs, whichexert a potent protective effect on theb-cell that persists for at least 3 years(34). Because the GLP-1 RAs cause signif-icant weight loss, they also improve insu-lin sensitivity in muscle. Thus, GLP-1 RAs,but not metformin, correct the two

    Table 1—Metabolic actions ofGLP-1 RAsc PancreasPotentiate glucose-mediated insulin

    secretionPreserve b-cell function/reverse b-cell

    failureInhibit glucagon secretion in

    a glucose-dependent fashion

    c Cardiovascular systemReduce MACEReduce systolic blood pressureReduce pulmonary capillary wedge

    pressureIncrease myocardial salvage following

    myocardial infarctionImprove endothelial dysfunction

    c GISlow gastric emptyingInhibit hepatic glucose productionDecrease liver fat contentDecrease visceral fat

    c Central nervous systemSuppress appetite

    c KidneyPreserve renal functionIncrease sodium excretion

    c GeneralPromote weight loss

    Figure 2—Not all antidiabetes agents are equal in their ability to reduce cardiovascular risk.

    care.diabetesjournals.org Abdul-Ghani and DeFronzo 1123

    http://care.diabetesjournals.org

  • major core defects in T2D patients, i.e.,b-cell dysfunction and muscle insulin re-sistance. The major mechanism of actionof metformin to reduce glycemia is in-hibition of hepatic gluconeogenesis(61,62) (Fig. 3C), whereas the GLP-1 RAsalso effectively reduce hepatic glucose

    production but by multiple other mecha-nisms, i.e., inhibition of glucagon secre-tion, stimulation of insulin secretion,direct effect on the liver, and depletionof liver fat (26,27,30,59,66–68).

    Although the UKPDS demonstratedthat metformin caused a reduction in

    cardiovascular events in T2D patients(65), the patient population consistedof a small number of obese T2D subjects(n 5 342); these results, by today’s stan-dards, would never be accepted as evi-dence for a cardiovascular benefit ofthe biguanide. Moreover, a beneficial

    Table 2—Benefits of GLP-1 RAs far outweigh those of metformin

    GLP-1 RAs Metformin

    Pathophysiological defects in T2D (see Fig. 1) Corrects 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

    Figure 3—Effect of metformin on glycemic control, insulin secretion, and insulin sensitivity in T2D. A: Metformin does not improve muscle insulinsensitivity (measured with euglycemic insulin clamp) in T2D individuals (n5 20) in the absence ofweight loss (72).B:Metformin has no effect onb-cell function in T2D individuals (n5 14) (measured with an oral glucose tolerance test [OGTT] and hyperglycemic clamp) (73). C: The primary effectvia which metformin reduces the HbA1c in T2D is related to the suppression of hepatic glucose production (HGP) via inhibition ofgluconeogenesis (72). FPG, fasting plasma glucose. D: Effect of metformin on HbA1c. Because metformin does not affect muscle insulin sensitivity orb-cell function, following an initial decline after metformin administration, the HbA1c rises progressively in T2D patients (5,6,74). KPNW, Kaiser Perma-nente Northwest.

    1124 Point Diabetes Care Volume 40, August 2017

  • effect on cardiovascular events was notobserved in other clinical studies with met-formin, i.e., the ADOPT study (5), whichincluded twice the number of patientsas the UKPDS (n5 818). To the contrary,subjects receiving metformin in ADOPTexperienced more cardiovascular eventsthan subjects receiving glyburide, al-though this difference was not statisti-cally significant. This emphasizes theproblemof interpreting results fromstud-ies that are markedly underpowered todetect clinically significant differences incardiac event rates. Conversely, the CVDbenefit of GLP-1 RAs has conclusivelybeen demonstrated in two very large,prospective cardiovascular interventiontrials, LEADER and SUSTAIN-6 (9,10).With regard to safety, both metformin

