Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic...

download Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Making (Printer-friendly)

of 25

Transcript of Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic...

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    1/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    www.medscape.or

    This article is a CME certified activity. To earn credit for this activity visit:

    http://www.medscape.org/viewarticle/775099

    CME Information

    CME Released: 12/14/2012; Valid for credit through 12/14/2013

    Target Audience

    This activity is intended for osteopathic physicians.

    Goal

    According to the American Association of Clinical Endocrinologists (AACE), more than 60% of patients with diabetes do not

    reach HbA1c targets. Advances in the understanding of glucose homeostasis have helped to identify new targeted

    treatments, such as incretin-based agents, which broaden the antidiabetic armamentarium and may address some of the

    shortcomings of conventional treatments for type 2 diabetes mellitus (T2DM). Incretin-based agents are associated with a

    low risk of unwanted adverse events and offer physicians additional options to deliver individualized care.

    Consensus guidelines for diabetes management are updated regularly and provide an excellent starting point for clinical

    decision-making. However, the rapid pace of research into new diabetic agents can cause confusion among physicians who

    would benefit from education updating them on the latest findings and providing practical, evidence-based ways to use this

    information.

    This program is designed to bridge the gap between clinicians' current understanding of the role of guidelines, HbA1c targets

    and incretin therapies in T2DM management and to support the confident application of that knowledge in clinical practice to

    help more patients safely reach target goals.

    Learning Objectives

    This activity was designed to address the following IOM competencies: provide patient-centered care, and employ evidence-

    based practice.

    At the conclusion of this activity, participants should be able to demonstrate the ability to:

    1. Recognize the pros and cons of using A1c to diagnose prediabetes and discuss prevention of diabetes by treating

    prediabetes with lifestyle management and GLP-1 agonists

    2. Summarize results from clinical trials demonstrating the long-term benefit of glycemic control in patients with T2DM,

    and compare these results to those of the tight glucose control trials

    3. Describe the mechanisms of action of incretin-based agents and determine their place in the current armamentarium

    to facilitate early attainment and ongoing maintenance of appropriate HbA1c targets in patients with T2DM

    4. Identify treatments and treatment combinations that can enhance glycemic control, facilitate weight loss, and prevent

    or slow the pathophysiology of diabetes, including reduction of insulin resistance, improvement of glucose tolerance

    and beta cell function, and reduction of cardiovascular (CV) risk factors5. Employ new strategies and medication treatment formulations to optimize antidiabetic treatment adherence

    Credits Available

    Osteopathic Physicians - maximum of 1.00 Category 1-B AOA Credit

    All other healthcare professionals completing continuing education credit for this activity will be issued a certificate of

    participation.

    Physicians should claim only the credit commensurate with the extent of their participation in the activity.

    http://www.medscape.org/viewarticle/775099http://www.medscape.org/http://www.medscape.org/viewarticle/775099http://www.medscape.org/
  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    2/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    2ww.medscape.org/viewarticle/775099_print

    Accreditation Statements

    For Physicians

    The AOA's continuing medical education program (CME) strives for growth of knowledge, refinement of skills, and the

    deepening of understanding for the osteopathic profession. The AOA Board of Trustees establishes accreditation policy for

    osteopathic CME sponsors. The Council on Continuing Medical Education (CCME) has been delegated authority by the

    AOA Board of Trustees to monitor osteopathic CME and to grant or deny Category 1 accreditation status to osteopathic

    CME sponsors.

    The American Osteopathic Association's Council on Continuing Medical Education approved the activity for 1 hour of

    Category 1-B AOA credit.

    Contact This Provider

    For questions regarding the content of this activity, contact the accredited provider for this CME/CE activity noted above. For

    technical assistance, contact [email protected]

    Instructions for Participation and Credit

    There are no fees for participating in or receiving credit for this online educational activity. For information on applicability and

    acceptance of continuing education credit for this activity, please consult your professional licensing board.

    This activity is designed to be completed within the time designated on the title page; physicians should claim only those

    credits that reflect the time actually spent in the activity. To successfully earn credit, participants must complete the activity

    online during the valid credit period that is noted on the title page. To receiveAMA PRA Category 1 Credit, you must

    receive a minimum score of 70% on the post-test.

    Follow these steps to earn CME/CE credit*:

    1. Read the target audience, learning objectives, and author disclosures.

    2. Study the educational content online or printed out.

    3. Online, choose the best answer to each test question. To receive a certificate, you must receive a passing score as

    designated at the top of the test. In addition, you must complete the Activity Evaluation to provide feedback for future

    programming.

    You may now view or print the certificate from your CME/CE Tracker. You may print the certificate but you cannot alter it.

    Credits will be tallied in your CME/CE Tracker and archived for 6 years; at any point within this time period you can print out

    the tally as well as the certificates from the CME/CE Tracker.

    *The credit that you receive is based on your user profile.

    Hardware/Software Requirements

    To access activities, users will need:

    A computer with an Internet connection.

    Internet Explorer 7.x or higher, Firefox 4.x or higher, Safari 2.x or higher, or any other W3C standards compliant

    browser.

    Adobe Flash Playerand/or an HTML5 capable browser may be required for video or audio playback.

    Occasionally other additional software may be required such as PowerPoint orAdobe Acrobat Reader.

    http://get.adobe.com/reader/http://office.microsoft.com/en-us/powerpoint/http://get.adobe.com/flashplayer/mailto:[email protected]:[email protected]
  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    3/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    3ww.medscape.org/viewarticle/775099_print

    Off-label Use

    This educational activity may contain discussion of published and/or investigational uses of agents that are not indicated by

    the FDA.

    The American Osteopathic Association and Rockpointe Corporation do not recommend the use of any agent outside of the

    labeled indications.

    Disclosure

    The American Osteopathic Association and Rockpointe Corporation adhere to the policies and guidelines, including the

    Standards for Commercial Support, set forth to providers by the Accreditation Council for Continuing Medical Education

    (ACCME) and all other professional organizations, as applicable, stating those activities where continuing education credits

    are awarded must be balanced, independent, objective, and scientifically rigorous. All persons in a position to control the

    content of a continuing medical education program are required to disclose any relevant financial relationships with any

    commercial interest to AOA or Rockpointe as well as to learners. All conflicts are identified and resolved by AOA and

    Rockpointe in accordance with the Standards for Commercial Support in advance of delivery of the activity to learners. The

    content of this activity was vetted by an external medical reviewer to ensure objectivity and that the activity is free of

    commercial bias.

    The faculty reported the following relevant financial relationships that they or their spouse/partner have with commercial

    interests:

    Authors

    C. W. Spellman, DO, PhD, Chair

    Professor and Associate Dean of Research, Director, Center for Diabetes and Metabolic Disorders, Department of Internal

    Medicine, Division of Endocrinology, Texas Tech University Health Sciences Center at Permian Basin, Odessa, Texas

    Dr. Spellman reports that he has nothing to disclose.

    James LaSalle, DO, FAAFP

    Director, Medical Arts Centers, Excelsior Springs, Missouri

    Disclosure: Consultant: Novo Nordisk; Speakers' Bureau: Boehringer Ingelheim-Lilly, GlaxoSmithKline, Novo Nordisk

    Michael D. Shapiro, DO, FACC, FSCCT

    Diplomate, American Board of Clinical Lipidology; Assistant Professor of Medicine and Radiology and Director, Preventive

    Cardiology and Atherosclerosis Imaging, Division of Cardiovascular Medicine, Oregon Health & Science University, Portland,Oregon

    Disclosure: Consultant: LipoScience; Speaker's Bureau: Abbott, LipoScience, Merck

    Non-faculty content contributors and/or reviewers reported the following relevant financial relationships that they or their

    spouse/partner have with commercial interests:

    Carole Drexel, PhD; Bradley Pine; Blair St. Amand; Jay Katz; Dana Simpler, MD; and Paula Larson report that they

    have nothing to disclose.

    Evolving Treatment for Patients with Type 2 Diabetes: Current

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    4/25

    3/13

    4ww.medscape.org/viewarticle/775099_print

    C.W. Spellman, DO, PhD, Chair; James R. LaSalle, DO, FAAFP; Michael D. Shapiro, DO, FACC, FSCCT

    How would you rate your level of knowledge regarding incretin therapies?

    Not knowledgeable

    Somewhat knowledgeable

    Knowledgeable

    Very knowledgeable

    Expert

    How competent do you feel in managing type 2 diabetes mellitus (T2DM) patients with incretin therapies?

    Not competent

    Somewhat competent

    Competent

    Very competent

    Expert

    Approximately what proportion of people with prediabetes will progress to T2DM?

    50%

    30%

    70%

    90%

    Donna is a 59-year-old woman who has had a diagnosis of diabetes for 4 years. At her last office visit, her

    body mass index (BMI) was 26.4 kg/m2 and her HbA1c was 7.9%. She has a strong family history of

    cardiovascular disease (CVD), and she is currently being treated for hypertension and dyslipidemia.

    According to American Diabetes Association (ADA) recommendations, what is Donnas HbA1c target?

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    5/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    5ww.medscape.org/viewarticle/775099_print

    True or False: In the ACCORD, ADVANCE, and VADT trials, intensive glycemic control that exceeds an A1c

    goal of < 7.0% yielded no significant reduction in CVD outcomes compared to standard glycemic control.

