154
Views
21
CrossRef citations to date
0
Altmetric
Review

Pharmacology, efficacy and safety of liraglutide in the management of type 2 diabetes

, , , &
Pages 215-226 | Published online: 14 Jul 2010

Abstract

Liraglutide is a glucagon-like peptide-1 analog with pharmacokinetic properties suitable for once-daily administration approved by the Food and Drug Administration for the treatment of patients with type 2 diabetes. Clinical trial data from large, controlled studies demonstrate the safety and efficacy of liraglutide in terms of hemoglobin A1c (HbA1c) reduction, reductions in body weight, and the drug’s low risk for hypoglycemic events when used as monotherapy. Liraglutide has been studied as monotherapy and in combination with metformin, glimepiride, and rosiglitazone for the treatment of type 2 diabetes. Additionally, comparative data with insulin glargine and exenatide therapy are available from Phase III trials. Once-daily administration may provide a therapeutic advantage for liraglutide over twice-daily exenatide, with similar improvements in HbA1c and body weight observed when liraglutide was compared with exenatide. The glucose-dependent mechanism of insulin release with incretin analog therapy holds potential clinical significance in the management of postprandial hyperglycemic excursions, with minimal risk of hypoglycemia when used with non-secretagogue medications. Data to date on patient-reported outcomes with liraglutide treatment are encouraging. The most common adverse events associated with liraglutide therapy are dose-dependent nausea, vomiting, and diarrhea. Diligent postmarketing surveillance to elucidate the risk of pancreatitis and medullary thyroid carcinoma in a heterogeneous population are likely warranted.

Introduction

The burden of diabetes continues to grow, both globally to more than 220 million people worldwide with diabetesCitation1 and in the US, where more than 23.6 million people have the disease.Citation2 Although numerous interventions and medications exist to treat diabetes, less than half of adults in the US with diabetes are able to reach the target glycosylated hemoglobin A1c (HbA1c) level, as set by the American Diabetes Association (ADA), of less than 7% for most patients.Citation3,Citation4 Attaining and maintaining glycemic control in type 2 diabetes mellitus (T2DM) is complicated by disease progression and continued β-cell deterioration.Citation5 Benefits of intensive glucose control include a reduction in microvascular complications, as well as the so-called “legacy effect”. This effect refers to the results of a 10-year follow-up study to the United Kingdom Prospective Diabetes Study, which found that intensive glucose control in newly diagnosed T2DM patients provided long-term benefits on cardiovascular outcomes and mortality, even if intensive control was not sustained in the long term.Citation6 Considering these findings, and that 18% of patients developed a diabetes-related complication within 6 years of diagnosis in the United Kingdom Prospective Diabetes Study,Citation7 it is clear that glycemic control starting at the time of T2DM diagnosis is important.

While lifestyle modifications, including diet and exercise, were once the initial treatment for patients with T2DM, it is now recognized that these interventions are insufficient for most patients, and pharmacotherapy should not be delayed.Citation8 Thus, the initial management of a patient presenting with T2DM consists of both lifestyle modification and medication, most specifically metformin, as recommended by the ADA.Citation9 While monotherapy may suffice in the short term, most patients will need polypharmacy to achieve and sustain glycemic control.Citation5 The ADA recommends initial combination therapy in newly diagnosed patients with an HbA1c of >8.5%,Citation9 while a consensus panel for the American Association of Clinical Endocrinologists (AACE) and the American College of Endocrinology (ACE) is even more aggressive, recommending dual therapy for patients with an HbA1c between 7.6% and 9%, and triple therapy or insulin for those with an initial HbA1c of >9%.Citation8

Because the choice of initial therapy and adjunctive therapy for intensification is increasingly individualized to the patient, agents that were once viewed solely as add-on therapy are now being considered much earlier in the course of treatment. Glucagon-like peptide-1 (GLP-1) agonists and dipeptidyl peptidase-4 (DPP-4) inhibitors are examples of such agents. The AACE/ACE diabetes algorithm recommends monotherapy for patients with an initial HbA1c of 6.5%–7.5%.Citation8 While metformin is the preferred initial agent, GLP-1 receptor agonists, DPP-4 inhibitors, thiazolidinediones, and alpha-glucosidase inhibitors are included as alternatives.Citation8 For patients requiring dual or triple therapy, the AACE/ACE preferentially recommends the addition of a GLP-1 agonist or a DPP-4 inhibitor, citing their efficacy and safety profiles, over thiazolidinediones or sulfonylureas.Citation8 Following initial treatment with metformin, the ADA, in contrast, recommends intensification with either basal insulin or a sulfonylurea (both Tier 1, or well validated therapies), pioglitazone or a GLP-1 agonist (both Tier 2, or less well validated therapies).Citation9 Not only have such treatment algorithms changed in recent years to recommend GLP-1 agonists earlier, but package labeling is also changing to expand their use. Exenatide, the GLP-1 agonist approved by the Food and Drug Administration (FDA) in 2005 for combination therapy, received an indication for monotherapy in 2009.Citation10

This relatively new class of GLP-1 agonists has gained increasing use for a variety of reasons. Agents which mimic the incretin system, such as GLP-1 agonists, have a low incidence of hypoglycemia, often cause weight loss, and may preserve β-cells or even stimulate their proliferation.Citation11,Citation12 In addition to the approval of exenatide, liraglutide was approved by the FDA in January 2010.Citation13 This paper will provide an overview of liraglutide and attempt to compare this new incretin analog with exenatide in terms of efficacy, safety, and utility in the treatment of patients with T2DM.