    andGLP-1 RAs are associatedwith gastro-intestinal (GI) adverse events. Approxi-mately 15–20% of T2D patients do nottolerate metformin because of GI sideeffects (66). The incidence of GI side ef-fects with the long-acting GLP-1 RAs issimilar to that of metformin. Further,and unlike those with metformin, the GIside effects usually are mild to moderate,waning over the first 4–6 weeks of initi-ating therapy. The percentage of patientswho discontinue long-acting GLP-1 RAsbecause of GI side effects is signifi-cantly lower than that of metformin(67). Some postmarketing reports havesuggested an increased risk of acute pan-creatitis with GLP-1 RA use. However,three large, prospective, double-blind,placebo-controlled clinical trials including;20,000 patients followed for 2–4 yearshave demonstrated no increased risk ofacute pancreatitis with GLP-1 RA use(9,10,68).Metformin is administered orally ver-

    sus via injection for GLP-1 RAs. However,intermediate-acting metformin requiresmultiple daily dosing, whereas two long-acting GLP-1 RAs (exenatide and dulaglu-tide) are available as weekly injectionsand a third (semaglutide) is under reviewby the U.S. Food and Drug Administra-tion. A subcutaneously implanted os-motic mini pump that continuouslydelivers exenatide for 6 months is ex-pected to be approved within the nextyear (69), and an oral formulation of theGLP-1 RA semaglutide is in phase 3 trials(70) and is anticipated to be availablewithin 3–4 years. These modern deliverymethodswill improve patient compliancefor GLP-1 RAs versus metformin.

    Lastly, metformin is generic and inex-pensive, whereas GLP-1 RAs are still un-der patent and, therefore, expensive.However, most large health care planshave at least one GLP-1 RA on formularywith a modest copay. Moreover, lira-glutide (Victoza) is expected to becomegeneric by the end of 2017, and thisshould significantly reduce its cost. Acost-effective analysis is beyond thescope of this discussion, and the appro-priate long-term, clinical, real-world stud-ies to perform such an analysis are notavailable. However, a recent cost analysisfor the treatment of T2D patients in theU.S. by the American Diabetes Associa-tion (71) demonstrated that only a smallportion (12%) of the cost of T2D is due tothe direct cost of antihyperglycemic med-ications. The vast majority of the cost ofdiabetes care is related to the develop-ment of diabetic vascular complica-tions, with CVD disease contributing50% of that cost, and two recent stud-ies, LEADER and SUSTAIN-6, have de-monstrated that GLP-1 RAs decreasecardiovascular events. Further, the costof medications to treat the complica-tions of diabetes is 50% greater thanthe cost of antihyperglycemic medi-cations. It remains to be determinedwhether, on a long-term basis, the useof GLP-1 RAs, which in addition to causinga durable reduction in the plasma glu-cose concentration (thereby decreas-ing microvascular complications) alsoreduce cardiovascular events, will becost-effective.

    In summary, the currently availableclinical and scientific evidence (Table 2)is overwhelmingly in favor of the useof GLP-1 RAs over metformin as first-line therapy in newly diagnosed T2D pa-tients.

    Funding. M.A.-G. receives funding from theNational Institutes of Health (DK 097554-01)and the Qatar Foundation (National PrioritiesResearch Program 4-248-3-076), and R.A.D. re-ceives funding from the National Institutes ofHealth (DK 024092-42). R.A.D.’s salary is, in part,supported by the South Texas Veterans HealthCare System, Audie L. Murphy Division.Duality of Interest. R.A.D. is on the advisoryboard of AstraZeneca, Janssen, Novo Nordisk,Boehringer Ingelheim, and Intarcia. R.A.D. has re-ceived grants from AstraZeneca, Janssen, andBoehringer Ingelheim. R.A.D. is a member ofthe speakers’ bureau for AstraZeneca and NovoNordisk. No other potential conflicts of interestrelevant to this article were reported.