    True

    False

    Incretin-based therapies significantly reduce HbA1c levels in which of the following scenarios?

    Add-on to metformin

    Add-on to sulfonylurea (SU)

    Add-on to metformin and SU

    All of the above

    Which of the following antidiabetes therapies is/are associated with a reduction in body weight?

    Metformin

    Insulin

    GLP-1 agonists

    DPP-4 inhibitors

    What are the most common adverse events associated with GLP-1 receptor agonists?

    Nausea and vomiting

    Hypoglycemia

    Weight gain

    Pancreatitis

    Which is true regarding incretin therapy?

    Low risk of hypoglycemia, except when combined with SUs

    Cannot be used with renal diseaseLow risk of gastrointestinal adverse events

    All of the above

    Which of the following may help improve patient adherence to antidiabetic therapy?

    Reduced administration frequency

    Lowered risk of hypoglycemia

    Neutral or beneficial effects on body weight

    All of the above

    Save and Proceed

    This CME activity is based on the slides and lectures presented by the faculty at the American Osteopathic Association

    (AOA)accredited breakfast symposium, Evolving Treatment for Patients with Type 2 Diabetes: Current Guidelines and

    Emerging Therapeutic Decision-Making, on October 9, 2012, at the Marriott Marquis & Marina, San Diego, California.

    C. W. Spellman, DO, PhD

    Diabetes is a growing epidemic with major public health implications for patients, providers, and the healthcare system. More

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    6/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    6ww.medscape.org/viewarticle/775099_print

    than 23 million patients have been diagnosed with type 2 diabetes mellitus (T2DM) in the United States, and another 11

    million have undiagnosed disease, for a total prevalence of approximately 32 million. Furthermore, an estimated 50 million

    individuals have prediabetes, meaning they have blood glucose levels outside the normal range, but have not yet met the

    formal criteria for a diabetes diagnosis. Overall, more than 80 million Americans have T2DM, undiagnosed diabetes, or

    prediabetes.[1]

    Without effective intervention, the burden of T2DM is expected to continue to increase globally. [2] One strategy for

    addressing the diabetes epidemic is to target patients with prediabetes using lifestyle modifications and pharmacotherapy to

    re-establish normal glucose metabolism (ie, normoglycemia), reduce the incidence of progression to T2DM, and prevent the

    onset of diabetes-related complications.

    What Is Prediabetes?

    The formal definition of "prediabetes" has evolved to reflect advances in the understanding of diabetes progression. Until

    2003, the American Diabetes Association (ADA) used the term "glucose intolerance," to describe a state of intermediate

    hyperglycemia marked by impaired glucose tolerance (IGT), impaired fasting glucose (IFG), or both. [1] In 2005, the diabetes

    community adopted the term "prediabetes" to describe intermediate hyperglycemia, [3] and in 2010, the ADA expanded the

    definition of prediabetes to include intermediate HbA1c values.[4]

    In current clinical practice, the American Association of Clinical Endocrinologists (AACE), the ADA, and other major medica

    societies around the world use different criteria to define prediabetes (Table 1).[4,5]

    Table 1. Criteria for Prediabetes[4,5]

    2hPG = 2-hour postload glucose; AACE = American Association of Clinical Endocrinologists; ADA = American

    Diabetes Association; FPG = fasting plasma glucose; HbA1c = glycated hemoglobin; IFG = impaired fasting glucose;

    IGT = impaired glucose tolerance

    Significance and Diagnosis of Prediabetes

    Prediabetes now affects approximately 35% of adults in the United States. [6] Up to 70% of people with prediabetes will

    progress to develop T2DM and cardiovascular comorbidities.[7] Despite the magnitude of this public health crisis, basic

    questions related to the diagnosis of prediabetes remain unanswered. [2] For instance, HbA1c levels reflect blood glucose

    averages over the previous 812 weeks, and, as a result, may more accurately represent overall glucose control than

    measures such as IFG and IGT. Therefore, is HbA1c the preferred way to diagnose prediabetes?

    To address this question, Hollander and Spellman recently performed a systematic review of clinical trials, meta-analyses,

    and clinical practice guidelines to evaluate the utility of various criteria for the diagnosis of prediabetes and progression to

    T2DM.[1] The review incorporated studies published between 2006 and 2012 using the terms "prediabetes," "HbA1c," or

    "glucose tolerance test." Findings from the analysis illustrate current variations in the definition of prediabetes, as well of the

    implications of these variations on the diagnosis and management of patients who are at risk for T2DM and diabetes-related

    complications.

    In 1 study, investigators evaluated the strength of various diagnostic criteria for prediabetes in a cohort of 2092 Japanese

    patients.[8] At the time of study enrollment, all patients met the ADA prediabetes diagnostic criteria of IFG (fasting plasma

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    7/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    7ww.medscape.org/viewarticle/775099_print

    glucose [FPG] level of 100125 mg/dL), HbA1c level of 5.7% to 6.4%, or both. Within this group, 20% of patients met the

    HbA1c criterion alone, while another 20% met both the IFG and HbA1c criteria for prediabetes. By comparison, 60% of

    patients met only the IFG criterion, suggesting that the majority of patients would be missed if clinicians relied only on

    HbA1c measurements to diagnose prediabetes.

    Other studies have examined the role of IFG alone, IGT alone, or both values in identifying prediabetes in various patient

    groups around the world. In a cohort of 1546 US adults, the overall prevalence of prediabetes was 34%, as diagnosed on the

    basis of IFG alone (19%), IGT alone (5%), or both IFG and IGT (10%). [9] In a French study of 1283 overweight or obese

    individuals, the overall prevalence of diabetes was 20%.[10] In this group, 15% were prediabetic on the basis of IFG alone,

    while 2% met the criteria for prediabetes according to IGT alone, and 3% on the basis of both IFG and IGT. These findings

    suggest that measuring IFG alone, rather than IFG and IGT, would have missed 70% of prediabetic cases.

    Another French study examined the diagnostic utility of HbA1c in a high-risk population of 1157 patients who fulfilled the

    ADA criteria for diabetes screening, including obesity, sedentary lifestyle, first-degree relatives with diabetes, gestational

    diabetes, hypertension, and hyperlipidemia.[11] Using the current ADA threshold for HbA1c to define prediabetes (HbA1c:

    5.7%6.4%), 41% of patients met the criteria for prediabetes. By comparison, only 27% of these patients met the criteria for

    prediabetes on the basis of oral glucose tolerance testing (OGTT), which includes both IFG and IGT.

    In a Spanish study, investigators evaluated the diagnostic value of HbA1c and OGTT in a group of 1144 patients who were

    determined to have an elevated risk for diabetes according to an initial screening with the Finnish Diabetes Risk Score

    questionnaire.

    [12,13]

    In this high-risk group, 45% of patients met the criteria for prediabetes. Of these, 34% were diagnosedon the basis of HbA1c values, while 66% were diagnosed using FPG level, 2 hour postload glucose (2hPG) level, or both.

    These findings do not support the use of HbA1c testing alone to identify prediabetes in a high-risk population, and instead

    substantiate a strategy that combines HbA1c testing with traditional OGTT in the diagnosis of prediabetes.

    The DEAL (Diet Exercise Activity Lifestyle) study also evaluated the role of HbA1c testing in 242 patients with a provisional

    diagnosis of IFG based on a standard FPG test. [14] Using the formal OGTT, 56% of patients were classified with prediabetes

    on the basis of IFG alone, and 37% of patients met the criteria for prediabetes based on both IFG and IGT values. However,

    only 63% of patients diagnosed with prediabetes on the basis of IFG alone had HbA1c values that fell within the range of

    5.7%6.4%, while 70% of patients diagnosed on the basis of both IFG and IGT also met the HbA1c diagnostic criteria. This

    suggests that HbA1c testing would miss approximately one-third of individuals who were diagnosed with prediabetes on the

    basis of IFG and IGT.

    Prevention of T2DM and Return to Normoglycemia

    Intensive lifestyle modifications and pharmacotherapy are effective tools for preventing progression to T2DM in patients who

    meet the criteria for prediabetes. Although many studies support the rationale for intervening in prediabetes, most clinical

    trial data gathered to date is limited by short follow-up periods (Table 2). Although patients with prediabetes can progress to

    T2DM relatively quicklyin the range of 3 to 5 yearsstudies that fall short of this timeline may not measure the full benefits

    of lifestyle changes or pharmacotherapy.[15]

    Table 2. Lifestyle Interventions and Pharmacotherapy in the Prevention or Delay of T2DM [15]

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    8/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    8ww.medscape.org/viewarticle/775099_print

    IFG = impaired fasting glucose; IGT = impaired glucose tolerance

    For patients with prediabetes, interventions such as intensive lifestyle modifications, metformin, and incretin-based therapy

    slow the progression to T2DM and promote the return to normal blood glucose levels. In 1 study, 77% of patients treated with

    exenatide in addition to lifestyle changes regressed to euglycemia, compared with 56% of patients managed with lifestyle

    modifications alone.[18] In another trial, treatment with liraglutide resulted in an 84% to 96% decrease in the prevalence of

    diabetes, depending on liraglutide dose.[15]

    In 2012, Perreault and colleagues reported long-term findings from an extension of DPPOS (Diabetes Prevention Program

    Outcomes Study), which randomly assigned 1990 patients who were at risk for diabetes to treatment with intensive lifestyle

    therapy, metformin, or placebo.[19] Patients who became normoglycemic at least once were 56% less likely to develop

    diabetes during 5.7 years of follow-up observation than patients who were persistently classified as having prediabetes,

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    9/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    9ww.medscape.org/viewarticle/775099_print

    regardless of the method of achieving normal blood glucose (ie, lifestyle changes or metformin). These findings underscore

    the importance of timely intervention with effective therapies that allow patients with prediabetes to achieve glucose control.

    Summary

    Nearly one-third of adults in the US have prediabetes, and 70% of patients with prediabetes will develop T2DM. Although

    early identification of prediabetes is necessary to initiate effective intervention, only 7% of patients with prediabetes are

    aware of their diagnosis. Better tools for screening and diagnosis are urgently needed to enhance the care of patients at risk

    for diabetes.

    No single test is 100% effective in the diagnosis of prediabetes, and using all options for screening (ie, FPG, IFG, and

    HbA1c) may not be feasible in routine clinical practice. HbA1c testing is emerging as a key tool for identifying patients with

    prediabetes, but current testing methods present challenges for health care providers. The sensitivity and specificity of

    HbA1c as a diagnostic test for prediabetes depend on the characteristics of the patient population, and may vary by body

    mass index (BMI), age, sex, and race. The ADA, AACE, and other societies in the diabetes community recommend HbA1c

    testing to identify patients with prediabetes, but not all societies use the same HbA1c cut-points to define prediabetes.

    Recent data suggest that HbA1c may have greater utility as a diagnostic test for prediabetes in overweight and obese

    patients, as well as those with multiple other cardiovascular risk factors.

    Although there is a continuum from prediabetes to diabetes, it is important to remember that patients can move in 2

    directions: progression toward T2DM and diabetes-related complications, or regression to normoglycemia. Preliminaryclinical trial evidence suggests that incretin-targeted therapies such as exenatide and liraglutide enable more patients with

    prediabetes to regress to normoglycemia than other medications or intensive lifestyle interventions. Future clinical trial data

    will clarify the best methods for diagnosing prediabetes, as well as the optimal interventions for preventing the progression to

    T2DM.

    James LaSalle, DO, FAAFP

    The incretin hormones regulate glucose homeostasis and play key roles in the pathophysiology and treatment of

    T2DM.[20,21] The "incretin effect" was first observed in studies showing that the pancreas secreted more insulin when

    glucose was administered orally through the gastrointestinal tract than when glucose bypassed the gut and was delivered by

    intravenous (IV) infusion. Nutrient intake stimulates the release of incretins such as glucagon-like peptide-1 (GLP-1), which

    act on the pancreas to potentiate insulin secretion from the beta cells and lower glucagon secretion from the islet cells in a

    glucose-dependent manner.[20,21]

    The glucose-dependent effects of incretins are well characterized. One trial examined changes in glucose, insulin, and

    glucagon levels in response to a pharmacologic infusion of GLP-1 in patients with poorly controlled T2DM (mean HbA1c of

    11.6%).[22] In the study, blood was drawn every 30 minutes during a 4-hour IV infusion of GLP-1 (and during a placebo

    infusion the following day) to evaluate glucose, insulin, and glucagon levels. The GLP-1 infusion reduced plasma glucose to

    normal basal levels in all patients, with significant mean reductions at each time point from 60 minutes onward compared

    with placebo (P< .05). Patients had a mean baseline plasma glucose level of 228.6 mg/dL. At the start of the GLP-1

    infusion, plasma insulin initially increased and glucagon decreased in response to GLP-1. However, as plasma glucose

    approached normal basal levels, insulin returned to near-baseline levels. Furthermore, glucagonwhich was suppressed

    when glucose levels were highalso started rebounding toward normal levels, even in the presence of the continuous GLP-1infusion. These patterns in glucose, insulin, and glucagon levels illustrate the glucose-dependent nature of GLP-1, and

    demonstrate the potential therapeutic benefits of exogenous GLP-1 for normalizing fasting plasma glucose concentrations in

    patients with poorly-controlled T2DM.[22]

    Incretin-based Therapies for T2DM

    Healthcare providers currently have several options for incretin-based therapies for the management of T2DM, and other

    investigational options may soon join the treatment armamentarium (Figure 1).[23,24]

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    10/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    10/ww.medscape.org/viewarticle/775099_print

    Figure 1. Incretin-Based Therapies for T2DM[23,24]

    DPP-4 Inhibitors

    One approach to incretin-based therapy involves blocking the degradation of endogenous incretins through the inhibition of

    dipeptidyl peptidase-4 (DPP-4). The DPP-4 inhibitors include sitagliptin, saxagliptin, and linagliptin, which are currently

    approved for the treatment of T2DM (Table 3), as well as the investigational agents vildagliptin and alogliptin. DPP-4 inhibitors

    can be taken orally without regard to meals.

    Table 3. DDP-4 Inhibitors in T2DM[23]

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    11/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    11/ww.medscape.org/viewarticle/775099_print

    The efficacy and safety of DPP-4 inhibitors have been well studied in patients with T2DM. Saxagliptin added to metformin

    reduces HbA1c (P< .0001), fasting plasma glucose (P< .0001), and postprandial glucose (P< .0001) after 24 weeks

    compared with metformin alone.[25] Linagliptin significantly reduces HbA1c by -0.5% to -0.7% when given as single-agent

    therapy or when added to metformin; thiazolidinedione (TZD); sulfonylurea; and metformin and sulfonylurea.[26-30] In a head-

    to-head trial, sitagliptin added to metformin showed similar glucose-lowering effects after 52 weeks of treatment compared

    with glipizide added to metformin, but with a more favorable effect on body weight (1.5 kg weight loss vs 1.1 kg weight gain;

    P< .001) and a reduced risk of hypoglycemia (4.9% vs 32.0%). [31]

    GLP-1 Receptor Agonists (RAs)

    GLP-1 RAs are indicated for the treatment of T2DM as an adjunct to lifestyle modifications including diet and exercise.

    Currently available agents include twice-daily exenatide, once-weekly exenatide, and once-daily liraglutide (Table 4). GLP-1

    analogs improve glycemic control in patients with T2DM by enhancing glucose-dependent insulin secretion and reducing

    postprandial glucagon secretion. The GLP-1 RAs also slow gastric emptying, resulting in increased satiety, reduced food

    intake, and weight loss. However, decreased gastric emptying may also lead to nausea, vomiting, and diarrhea for some

    patients.

    Table 4. Properties of GLP-1 Agonists[32]

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    12/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    12/ww.medscape.org/viewarticle/775099_print

    * Not head-to-head trials

    Liraglutide is a long-acting subcutaneous GLP-1 analog that is administered once daily. The LEAD-6 (Liraglutide Effect and

    Act ion in Diabetes) trial compared liraglutide 1.8 mg once daily and exenatide 10 g twice daily in patients with poorly

    controlled T2DM despite treatment with metformin and/or a sulfonylurea.[33]

    Add-on treatment with l iraglutide provided agreater reduction in HbA1c compared with add-on exenatide (1.1% vs -0.8%; P< .05) and a greater reduction in fasting

    plasma glucose (-29.0 mg/dL vs -10.8 mg/dL; P< .05). Moreover, more patients in the liraglutide group than in the exenatide

    group reached the treatment goal of HbA1c < 7.0% (54.0% vs 43.0%; P< .05). Both agents were well-tolerated, and

    liraglutide and exenatide promoted similar levels of weight loss (-3.24 kg vs -2.87 kg). [33]

    Newer formulations with reduced administration frequency may help improve patient adherence to antidiabetes therapy. The

    DURATION-1 (Diabetes Therapy Utilization: Researching Changes in A1c, Weight and Other Factors Through Intervention

    with Exenatide Once Weekly) trial evaluated long-term treatment with different formulations of exenatide in patients with

    T2DM.[34] Patients were randomly assigned to treatment with exenatide 2 mg once weekly or exenatide 10 g twice daily fo

    30 weeks, followed by 1.5 years of exenatide 2 mg once weekly for all patients. Compared with twice-daily exenatide,

    treatment with once-weekly exenatide provided a greater reduction in mean HbA1c (-1.5% vs -1.9%). The DURATION-5 trial

    compared exenatide twice daily with exenatide once weekly over 24 weeks in 252 patients with T2DM. [35] The once-weekly

    formulation provided superior glycemic control compared with standard twice-daily dosing. The mean reduction in HbA1c was

    -1.6% in the exenatide once-weekly group and -0.9% in the exenatide twice-daily group (P< .0001).