    References1. DeFronzoRA. Banting Lecture. From the trium-virate to the ominous octet: a new paradigm forthe treatment of type 2 diabetes mellitus. Diabe-tes 2009;58:773–7952. Inzucchi SE, Bergenstal RM,Buse JB, et al.Man-agement of hyperglycemia in type 2 diabetes,2015: a patient-centered approach: update to aposition statement of the American Diabetes As-sociation and the European Association for theStudy of Diabetes. Diabetes Care 2015;38:140–1493. Garber AJ, Abrahamson MJ, Barzilay JI, et al.;American Association of Clinical Endocrinologists(AACE); American College of Endocrinology (ACE).Consensus statement of theAmericanAssociationof Clinical Endocrinology and American College ofEndocrinology on the comprehensive type 2diabetes algorithm–2015 executive summary.Endocr Pract 2015;21:1403–14144. International Diabetes Federation ClinicalGuidelines Task Force. Global Guideline forType 2 Diabetes. Brussels, Belgium, InternationalDiabetes Federation, 2012, p. 44–465. Kahn SE, Haffner SM, Heise MA, et al.; ADOPTStudy Group. Glycemic durability of rosiglitazone,metformin, or glyburide monotherapy. N Engl JMed 2006;355:2427–24436. United Kingdom Prospective Diabetes Study(UKPDS). 13: relative efficacy of randomly allo-cated diet, sulphonylurea, insulin, or metforminin patients with newly diagnosed non-insulin de-pendent diabetes followed for three years. BMJ1995;310:83–887. Abdul-Ghani MA, Puckett C, Triplitt C, et al.Initial combination therapy with metformin,pioglitazone and exenatide is more effectivethan sequential add-on therapy in subjects withnew-onset diabetes. Results from the Efficacy andDurability of Initial Combination Therapy forType 2 Diabetes (EDICT): a randomized trial.Diabetes Obes Metab 2015;17:268–2758. Abdul-Ghani M, Mujahid O, Mujahid A,et al. Combination therapy with exenatide pluspioglitazone versus basal/bolus insulin in patientswith poorly controlled type 2 diabetes on sulfo-nylurea plus metformin: the QATAR Study. Diabe-tes Care 2017;40:325–3319. Marso SP, Daniels GH, Brown-Frandsen K,et al.; LEADER Steering Committee; LEADER TrialInvestigators. Liraglutide and cardiovascular out-comes in type 2 diabetes. N Engl J Med 2016;375:311–32210. Marso SP,Bain SC,Consoli A, et al.; SUSTAIN-6Investigators. Semaglutide and cardiovascu-lar outcomes in patients with type 2 diabetes.N Engl J Med 2016;375:1834–184411. Abdul-Ghani MA, DeFronzo RA. Pathogenesisof insulin resistance in skeletal muscle. J BiomedBiotechnol 2010;2010:47627912. Groop LC, Widén E, Ferrannini E. Insulin re-sistance and insulin deficiency in the pathogenesisof type 2 (non-insulin-dependent) diabetes melli-tus: errors of metabolism or of methods? Diabe-tologia 1993;36:1326–133113. Shalata A, Jazmawi W, Aslan O, et al. Earlymetabolic defects in Arab subjects with strongfamily history of type 2 diabetes. J Endocrinol In-vest 2013;36:417–42114. Reaven GM. Relationships among insulin re-sistance, type 2 diabetes, essential hypertension,and cardiovascular disease: similarities and