    The DURATION-6 trial compared once-weekly exenatide and daily liraglutide in 911 patients with T2DM.[36] After 26 weeks,

    the mean reduction in HbA1c was 1.26% in the exenatide group and 1.48% in the liraglutide group (P< .05). Once-weekly

    exenatide was associated with fewer adverse gastrointestinal events than daily liraglutide, including less nausea (9.3% vs

    20.4%), vomiting (3.7% vs 10.7%), and diarrhea (6.1% vs 13.1%). More than twice as many patients in the liraglutide group

    (5.3%) than in the exenatide group (2.6%) discontinued treatment due to adverse events. Patients in both groups had a

    modest reduction in body weight, with no significant differences between treatments. [36]

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    13/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    13/ww.medscape.org/viewarticle/775099_print

    Treatment Considerations

    Nausea: Nausea and vomiting related to delayed gastric emptying are the most common adverse events associated with

    GLP-1 RAs. Nausea is mostly mild-to-moderate, and is most common at initiation of therapy; it tends to decrease with

    continuing treatment. In 1 prospective study, the frequency of nausea was similar during weeks 26 to 52 of treatment for

    patients treated with liraglutide 1.8 mg/d, liraglutide 1.2 mg/d, or sitagliptin 100 mg/d. [37] In the LEAD-6 trial, the risk of

    nausea was significantly higher for patients treated with exenatide 10 g twice daily compared with those treated with

    liraglutide 1.8 mg/d (P< .0001).[33] In DURATION-6, nausea was less frequent among patients who received once-weekly

    exenatide (9.3%) than among those who received liraglutide (20.4%).[36]

    Weight loss: In a meta-analysis of GLP-1 RAs, the magnitude of weight loss was similar for exenatide once weekly (mean:

    -2.8 kg; 95% CI: -5.2 to -0.3 kg), exenatide twice daily (mean: -2.8 kg; 95% CI: -2.9 to -2.7 kg), and liraglutide (mean: -2.8

    kg; 95% CI: -3.5 to -0.9 kg).[38] Several studies have demonstrated that the weight loss associated with GLP-1 RA treatment

    is sustained for at least 2 years.[39-41]

    Use in patients on insulin therapy: Twice-daily exenatide has also been evaluated as add-on therapy in patients treated

    with glargine insulin.[42] In the prospective trial, 259 patients who were taking glargine insulin were randomly assigned to add

    on therapy with placebo or twice-daily exenatide. Compared with placebo, exenatide was associated with a greater reduction

    in HbA1c (-1.0% vs -1.7%), more favorable effects on body weight (+1.0 kg vs -1.8 kg), and a smaller increase in glargine

    dose (20 units vs 13 units). Patients had a similar risk of hypoglycemia regardless of treatment group. However, more

    patients in the exenatide group than in the placebo group dropped out of the study (13 vs 1).

    Use in patients with comorbid renal impairment: Renal impairment appears to impact the clearance of exenatide in

    patients with T2DM, but does not affect the metabolism of liraglutide. [43,44] This is because exenatide is eliminated

    predominantly via glomerular filtration, whereas no specific organ serves as the major route of elimination for liraglutide.[23]

    Table 5 summarizes the recommended dosing for exenatide and liraglutide for patients with T2DM and comorbid renal

    impairment.[23]

    Table 5. GLP-1 Receptor Agonist Dosing in Patients with Renal Impairment[23]

    *Hypovolemia due to nausea/vomiting may worsen renal function

    CrCl = creatinine clearance; ESRD = end-stage renal disease

    Safety Considerations

    Pancreatitis: In 2008, the Food and Drug Administration (FDA) issued an alert to healthcare professionals regarding the risk

    of pancreatitis with incretin-related drugs.[45] The FDA report described several issues with incretin agents including:

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    14/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    14/ww.medscape.org/viewarticle/775099_print

    Reports of pancreatitis in liraglutide clinical trials[46-48]

    Postmarketing reports of pancreatitis in patients taking exenatide, including cases of hemorrhagic/necrotizing

    pancreatitis that resulted in patient deaths[45]

    Later, an analysis from the FDA Adverse Event Reporting System (AERS) reported[45]:

    A 6-fold increase of pancreatitis with sitagliptin or exenatide compared with other antidiabetes medications, including

    rosiglitazone, nateglinide, repaglinide, and glipizide[49]

    An increase in the reporting rate of pancreatic cancer in patients treated with sitagliptin or exenatide[49]

    Despite the value of these safety warnings, the AERS has limitations for understanding true pancreatitis risk, including

    potential reporting bias.[50] In addition, the AERS reporting system did not collect information on BMI, a known risk factor for

    pancreatitis.

    Healthcare professionals should consider the following precautions related to pancreatitis when treating patients with

    incretin-based therapies[45,51-52]:

    Adhere to label warnings

    Observe patients for symptoms of acute pancreatitis (persistent severe abdominal pain that may be accompanied by

    vomiting)

    Discontinue drug if pancreatitis is suspected

    Do not restart drug if pancreatitis is confirmed

    Consider other antihyperglycemic therapies in patients with a history of pancreatitis

    Thyroid cancer: Liraglutide and once-weekly exenatide carry boxed warnings for thyroid C-cell tumors based on data from

    rodent models of T2DM.[53,54] Clinical trials and postmarketing data show conflicting results regarding the effects of GLP-1

    analogs on calcitonin levels, with small increases in calcitonin levels in some studies and reduced levels in others. [33,53] To

    date, no definitive cases of medullary thyroid carcinoma related to liraglutide have been detected. [53,54] No cases of thyroid

    cancer have been reported in the exenatide clinical trials.

    Renal impairment: Postmarketing reports have also tracked renal outcomes in patients treated with GLP-1 RAs. To date,

    no evidence indicates that GLP-1 RAs are directly toxic to kidney cells. [23,55,56] However, renal impairment may occur in

    patients who have experienced nausea, vomiting, diarrhea, and dehydration.[23,56-60] In some cases, renal impairment has

    been observed in patients who are taking concurrent medication known to affect renal function or hydration status (eg,

    angiotensin-converting enzyme [ACE] inhibitors, nonsteroidal anti-inflammatory drugs [NSAIDs], or diuretics). [23,56-60] In

    addition, cases of patients requiring hemodialysis or transplantation have also been reported. [23] Impaired renal function has

    also been observed in patients without known underlying renal disease.[23]

    In many cases, treatment-emergent renal impairment is reversible with supportive treatment and discontinuation of potentiall

    causative agents.[23] Current labeling includes a warning against GLP-1 RA use in patients with severe renal impairment or

    end-stage renal disease (ESRD).[23] Furthermore, GLP-1 RAs should be used with caution in patients with a history of renal

    transplantation (exenatide and exenatide once weekly), and when initiating or escalating exenatide doses in patients with

    T2DM and comorbid renal impairment (exenatide, exenatide once weekly, and liraglutide). [23]

    Patient Case: Uncontrolled T2DM in a Patient with Poor Treatment Adherence

    Robert is a 58-year-old man with a history of hypertension, T2DM, and dyslipidemia. His physical examination shows that he

    is overweight (BMI of 38.8 kg/m2) with a blood pressure of 130/78 mm Hg, a heart rate of 76 bpm, and 2-mm pitting edema.

    His laboratory results include the following: HbA1c: 7.8%; LDL: 105 mg/dL; HDL: 32 mg/dL; triglycerides: 225 mg/dL; blood

    urea nitrogen (BUN): 19 mg/dL; and serum creatinine: 1.5 mg/dL.

    Robert has been trying to lose weight for years but cannot stick with any regimen. He also admits that he does not always

    remember to take his medications. His current medications include metformin 2000 mg/d, glimepiride 4 mg/d, pioglitazone

    30 mg/d, metoprolol 100 mg twice daily, simvastatin 20 mg/d, and aspirin 75 mg/d.

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    15/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    15/ww.medscape.org/viewarticle/775099_print

    In addition to lifestyle changes and referral to a dietitian, what changes would you make to get the patients

    HbA1c to the ADAs recommended goals?

    Increase the dose of metformin

    Discontinue metformin, glimepiride, and pioglitazone, and initiate basal insulin therapy

    Discontinue metformin and pioglitazone, continue glimepiride, and add incretin-based therapy

    Discontinue pioglitazone, continue metformin and glimepiride, and initiate GLP-1 therapy

    Save and Proceed

    Summary

    The incretin hormone system is a powerful driver of postprandial blood glucose control. In patients with T2DM, this system is

    defective, leading to a range of adverse effects on glycemic control. Incretin-based therapies provide an intriguing

    constellation of effects for patients with T2DM. The GLP-1 RAs provide effective postprandial glucose control and stimulate

    weight loss with the convenience of once-weekly dosing. The DPP-4 inhibitors also control postprandial glucose with weight-

    neutral effects, and can be used in combination with other oral agents in a single tablet. The incretin-based therapies are

    associated with a low risk of hypoglycemia, except when used in combination with sulfonylurea. In addition, these agents

    are safe in patients with renal disease, although they may require dose adjustments depending on the severity of renal

    impairment. The gastrointestinal side effects associated with GLP-1 RAs are typically transient, with longer-acting

    formulations associated with less severe nausea.

    Achieving effective glycemic control in T2DM is a challenge for both patients and healthcare providers. Incretin-based

    therapies represent an important new treatment option for patients who remain poorly controlled despite lifestyle

    modifications and standard first-line pharmacotherapy. During treatment with incretin-based therapies, patients and clinicians

    should be aware of the signs and symptoms of pancreatitis and other possible side effects of therapy. These agents should

    be used with caution in patients with a history of thyroid cancer, and renal status should be monitored for the duration of

    treatment. When necessary, treatment regimens should be modified to improve tolerability and/or convenience to increase

    the likelihood of patient adherence and improved glycemic control.

    Michael D. Shapiro, DO, FACC, FSCCT

    Cardiovascular disease (CVD) is a major burden for patients with diabetes. Indeed, CVD is the major cause of morbidity and

    mortality in patients with T2DM, and the largest contributor to direct and indirect costs related to diabetes care.[62] T2DM is

    regarded as a coronary artery disease (CAD)-equivalent, because it elevates patients with T2DM to the same risk category

    as nondiabetic patients with a history of myocardial infarction (MI). In a long-term study of MI risk in diabetic and nondiabetic

    patients, those with T2DM and no history of MI were just as likely to suffer a future MI as were nondiabetic patients who had

    already sustained an MI (20% vs 19%, respectively). [63]

    Atherosclerosis, the progressive accumulation of cholesterol in the arterial wall and resultant narrowing of the arterial lumen,

    is the central pathologic mechanism of CAD and other macrovascular complications such as peripheral artery disease (PAD

    and stroke in patients with diabetes.[64] The Emerging Risk Factors Collaboration (ERFC) group conducted a meta-analysis

    of 102 prospective studies enrolling 698,782 patients to measure the association between T2DM and the development of

    vascular disease.[65] Compared with normal fasting blood glucose, a diagnosis of T2DM approximately doubled the risk for

    vascular disease and adverse vascular outcomes, including coronary heart disease (CHD) (HR: 2.0); ischemic stroke (HR:

    2.27); and vascular deaths (HR: 1.73).

    T2DM represents a ticking clock of cardiovascular risk, as the deleterious effects of endothelial dysfunction begin to

    accumulate long before a clinical diagnosis of T2DM is made. The Nurses' Health Study evaluated long-term cardiovascular

    risk in 117,629 women without CVD at baseline. [66] Of these, 5894 women developed T2DM during the 20-year follow-up.

    Compared with women who never developed diabetes, those who eventually developed T2DM were 3 times more likely to

    have an MI in the time period before their diabetes diagnosis (RR: 3.17; 95% CI: 2.613.85), and nearly 4 times as likely to

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    16/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    16/ww.medscape.org/viewarticle/775099_print

    have an MI after they were diagnosed with T2DM (RR: 3.97; 95% CI: 3.35-4.71). [66] These findings underscore the

    importance of early and aggressive detection and treatment of cardiovascular risk factors for patients with T2DM.

    There is unequivocal evidence that aggressive management of traditional cardiovascular risk factors in patients with T2DM

    results in improved clinical outcomes. Numerous trials have demonstrated the benefits of blood pressure control on both

    micro- and macrovascular events.67-77] In addition, aggressive LDL cholesterol control, specifically with statins, has resulted

    in improvements in macrovascular outcomes.[67,75,76,78] With that in mind, there have been several large trials to test the

    hypotheses that intensive glucose lowering would improve both micro- and macrovascular event rates. Indeed, the clinical

    trials have demonstrated consistent reductions in microvascular disease outcomes with aggressive glucose control in both

    patients with T1DM and T2DM.[67,74-76,78]

    Intensive glycemic control confers a range of benefits for patients with T2DM, and has become the standard of care for

    reducing the microvascular complications of diabetes. The first major trial to demonstrate the benefits of intensive therapy in

    patients with T2DM was the landmark UKPDS (United Kingdom Prospective Diabetes Study), which compared conventional

    dietary therapy with intensive glucose-lowering therapy with sulfonylurea, metformin, or insulin in patients with newly-

    diagnosed T2DM.[78] A long-term follow-up analysis of data from UKPDS revealed a "legacy effect" of intensive glucose

    control in T2DM, showing that the magnitude of the benefits of intensive therapy grew over time.[67] Compared with patients

    who received standard treatment during the UKPDS trial, those in the sulfonylurea-insulin group had a 24% reduction in

    microvascular disease (P= .001), a 15% reduction in MI (P= .014), a 13% reduction in all-cause mortality (P= .007), and a

    9% reduction in any diabetes-related endpoint (P= .040) that persisted during 10 years of posttrial follow-up.[67]

    Glycemic Control: How Much Is Too Much?

    While intensive glucose lowering has translated into a consistent reduction in microvascular complications, demonstrating a

    beneficial effect of intensive glucose lowering on macrovascular complications has been elusive. However, recent trials have

    shown a potential for harm associated with intensive glucose control, including an increased risk of severe hypoglycemia and

    death. These findings have led the diabetes community to re-evaluate the appropriateness of aggressive treatment goals for

    certain subgroups of patients with T2DM.

    The ACCORD (Action to Control Cardiovascular Risk in Diabetes) trial included 10,251 patients with a mean age of 62 years,

    a median duration of T2DM of 10 years, and known heart disease or at least 2 risk factors for CHD at the time of

    enrollment.[72] Patients in the intensive glucose-lowering arm were treated to an HbA1c goal of < 6.0%, while those in the

    standard therapy group were treated to an HbA1c goal of 7.0% to 7.9%. The primary endpoint was the composite outcome o

    nonfatal MI, nonfatal stroke, or death from CVD. The ACCORD trial was discontinued early due to excess deaths in the

    intensive therapy group and no reduction in total cardiovascular events compared with standard therapy (HR: 0.90; P= .16).

    In particular, more cardiovascular deaths occurred with intensive therapy than with standard therapy (14 vs 11 deaths per

    1000 patients per year).

    The excess deaths in the intensive therapy group in ACCORD contributed to the growing concern that there may be a

    threshold for target HbA1c levels, below which the risk of adverse events increases. Moreover, these findings highlight the

    importance of selecting patients for whom the risk/benefit ratio favors aggressive therapy. For instance, subgroup analyses

    from ACCORD showed a significant reduction in the primary endpoint for patients with no history of prior cardiovascular

    events (P= .04 vs a history of prior cardiovascular events) and in those with baseline HbA1c levels 8.0% ( P= .03 vs

    HbA1c > 8.0%).[72]

    Other major studies in T2DM provide conflicting results about the role of intensive glucose-lowering therapy in controlling

    diabetes-related complications. The VADT (VA Diabetes Trial) included 10,251 patients with a mean age of 60 years, a

    median duration of T2DM of 11.5 years, and a mean HbA1c of 9.4% at the time of enrollment. [73] Patients were randomly

    assigned to treatment with standard (HbA1c < 9.0%) or intensive (HbA1c < 6.0%) treatment goals. In VADT, intensive

    glucose-lowering therapy in this population of patients with advanced, long-standing T2DM increased the risks of

    hypoglycemia and weight gain without providing additional vascular benefits beyond those achieved with s tandard therapy. In

    contrast with VADT, the ADVANCE (Action in Diabetes and Vascular Disease: Preterax and Diamicron MR Controlled

    Evaluation) study found that intensive glucose-lowering therapy may provide some benefit to high-risk patients with T2DM.

    ADVANCE enrolled 11,140 patients with a mean age of 66 years, a median duration of T2DM of 8 years, a mean HbA1c of

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    17/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    17/ww.medscape.org/viewarticle/775099_print

    7.48%, and a history of a major cardiovascular event or at least one risk factor at baseline.[74] These patients were randomly

    assigned to standard care or to an intensive regimen targeting an HbA1c level 6.5%. In this high-risk population, intensive

    glucose-lowering therapy lowered the risk of renal complications by 21%.

    In general, the major trials of intensive glucose-lowering therapy in T2DM suggest a benefit related to microvascular

    complications such as nephropathy, retinopathy, and neuropathy (Table 6). Long-term follow-up of patients enrolled in the

    UKPDS study demonstrated statistically significant reductions in nonfatal MI (RR: 15%; P= .014) and all-cause mortality

    (RR: 13%; P= .007) as a result of intensive therapy.[67,78] By comparison, there was no evidence of improvement in

    cardiovascular endpoints in the ACCORD, ADVANCE, or VADT trials.[72-74,77]

    Table 6. Impact of Intensive Glycemic Therapy on T2DM Outcomes[67,72-78]

    Intensive therapy confers some cardiovascular benefits, but does not protect patients from all adverse outcomes. A meta-

    analysis of the UKPDS, ACCORD, ADVANCE, and VADT trials included 27,802 patients with T2DM who were treated with

    intensive glucose-lowering therapy or standard care.[79] In the pooled analysis, intensive therapy was associated with a 16%reduction in nonfatal MI (RR: 0.84; 95% CI: 0.750.94), with an absolute overall risk reduction of 9 events per 1000 patients

    over 5 years of treatment. However, intensive glucose-lowering therapy had no significant effect on either cardiovascular

    mortality (RR: 0.97; 95% CI: 0.761.24) or all-cause mortality (RR: 0.98; 95% CI: 0.841.15). [79]

    New Options for Cardiovascular Risk Reduction in T2DM

    Patients with T2DM may benefit from interventions that reduce the risk of CVD over and above what can be expected from

    glucose lowering itself. New therapeutic targets with important implications for cardiovascular risk reduction include the

    incretin-based therapies, including the GLP-1 RAs and the DPP-4 antagonists.