    care.diabetesjournals.org Abdul-Ghani and DeFronzo 1125

    http://care.diabetesjournals.org

  • differences. J Clin Hypertens (Greenwich) 2011;13:238–24315. Kashyap S, Belfort R, Gastaldelli A, et al. Asustained increase in plasma free fatty acids im-pairs insulin secretion in nondiabetic subjects ge-netically predisposed to develop type 2 diabetes.Diabetes 2003;52:2461–247416. Kahn SE, Cooper ME, Del Prato S. Pathophys-iology and treatment of type 2 diabetes: perspec-tives on the past, present, and future. Lancet2014;383:1068–108317. Alejandro EU, Gregg B, Blandino-Rosano M,Cras-Méneur C, Bernal-Mizrachi E. Natural historyofb-cell adaptation and failure in type 2 diabetes.Mol Aspects Med 2015;42:19–4118. Halban PA, Polonsky KS, Bowden DW, et al.b-Cell failure in type 2 diabetes: postulatedmech-anisms and prospects for prevention and treat-ment. Diabetes Care 2014;37:1751–175819. Saad MF, Knowler WC, Pettitt DJ, Nelson RG,Mott DM, Bennett PH. The natural history of im-paired glucose tolerance in the Pima Indians.N Engl J Med 1988;319:1500–150620. Ferrannini E, Gastaldelli A, Miyazaki Y,MatsudaM,Mari A, DeFronzo RA. b-Cell functionin subjects spanning the range from normal glu-cose tolerance to overt diabetes: a new analysis.J Clin Endocrinol Metab 2005;90:493–50021. Ferrannini E, Mari A. b-Cell function in type 2diabetes. Metabolism 2014;63:1217–122722. Polonsky KS. Lilly Lecture 1994. The b-cell indiabetes: from molecular genetics to clinical re-search. Diabetes 1995;44:705–71723. Jornayvaz FR, Samuel VT, Shulman GI. Therole of muscle insulin resistance in the pathogen-esis of atherogenic dyslipidemia and nonalcoholicfatty liver disease associated with the metabolicsyndrome. Annu Rev Nutr 2010;30:273–29024. Reaven GM. Insulin resistance: the link be-tween obesity and cardiovascular disease. MedClin North Am 2011;95:875–89225. DeFronzo RA. Insulin resistance, lipotoxicity,type 2 diabetes and atherosclerosis: the missinglinks. The Claude Bernard Lecture 2009. Diabeto-logia 2010;53:1270–128726. Nauck MA, Meier JJ. The incretin effect inhealthy individuals and those with type 2 diabe-tes: physiology, pathophysiology, and response totherapeutic interventions. Lancet Diabetes Endo-crinol 2016;4:525–53627. Holst JJ, Gribble F, Horowitz M, Rayner CK.Roles of the gut in glucose homeostasis. DiabetesCare 2016;39:884–89228. Matsuda M, DeFronzo RA, Glass L, et al. Glu-cagon dose-response curve for hepatic glucoseproduction and glucose disposal in type 2 diabeticpatients and normal individuals. Metabolism2002;51:1111–111929. Knop FK, Vilsbøll T, Madsbad S, Holst JJ,Krarup T. Inappropriate suppression of glucagonduring OGTT but not during isoglycaemic i.v. glu-cose infusion contributes to the reduced incretineffect in type 2 diabetes mellitus. Diabetologia2007;50:797–80530. Sandoval DA, D’Alessio DA. Physiology ofproglucagon peptides: role of glucagon andGLP-1 in health and disease. Physiol Rev 2015;95:513–54831. Gamble JM, ClarkeA,Myers KJ, et al. Incretin-based medications for type 2 diabetes: an over-view of reviews. Diabetes Obes Metab 2015;17:649–658