    GLP-1 is associated with diverse physiologic effects across several organ systems that may translate into cardiovascular

    benefits. Such favorable effects include deceleration of gastric emptying; reduction in appetite; earlier induction of satiety;

    weight reduction; increased beta-cell mass; enhanced insulin sensitivity in the liver, muscle, and adipose tissue; and

    improvements in traditional cardiovascular risk factors and function. The GLP-1 receptor (GLP-1R) is expressed in several

    key structures that affect cardiac function, including myocytes, the pericardium, vascular epithelium, and vascular smooth

    muscle. In preclinical studies, GLP-1R agonism induces dose-dependent vasodilation.[80] Furthermore, GLP-1R agonism is

    associated with increased myocardial glucose uptake; attenuation of ischemic injury; limitation of infarct size; improvement

    in hemodynamic measures such as stroke volume, cardiac output, and ejection fraction; and systemic resistance.[81-85]

    Conversely, the protective effects on cardiac function are diminished by GLP-1R antagonism. [81]

    Treatment with GLP-1 agonists also shows beneficial effects on cardiometabolic markers in patients with diabetes. In a

    prospective study of 217 patients with T2DM, treatment with add-on exenatide was associated with sustained improvements

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    18/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    18/ww.medscape.org/viewarticle/775099_print

    in systolic blood pressure (BP) (P< .01) and diastolic BP (P< .0001) over 3 years of treatment compared with baseline.[39]

    Long-term treatment with adjuvant exenatide also significantly improves endothelial function, as measured by flow-mediated

    vasodilation, in patients with T2DM (P< .05 vs baseline).[86]

    GLP-1 agonists also promote weight reduction in overweight and obese patients with T2DM. The phase 3 AMIGO (AC2993

    Diabetes Management for Improving Glucose Outcomes) trials compared twice-daily subcutaneous exenatide with placebo in

    patients with T2DM who were also taking metformin (AMIGO 1), a sulfonylurea (AMIGO 2), or both metformin and a

    sulfonylurea (AMIGO 3).[87-89] In each of these patient groups, add-on therapy with exenatide significantly reduced HbA1c

    compared with placebo.[87-89] The mean absolute reduction in HbA1c varied from -0.4% to -0.6% with exenatide 5 g twice

    daily and from -0.8% to -0.9% with exenatide 10 g twice daily in patients on background oral therapy. [87-89] In the AMIGO

    clinical trials program, add-on treatment with exenatide also resulted in significant weight loss compared with placebo. Mean

    reductions in body weight with high-dose exenatide ranged from -1.6 kg to -2.8 kg after 30 weeks (P< .05 vs placebo) of

    treatment.[87-89]

    Exenatide also compares favorably to insulin in patients with poorly controlled T2DM. In 3 head-to-head studies of exenatide

    compared with insulin glargine or insulin aspart, the glycemic effects of treatment were comparable. [90-92] The mean HbA1c

    reduction ranged from -0.9% to -1.4% in the insulin groups and from -1.0% to -1.4% in the exenatide groups. However,

    patients in the insulin groups gained weight during the study (mean increase: 1.8 kg to 2.9 kg), while those in the exenatide

    groups lost weight (mean decrease: -2.2 kg to -2.5 kg). [90-92]

    Liraglutide, another GLP-1 analog, is also associated with a dose-dependent reduction in body weight.[93]

    In a phase 2 studyof patients with T2DM, those in the liraglutide 1.9 mg group had a mean loss of -3.0 kg after 14 weeks of treatment (P= .04

    vs placebo).[93] In addition to promoting weight loss, both exenatide and liraglutide improve other markers of cardiovascular

    risk (Table 7).

    Table 7. GLP-1 Agonists and Cardiovascular Risk Factors[39,94,95]

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    19/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    19/ww.medscape.org/viewarticle/775099_print

    LAR = long-acting release; NR = not reported

    *P< .05 vs baseline; P< .05 vs placebo

    Beyond improving biomarkers of cardiovascular risk, preliminary data suggest that treatment with GLP-1 agonists confers

    direct benefits on cardiac function in the setting of established CVD. For example, in a preclinical model of T2DM, GLP-1

    administration following coronary artery occlusion reduced infarct s ize and activated other prosurvival signaling pathways

    associated with improved outcomes and prolonged survival. [96] In a small trial of patients with severe heart failure, infusion of

    GLP-1 over 5 weeks significantly improved left ventricular function, functional status, and quality of life. [97]

    Recent safety data provide additional insight on the potential role of incretin-based therapy in the management of patients

    with T2DM. A meta-analysis of 8 randomized, double-blind, phase 2 and 3 trials evaluated the cardiovascular safety of the

    DPP-4 inhibitor saxagliptin in 4607 patients with T2DM. [98] There was no evidence of increased cardiovascular risk with

    saxagliptin used either as monotherapy or in combination with other oral antidiabetes agents. Indeed, treatment with

    saxagliptin was associated with a 66% reduction in the risk of major adverse cardiovascular events (HR: 0.44), and a 41%

    reduction in the risk of acute clinically significant events (HR: 0.59) compared with control. Saxagliptin was also associated

    with a lower risk of all-cause mortality compared with control (0.3% vs 1.0%), as well as a lower risk of cardiovascular death

    (0.2% vs 0.8%).

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    20/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    20/ww.medscape.org/viewarticle/775099_print

    Ongoing and planned clinical trials will provide further evidence to guide the safe and effective use of incretin-based therapies

    in patients with T2DM. Together these trials will enroll more than 60,000 patients and collect 304,170 patient-years of follow-

    up data on hard cardiovascular endpoints (Table 8).

    Table 8. Ongoing and Planned Cardiovascular Outcome Trials[24]

    Source: National Institutes of Health

    Patient Case: Mary

    Mary is a 50-year-old woman who was diagnosed with T2DM 5 years ago. Her creatinine level is 1.4 mg/dL. She stopped

    metformin and started treatment with glimepiride. She has had 2 episodes of hypoglycemia with glucose levels of 5055

    mg/dL. She is a nonsmoker with a history of longstanding hypertension, hyperlipidemia, stage 3 chronic kidney disease,

    sleep apnea, and osteoarthritis. On her last exam, her ejection fraction (EF) was 45% by echocardiography. Her mother died

    of heart failure at age 70, and her father died following an MI at age 50. Her current medications include aspirin 81 mg/d,

    glimepiride 4 mg/d, lisinopril 20 mg/d, simvastatin 40 mg/d, and fish oil 1 g/d.

    On her physical exam, she is obese (BMI: 46.7 kg/m2) and has 2+ pitting edema. Her BP is 126/70 mm Hg. Her laboratory

    findings include the following: LDL: 90 mg/dL; HDL: 35 mg/dL; triglycerides: 170 mg/dL; HbA1c: 7.5%; creatinine: 1.4 mg/dL

    Stress echocardiography demonstrated no evidence of ischemia.

    What is your next step regarding Marys glucose-lowering therapy?

    No change in therapy; schedule follow-up in 3 months

    Discontinue glimepiride and restart metformin

    Add an incretin agent to background glimepiride

    Decrease glimepiride and initiate an incretin agent

    Discontinue glimepiride and initiate a TZD

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    21/25

    Discontinue glimepiride and initiate basal insulin

    Save and Proceed

    This article is a CME certified activity. To earn credit for this activity visit:

    http://www.medscape.org/viewarticle/775099

    1. Hollander P, Spellman C. Controversies in prediabetes: do we have a diagnosis? Postgrad Med. 2012;124:109-118.

    2. World Health Organization. Use of glycated haemoglobin (HbA1c) in the diagnosis of diabetes mellitus. 2011.

    http://www.who.int/diabetes/publications/report-hba1c_2011.pdf. Accessed on November 15, 2012.

    3. Alberti G, et al. Program and abstracts of the 1st International Congress on "Prediabetes" and the Metabolic

    Syndrome; Berlin, Germany. 2005;13-16.

    4. Standards of medical care in diabetes--2010. Diabetes Care. 2010;33 Suppl 1:S11-61.

    5. Handelsman Y, Mechanick JI, Blonde L, et al. AACE Task Force for Developing Diabetes Comprehensive Care Plan.

    American Association of Clinical Endocrinologists Medical Guidelines for Clinical Practice for developing a diabetes

    mellitus comprehensive care plan. Endocr Pract. 2011;17(suppl 2):1-53.

    6. Karve A, Hayward RA. Prevalence, diagnosis, and treatment of impaired fasting glucose and impaired glucose

    tolerance in nondiabetic U.S. adults. Diabetes Care. 2010;33:2355-2359.7. Buysschaert M, Bergman M. Definition of prediabetes. Med Clin North Am. 2011;95:289-297, vii.

    8. Heianza Y, Hara S, Arase Y, et al. HbA1c 5.7-6.4% and impaired fasting plasma glucose for diagnosis of prediabetes

    and risk of progression to diabetes in Japan (TOPICS 3): a longitudinal cohort study. Lancet. 2011;378:147-155.

    9. Karve A, Hayward RA. Prevalence, diagnosis, and treatment of impaired fasting glucose and impaired glucose

    tolerance in nondiabetic U.S. adults. Diabetes Care. 2010;33:2355-2359.

    10. Cosson E, Hamo-Tchatchouang E, Banu I, et al. A large proportion of prediabetes and diabetes goes undiagnosed

    when only fasting plasma glucose and/or HbA1c are measured in overweight or obese patients. Diabetes Metab.

    2010;36:312-318.

    11. Cosson E, Nguyen MT, Hamo-Tchatchouang E, et al. What would be the outcome if the American Diabetes

    Association recommendations of 2010 had been followed in our practice in 1998-2006? Diabet Med. 2011;28:567-574

    12. Silventoinen K, Pankow J, Lindstrom J, et al. The validity of the Finnish Diabetes Risk Score for the prediction of theincidence of coronary heart disease and stroke, and total mortality. Eur J Cardiovasc Prev Rehabil. 2005;12:451-458.

    13. Costa B, Barrio F, Cabre JJ, et al. Shifting from glucose diagnostic criteria to the new HbA(1c) criteria would have a

    profound impact on prevalence of diabetes among a high-risk Spanish population. Diabet Med. 2011;28:1234-1237.

    14. Bersoux S, Cook CB, Wu Q, et al. Hemoglobin A1c testing alone does not sufficiently identify patients with

    prediabetes.Am J Clin Pathol. 2011;135:674-677.

    15. Ast rup A, Rossner S, Van Gaal L, et al. Effects of liraglutide in the treatment of obesity: a randomised, double-blind,

    placebo-controlled study. Lancet. 2009;374:1606-1616.

    16. Lu YH, Lu JM, Wang SY, et al. Outcome of intensive integrated intervention in participants with impaired glucose

    regulation in China.Adv Ther. 2011;28:511-519.

    17. Saito T, Watanabe M, Nishida J, et al. Lifestyle modification and prevention of type 2 diabetes in overweight Japanese

    with impaired fasting glucose levels: a randomized controlled trial.Arch Intern Med. 2011;171:1352-1360.

    18. Rosenstock J, Klaff LJ, Schwartz S, et al. Effects of exenatide and lifesty le modification on body weight and glucose

    tolerance in obese subjects with and without pre-diabetes. Diabetes Care. 2010;33:1173-1175.

    19. Perreault L, Pan Q, Mather KJ, et al. 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;379:2243-

    2251.

    20. Holst JJ, Orskov C. The incretin approach for diabetes treatment: modulation of islet hormone release by GLP-1

    agonism. Diabetes. 2004;53 Suppl 3:S197-204.

    21. Meier JJ, Nauck MA. Glucagon-like peptide 1 (GLP-1) in biology and pathology. Diabetes Metab Res Rev.

    2005;21:91-117.

    22. Nauck MA, Kleine N, Orskov C, et al. Normalization of fasting hyperglycaemia by exogenous glucagon-like peptide 1

    (7-36 amide) in type 2 (non-insulin-dependent) diabetic patients. Diabetologia. 1993;36:741-744.

    http://www.medscape.org/viewarticle/775099
  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    22/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    22/ww.medscape.org/viewarticle/775099_print

    23. US Food and Drug Administration. Drugs@FDA Web site. http:/ /www.accessdata.fda.gov/Scripts/cder/DrugsatFDA.

    Accessed November 15, 2012.

    24. US National Institutes of Health. Clinicaltrials.gov. http://www.clinicaltrials.gov/ct2/home. Accessed November 15,

    2012.

    25. DeFronzo RA, Hissa MN, Garber AJ, et al. 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;32:1649-1655.

    26. Del Prato S, Barnett AH, Huisman H, et al. Effect of linagliptin monotherapy on glycaemic control and markers of

    beta-cell function in patients with inadequately controlled type 2 diabetes: a randomized controlled trial. Diabetes

    Obes Metab. 2011;13:258-267.

    27. 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;13:65-74.

    28. Gomis R, Espadero RM, Jones R, et al. Efficacy and safety of initial combination therapy with linagliptin and

    pioglitazone in patients with inadequately controlled type 2 diabetes: a randomized, double-blind, placebo-controlled

    study. Diabetes Obes Metab. 2011;13:653-661.

    29. Lewin A, Arvay L, Liu D, et al. Safety and efficacy of linagliptin as add-on therapy to a sulphonylurea in inadequately

    controlled type 2 diabetes. Diabetologia 2010; 53 (suppl1):S326.

    30. Owens DR, Swallow R, Dugi KA, et al. 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. Diabetic Medicine.

    2011;28:1352-1361.

    31. Nauck MA, Meininger G, Sheng D, et al. 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;9:194-205.

    32. Gallwitz B. Glucagon-like peptide-1 analogues for Type 2 diabetes mellitus: current and emerging agents. Drugs.

    2011;71:1675-1688.

    33. Buse JB, Rosenstock J, Sesti G, et al. Liraglutide 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;374:39-47.

    34. Taylor K, Gurney K, Han J, et al. Exenatide once weekly treatment maintained improvements in glycemic control and

    weight loss over 2 years. BMC Endocr Disord. 2011;11:9.

    35. Blevins T, Pullman J, Malloy J, et al. DURATION-5: exenatide once weekly resulted in greater improvements in

    glycemic control compared with exenatide twice daily in patients with type 2 diabetes. J Clin Endocrinol Metab.

    2011;96:1301-1310.36. Buse J, et al. Efficacy and safety of exenatide once weekly versus liraglutide in subjects with type 2 diabetes

    (DURATION-6): a randomised, open-label study. Presented at: 47th European Association for the Study of Diabetes;

    September 12-16, 2011; Lisbon, Portugal. Abstract 75.

    37. Pratley R, Nauck M, Bailey T, et al. 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;65:397-407.

    38. Vilsboll T, Christensen M, Junker AE, et al. Effects of glucagon-like peptide-1 receptor agonists on weight loss:

    systematic review and meta-analyses of randomised controlled trials. BMJ. 2012;344:d7771.

    39. 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.

    40. Garber A, Henry RR, Ratner R, et al. Liraglutide, a once-daily human glucagon-like peptide 1 analogue, providessustained improvements in glycaemic control and weight for 2 years as monotherapy compared with glimepiride in

    patients with type 2 diabetes. Diabetes Obes Metab. 2011;13:348-356.

    41. MacConell L, Walsh B, Li Y, et al. Exenatide once weekly: sustained improvement in glycemic control and weight

    loss through 3 years. ADA 71st Scientific Sessions. San Diego, CA; June 24-28, 2011. Abstract 969-P.

    42. 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;154:103-112.

    43. Linnebjerg H, Kothare PA, Park S, et al. Effect of renal impairment on the pharmacokinetics of exenatide. Br J Clin

    Pharmacol. 2007;64:317-327.

    44. Jacobsen LV, Hindsberger C, Robson R, et al. Effect of renal impairment on the pharmacokinetics of the GLP-1

    analogue liraglutide. Br J Clin Pharmacol. 2009;68:898-905.

    45. US FDA Web site. Exenatide (marketed as Byetta) Healthcare Professional Sheet text version (8/2008). Available

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    23/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    23/ww.medscape.org/viewarticle/775099_print

    at: www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProviders/ucm124713.htm.

    Accessed November 15, 2012.

    46. Marre M, Shaw J, Brandle M, et al. Liraglutide, a once-daily human GLP-1 analogue, added to a sulphonylurea over

    26 weeks produces greater improvements in glycaemic and weight control compared with adding rosiglitazone or

    placebo in subjects with Type 2 diabetes (LEAD-1 SU). Diabet Med. 2009;26:268-278.

    47. Nauck M, Frid A, Hermansen K, et al. Efficacy 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;32:84-90.

    48. Garber A, Henry R, Ratner R, et al. Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a

    randomised, 52-week, phase III, double-blind, parallel-treatment trial. Lancet. 2009;373:473-481.

    49. Elashoff M, Matveyenko AV, Gier B, et al. Pancreatit is, pancreatic, and thyroid cancer with glucagon-like peptide-1-

    based therapies. Gastroenterology. 2011;141:150-156.

    50. 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;25:1019-1027.

    51. Byetta fact sheet. Available at: www.byettahcp.com/pdf/BYETTA_Fact_Sheet_Safety_Update.pdf. Accessed

    November 15, 2012.

    52. Victoza Full Prescribing Information, Novo Nordisk, Inc. 2010.

    53. FDA briefing materials liraglutide. Available at: www.fda.gov/ohrms/dockets/ac/09/briefing/2009-4422b2-01-FDA.pdf.

    Accessed November 15, 2012.

    54. Update on FDA Advisory Committee meeting on liraglutide for the treatment of type 2 diabetes (2 April 2009).

    Available at: http:/ /www.novonordisk.com/press/sea/sea.asp?sShowNewsItemGUID=a593b32a-0302-45cb-a0a8-

    7173188b9553&sShowLanguageCode=en-GB. Accessed November 15, 2012.

    55. Pendergrass M, Chen W. Exenatide is not associated with increased acute renal failure. ADA 70th Scientific

    Sessions. Orlando, FL; June 25-29, 2010. Abstract 11-LB.

    56. Tuttle K, Brown C, Lee D, et al. Effect of exenatide BID on kidney function and adverse events in clinical trials. ADA

    71st Scientific Sessions. San Diego, CA; June 24-28, 2011.