    32. UmapathysivamMM, LeeMY, Jones KL, et al.Comparative effects of prolonged and intermittentstimulation of the glucagon-like peptide 1 receptoron gastric emptying and glycemia. Diabetes 2014;63:785–79033. Chang AM, Jakobsen G, Sturis J, et al. TheGLP-1 derivative NN2211 restores b-cell sensitiv-ity to glucose in type 2 diabetic patients after asingle dose. Diabetes 2003;52:1786–179134. Kapitza C, Dahl K, Jacobsen JB, Axelsen MB,Flint A. The effects of semaglutide on b-cell func-tion in subjects with type 2 diabetes (Abstract).Diabetes 2016;65(Suppl. 1):A26235. Bunck MC, Cornér A, Eliasson B, et al. Effectsof exenatide on measures of b-cell function after3 years in metformin-treated patients with type2 diabetes. Diabetes Care 2011;34:2041–204736. Klonoff DC, Buse JB, Nielsen LL, et al. Exena-tide effects on diabetes, obesity, cardiovascularrisk factors and hepatic biomarkers in patientswith type 2 diabetes treated for at least 3 years.Curr Med Res Opin 2008;24:275–28637. DiamantM, Van Gaal L, Guerci B, et al. Exena-tide onceweekly versus insulin glargine for type 2diabetes (DURATION-3): 3-year results of anopen-label randomised trial. Lancet DiabetesEndocrinol 2014;2:464–47338. Eldor R, Daniele G, Huerta C, et al. Discor-dance between central (brain) and pancreatic ac-tion of exenatide in lean and obese subjects.Diabetes Care 2016;39:1804–181039. van Bloemendaal L, Ten Kulve JS, la Fleur SE,Ijzerman RG, Diamant M. Effects of glucagon-likepeptide 1 on appetite and body weight: focus onthe CNS. J Endocrinol 2014;221:T1–T1640. ArmstrongMJ, Hull D, Guo K, et al. Glucagon-like peptide 1 decreases lipotoxicity in non-alcoholicsteatohepatitis. J Hepatol 2016;64:399–40841. Cusi K. Treatment of patients with type 2 di-abetes and non-alcoholic fatty liver disease: currentapproaches and future directions. Diabetologia2016;59:1112–112042. 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 SocNephrol 2016;27(Abstract Edition):1B43. Buse JB, DeFronzo RA, Rosenstock J, et al. Theprimary glucose-lowering effect of metformin re-sides in the gut, not the circulation: results fromshort-term pharmacokinetic and 12-week dose-ranging studies. Diabetes Care 2016;39:198–20544. DeFronzo RA, Buse JB, Kim T, et al. Once-dailydelayed-release metformin lowers plasma glu-cose and enhances fasting and postprandialGLP-1 and PYY: results from two randomised tri-als. Diabetologia 2016;59:1645–165445. Sarwar N, Gao P, Seshasai SR, et al.; EmergingRisk Factors Collaboration. Diabetesmellitus, fast-ing blood glucose concentration, and risk of vas-cular disease: a collaborative meta-analysis of102 prospective studies. Lancet 2010;375:2215–222246. Di Angelantonio E, Kaptoge S, Wormser D,et al.; Emerging Risk Factors Collaboration. Asso-ciation of cardiometabolic multimorbidity withmortality [published correction appears in JAMA205;314:1179]. JAMA 2015;314:52–6047. Dormandy JA, Charbonnel B, Eckland DJ,et al.; PROactive Investigators. Secondary preven-tion of macrovascular events in patients with type2 diabetes in the PROactive Study (PROspectivepioglitAzone Clinical Trial InmacroVascular Events):

    a randomised controlled trial. Lancet 2005;366:1279–128948. Kernan WN, Viscoli CM, Furie KL, et al.; IRISTrial Investigators. Pioglitazone after ischemicstroke or transient ischemic attack. N Engl J Med2016;374:1321–133149. Zinman B,Wanner C, Lachin JM, et al.; EMPA-REG OUTCOME Investigators. Empagliflozin, car-diovascular outcomes, and mortality in type 2diabetes. N Engl J Med 2015;373:2117–212850. Wu JH, Foote C, Blomster J, et al. Effects ofsodium-glucose cotransporter-2 inhibitors on car-diovascular events, death, and major safety out-comes in adultswith type 2 diabetes: a systematicreview and meta-analysis. Lancet Diabetes Endo-crinol 2016;4:411–41951. Simpson SH,Majumdar SR, Tsuyuki RT, EurichDT, Johnson JA. Dose-response relation betweensulfonylurea drugs and mortality in type 2 diabe-tes mellitus: a population-based cohort study.CMAJ 2006;174:169–17452. Evans JM, Ogston SA, Emslie-Smith A, MorrisAD. Risk of mortality and adverse cardiovascularoutcomes in type 2 diabetes: a comparison ofpatients treated with sulfonylureas and metfor-min. Diabetologia 2006;49:930–93653. Scirica BM, Bhatt DL, Braunwald E, et al.;SAVOR-TIMI 53 Steering Committee and Investi-gators. Saxagliptin and cardiovascular outcomesin patients with type 2 diabetes mellitus. N Engl JMed 2013;369:1317–132654. White WB, Cannon CP, Heller SR, et al.;EXAMINE Investigators. Alogliptin after acute cor-onary syndrome in patients with type 2 diabetes.N Engl J Med 2013;369:1327–133555. Green JB, Bethel MA, Armstrong PW, et al.;TECOS Study Group. Effect of sitagliptin on cardio-vascular outcomes in type 2 Diabetes. N Engl JMed 2015;373:232–24256. Gerstein HC, Bosch J, Dagenais GR, et al.;ORIGIN Trial Investigators. Basal insulin and car-diovascular and other outcomes in dysglycemia.N Engl J Med 2012;367:319–32857. Verge D, López X. Impact of GLP-1 and GLP-1receptor agonists on cardiovascular risk factors intype2diabetes. CurrDiabetesRev2010;6:191–20058. Koska J, Sands M, Burciu C, et al. Exenatideprotects against glucose- and lipid-induced endo-thelial dysfunction: evidence for direct vasodila-tion effect of GLP-1 receptor agonists in humans.Diabetes 2015;64:2624–263559. Campbell JE, Drucker DJ. Pharmacology,physiology, and mechanisms of incretin hormoneaction. Cell Metab 2013;17:819–83760. Drucker DJ. The cardiovascular biology ofglucagon-like peptide-1. Cell Metab 2016;24:15–3061. DeFronzo RA, GoodmanAM; TheMulticenterMetformin Study Group. Efficacy of metformin inpatients with non-insulin-dependent diabetesmellitus. N Engl J Med 1995;333:541–54962. Cusi K,DeFronzoRA.Metformin: areviewof itsmetaboliceffects. Diabetes Reviews 1998;6:89–13163. Natali A, Ferrannini E. Effects of metforminand thiazolidinediones on suppression of hepaticglucose production and stimulation of glucose up-take in type 2 diabetes: a systematic review. Dia-betologia 2006;49:434–44164. Jensen JB, Gormsen LC, Sundelin E, et al.Organ-specific uptake and elimination of metfor-min can be determined in vivo inmice and humansby PET-imaging using a novel 11C-metformin tracer(Abstract). Diabetes 2015;64(Suppl. 1):LB33

    1126 Point Diabetes Care Volume 40, August 2017

  • 65. UK Prospective Diabetes Study (UKPDS)Group. Effect of intensive blood-glucose controlwith metformin on complications in overweightpatients with type 2 diabetes (UKPDS 34). Lancet1998;352:854–86566. Hare KJ, Knop FK, Asmar M, et al. Preservedinhibitory potency of GLP-1 on glucagon secretionin type 2 diabetes mellitus. J Clin EndocrinolMetab 2009;94:4679–468767. Jendle J, Grunberger G, Blevins T, Giorgino F,Hietpas RT, Botros FT. Efficacy and safety of dula-glutide in the treatment of type 2 diabetes: acomprehensive review of the dulaglutide clinicaldata focusing on the AWARD phase 3 clinical trialprogram. Diabetes Metab Res Rev 2016;32:776–790

    68. Pfeffer MA, Claggett B, Diaz R, et al.; ELIXAInvestigators. Lixisenatide in patients with type 2diabetes and acute coronary syndrome. N Engl JMed 2015;373:2247–225769. Henry RR, Rosenstock J, Logan D, Alessi T,Luskey K, BaronMA. Continuous subcutaneous de-livery of exenatide via ITCA 650 leads to sustainedglycemic control and weight loss for 48 weeks inmetformin-treated subjects with type 2 diabetes.J Diabetes Complications 2014;28:393–39870. Novo Nordisk A/S. Efficacy and long-termsafety of oral semaglutide versus sitagliptin in sub-jects with type 2 diabetes (PIONEER 3). In:ClinicalTrials.gov [Internet]. Bethesda, MD, Na-tional Library of Medicine, 2017. Available fromhttps://clinicaltrials.gov/ct2/show/NCT2607865.

    NLM Identifier: NCT02607865. Accessed 18 May201771. American Diabetes Association. Economiccosts of diabetes in the U.S. in 2012. DiabetesCare 2013;36:1033–104672. Cusi K, Consoli A, DeFronzo A. Metaboliceffects ofmetformin on glucose and lactatemetab-olism in noninsulin-dependent diabetes mellitus.J Clin Endocrinol Metab 1996;81:4059–406773. DeFronzo RA, Barzilai N, Simonson DC.Mech-anismofmetformin action in obese and lean non-insulin-dependent diabetic subjects. J ClinEndocrinol Metab 1991;73:1294–130174. Brown JB, Conner C, Nichols GA. Secondaryfailure of metformin monotherapy in clinicalpractice. Diabetes Care 2010;33:501–506

    care.diabetesjournals.org Abdul-Ghani and DeFronzo 1127

    http://ClinicalTrials.govhttps://clinicaltrials.gov/ct2/show/NCT2607865http://care.diabetesjournals.org