    57. Johansen OE, Whitfield R. Exenatide may aggravate moderate diabetic renal impairment: a case report. Br J Clin

    Pharmacol. 2008;66:568-569.

    58. Weise WJ, Sivanandy MS, Block CA, et al. Exenatide-associated ischemic renal failure. Diabetes Care.

    2009;32:e22-23.

    59. Ferrer-Garcia JC, Martinez-Chanza N, Tolosa-Torrens M, et al. Exenatide and renal failure. Diabet Med. 2010;27:728-729.

    60. Lopez-Ruiz A, del Peso-Gilsanz C, Meoro-Aviles A, et al. Acute renal failure when exenatide is co-administered with

    diuretics and angiotensin II blockers. Pharm World Sci. 2010;32:559-561.

    61. Physicians' Desk Reference, NJ: Thomson Reuters. 2010.

    62. American Diabetes Association. Standards of Medical Care in Diabetes2011. Diabetes Care. 2011;34:S11-S61.

    63. Haffner SM, Lehto S, Ronnemaa T, et al. Mortality from coronary heart disease in subjects with type 2 diabetes and

    in nondiabetic subjects with and without prior myocardial infarction. N Engl J Med. 1998;339:229-234.

    64. Fowler MJ. Microvascular and Macrovascular Complications of Diabetes. Clinical Diabetes. 2008;26:77-82.

    65. The Emerging Risk Factors C. Diabetes mellitus, fasting blood glucose concentration, and risk of vascular disease: a

    collaborative meta-analysis of 102 prospective studies. Lancet. 2010;375:2215-2222.

    66. Hu FB, Stampfer MJ, Haffner SM, et al. Elevated risk of cardiovascular disease prior to clinical diagnosis of type 2diabetes. Diabetes Care. 2002;25:1129-1134.

    67. Holman RR, Paul SK, Bethel MA, et al. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J

    Med. 2008;359:1577-1589.

    68. Shepherd J, Barter P, Carmena R, et al. Effect of lowering LDL cholesterol substantially below currently

    recommended levels in patients with coronary heart disease and diabetes: the Treating to New Targets (TNT) study.

    Diabetes Care. 2006;29:1220-1226.

    69. Sever PS, Poulter NR, Dahlf B, et al. Reduction in cardiovascular events with atorvastatin in 2,532 patients with type

    2 diabetes. Diabetes Care. 2005;28:1151-1157.

    70. Colhoun HM, Betteridge DJ, Durrington PN, et al. Primary prevention of cardiovascular disease with atorvastatin in

    type 2 diabetes in the Collaborative Atorvastatin Diabetes Study (CARDS): multicentre randomised placebo-controlled

    trial. Lancet. 2004;364:685-696.

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    24/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    24/ww.medscape.org/viewarticle/775099_print

    71. Collins R, Armitage J, Parish S, et al. MRC/BHF Heart Protection Study of cholesterol-lowering with simvastatin in

    5963 people with diabetes: a randomised placebo-controlled trial. Lancet. 2003;361:2005-2016.

    72. Gerstein HC, Miller ME, Byington RP, et al. Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med.

    2008;358:2545-2559.

    73. Duckworth W, Abraira C, Moritz T, et al. Glucose control and vascular complications in veterans with type 2 diabetes

    N Engl J Med. 2009;360:129-139.

    74. 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;358:2560-2572.

    75. Nathan DM, Cleary PA, Backlund JY, et al. Intensive diabetes treatment and cardiovascular disease in patients with

    type 1 diabetes. N Engl J Med. 2005;353:2643-2653.

    76. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin

    dependent diabetes mellitus. The Diabetes Control and Complications Trial Research Group. N Engl J Med.

    1993;329:977-986.

    77. Ismail-Beigi F, Craven T, Banerji MA, et al. Effect of intensive treatment of hyperglycaemia on microvascular

    outcomes in type 2 diabetes: an analysis of the ACCORD randomised trial. Lancet. 2010;376:419-430.

    78. Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes

    (UKPDS 34). UK Prospective Diabetes Study (UKPDS) Group. Lancet. 1998;352:854-865.

    79. Kelly TN, Bazzano LA, Fonseca VA, et al. Systematic Review: Glucose Control and Cardiovascular Disease in Type

    2 Diabetes.Ann Intern Med. 2009;151:394-403.

    80. Ban K, Noyan-Ashraf MH, Hoefer J, et al. Cardioprotective and vasodilatory actions of glucagon-like peptide 1 recepto

    are mediated through both glucagon-like peptide 1 receptor-dependent and -independent pathways. Circulation.

    2008;117:2340-2350.

    81. Bose AK, Mocanu MM, Carr RD, et al. Glucagon-like peptide 1 can directly protect the heart against

    ischemia/reperfusion injury. Diabetes. 2005;54:146-151.

    82. Nikolaidis LA, Elahi D, Hentosz T, et al. Recombinant glucagon-like peptide-1 increases myocardial glucose uptake

    and improves left ventricular performance in conscious dogs with pacing-induced dilated cardiomyopathy. Circulation.

    2004;110:955-961.

    83. Sonne DP, Engstrom T, Treiman M. Protective effects of GLP-1 analogues exendin-4 and GLP-1(9-36) amide against

    ischemia-reperfusion injury in rat heart. Regul Pept. 2008;146:243-249.

    84. Timmers L, Henriques JP, de Kleijn DP, et al. Exenatide reduces infarct size and improves cardiac function in a

    porcine model of ischemia and reperfusion injury. J Am Coll Cardiol. 2009;53:501-510.

    85. Zhao T, Parikh P, Bhashyam S, et al. Direct effects of glucagon-like peptide-1 on myocardial contractility and glucoseuptake in normal and postischemic isolated rat hearts. J Pharmacol Exp Ther. 2006;317:1106-1113.

    86. Basu A, Charkoudian N, Schrage W, et al. Beneficial effects of GLP-1 on endothelial function in humans: dampening

    by glyburide but not by glimepiride.Am J Physiol Endocrinol Metab. 2007;293:E1289-1295.

    87. DeFronzo RA, Ratner RE, Han J, et al. Effects of exenatide (exendin-4) on glycemic control and weight over 30 week

    in metformin-treated patients with type 2 diabetes. Diabetes Care. 2005;28:1092-1100.

    88. Buse JB, Henry RR, Han J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in sulfonylurea-

    treated patients with type 2 diabetes. Diabetes Care. 2004;27:2628-2635.

    89. 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;28:1083-1091.

    90. 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;143:559-569.91. 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 noninferiority trial. Clin Ther. 2007;29:2333-2348.

    92. 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;50:259-267.

    93. Vilsboll T, Zdravkovic M, Le-Thi T, et al. Liraglutide, a long-acting human glucagon-like peptide-1 analog, given as

    monotherapy significantly improves glycemic control and lowers body weight without risk of hypoglycemia in patients

    with type 2 diabetes. Diabetes Care. 2007;30:1608-1610.

    94. Zinman B, Gerich J, Buse JB, et al. 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

  • 7/29/2019 Evolving Treatment for Patients With Type 2 Diabetes_ Current Guidelines and Emerging Therapeutic Decision-Mak

    25/25

    3/13h Type 2 Diabetes: Current Guidelines and Emerging Therapeutic De

    2009;32:1224-1230.

    95. Bergenstal R, Kim T, Yan P, et al. Exenatide once weekly improved cardiometabolic risk factors in subjects with type

    2 diabetes during one year of treatment. Presented at: American Diabetes Association 69th Scientific Sessions; June

    5-9, 2009; New Orleans, LA. Abstract 165-OR.

    96. Noyan-Ashraf MH, Momen MA, Ban K, et al. GLP-1R agonist liraglutide activates cytoprotective pathways and

    improves outcomes after experimental myocardial infarction in mice. Diabetes. 2009;58:975-983.

    97. Sokos GG, Nikolaidis LA, Mankad S, et al. Glucagon-like peptide-1 infusion improves left ventricular ejection fraction

    and functional status in patients with chronic heart failure. J Card Fail. 2006;12:694-699.

    98. Wolf R, Frederich R, Fiedorek F, et al. Evaluation of CV risk in the saxagliptin clinical trials. Presented at: 69th

    Scientific Sessions of the American Diabetes Association; June 5-9, 2009; New Orleans, LA. Late Breaking Abstract

    Handout. 2009;8-LB:LB8.

    99. Kazory A, Walsh K, Harman E, et al. Is metformin safe in patients with mild renal insufficiency? Diabetes Care.

    2007;30:444.

    100. Nesto RW, Bell D, Bonow RO, et al. Thiazolidinedione use, fluid retention, and congestive heart failure. Circulation.

    2003;108:2941-2948.

    Disclaimer

    The material presented here does not necessarily reflect the views of Medscape, LLC, or companies that support educationa

    programming on medscape.org. These materials may discuss therapeutic products that have not been approved by the US

    Food and Drug Administration and off-label uses of approved products. A qualified healthcare professional should be

    consulted before using any therapeutic product discussed. Readers should verify all information and data before treatingpatients or employing any therapies described in this educational activity.

    2012 Rockpointe

    This article is a CME certified activity. To earn credit for this activity visit:

    http://www.medscape.org/viewarticle/775099

    http://www.medscape.org/viewarticle/775099