Methods

A MEDLINE search (1966 – February 2010) was conducted with the key words “liraglutide” and “incretin therapies” for clinical trials and pertinent review articles published in English. References of identified articles were searched for additional relevant sources. Abstracts from the ADA and European Association for the Study of Diabetes annual meetings presented in 2006, 2007, 2008, and 2009 were also searched for relevant data. English language articles pertinent to the pharmacology, pharmacokinetics, efficacy, safety, and patient-related outcomes of liraglutide treatment were reviewed. Six Phase III clinical trials from the Liraglutide Effects and Action in Diabetes (LEAD) program have been published. Reports on patient-reported outcomes and quality of life measures have also been published and are discussed herein.

Pharmacology

A role for an intestinal mediator of insulin secretion was initially conceived by the observation that the oral intake of glucose resulted in a greater insulin response when compared with intravenous glucose administration.Citation14,Citation15 This “incretin effect” is now known to be due to the stimulation of insulin release by the oral intake of nutrients which results in insulin secretion above and beyond the insulin release induced by increased blood glucose concentrations alone. The incretin effect is now recognized as being responsible for approximately 60% of the insulin response to a given meal.Citation16 Of clinical significance, the incretin effect has been shown to be greatly impaired in patients with T2DM.Citation17 The incretin effect is primarily attributed to 2 insulinotropic gut hormones, ie, GLP-1 and glucose-dependent insulinotropic polypeptide (GIP). GLP-1 is a 30-amino acid peptide released from L-cells of the intestine in response to a meal,Citation18 and GIP is released by duodenal cells of the proximal small bowel.Citation19 GLP-1 secretion is known to be deficient in patients with T2DM,Citation20,Citation21 and GLP-1 infusion has been shown experimentally to lower glucose levels via enhanced glucose-dependent insulin secretion in subjects with T2DM.Citation22Citation24 Further study with GLP-1 in subjects with T2DM has demonstrated beneficial effects of GLP-1 on decreasing inappropriate glucagon secretion,Citation25,Citation26 slowing of gastric emptying,Citation27,Citation28 and increased satiety and decreased food intake.Citation29

While GLP-1 has demonstrated clinical utility in the management of patients with T2DM, endogenous GLP-1 is rapidly degraded by the enzyme DPP-4, resulting in a GLP-1 half-life of approximately 1–2 minutes.Citation30 The development of DPP-4 resistant GLP-1 analogs has been one strategy by which to utilize the beneficial effects of GLP-1 in patients with T2DM. Liraglutide is the newest incretin analog currently available in the US, which is approved for once-daily administration.Citation13 In vitro studies indicate that liraglutide retains affinity for GLP-1 receptors despite these structural modifications.Citation31 The addition of the C16 acyl chain allows for noncovalent binding to albumin, both hindering DPP-4 access to the molecule and contributing to a prolonged half- life and duration of action.Citation32

Pharmacokinetics

The structure of liraglutide makes it kinetically unique when compared with the related compound exenatide; liraglutide incorporates a palmitate side chain at position 26 using a γ-glutamic acid spacer.Citation33,Citation34 This change allows for 99% albumin binding when compared with natural GLP-1, allowing liraglutide to escape glomerular filtration and extend its duration of action. Liraglutide is detected in the urine and feces as metabolites, and is hepatically metabolized and eliminated via the liver and kidneys.

The pharmacokinetic profile of liraglutide makes it a desirable agent for the treatment of T2DM, given the extended time to maximum plasma concentration (Tmax) and half-life (t1/2). In an initial study performed by Elbrond et al, 72 healthy male subjects received 8 consecutive subcutaneous doses (1.25–20.0 μg/kg per dose) of liraglutide.Citation35 Results from this study reported a Tmax of 9–12 hours after dosing, and a plasma t1/2 of elimination of 11–15 hours.Citation35 In a study of 30 healthy male subjects receiving 5 consecutive subcutaneous doses (1.25–12.5 μg/kg per dose) of liraglutide, the reported Tmax was 10–14 hours, while the plasma t1/2 of elimination was 11–13 hours.Citation34 Further kinetic studies were performed utilizing liraglutide in a multi-day fashion with different administration times. One study was performed in 11 subjects with T2DM, administering 10 μg/kg subcutaneously once daily at bedtime for 1 day.Citation36 The Tmax was found to be 10–14 hours, consistent with previous studies, while the plasma t1/2 was found to be 6–14 hours. A second study was performed with liraglutide, administering 6 μg/kg subcutaneously every morning over a 7-day period in 13 T2DM subjects.Citation37 Study results reported a Tmax of 7.1–13.1 hours, and a steady-state plasma t1/2 of 17.9 hours.

Liraglutide was also examined in clinical trials to determine if normal dosing pharmacokinetics would be impacted in both renally and hepatically impaired subjects. Jacobsen et al performed a study in 30 subjects, comprising 24 with varying degrees of renal impairment and 6 healthy subjects, in which 0.75 mg of liraglutide was administered subcutaneously, with 72-hour follow-up blood sampling.Citation38 Results from this study found that liraglutide did not adversely impact serum creatinine in mild-to-severe renal impairment, and was not associated with an increased risk of adverse events in this study population. A meta-analysis was performed examining the Phase III LEAD studies, looking at the impact of liraglutide on serum creatinine levels.Citation39 When compared with normal subjects, no significant change in serum creatinine occurred with either 1.2 mg daily or 1.8 mg daily dosing; this is considered to be due to the modification of liraglutide’s chemical structure when compared with natural GLP-1. Liraglutide was also evaluated in 24 subjects with mild, moderate, severe, or no hepatic impairment.Citation40 Subjects were administered 0.75 mg of liraglutide as a single dose, and were evaluated after a 72-hour period to determine if hepatic impairment influenced liraglutide’s kinetic and safety profile. After both renal and hepatic evaluations, it was concluded by the researchers that no hepatic or renal dosing adjustments are necessary with liraglutide.

When compared with exenatide, there are several differences that may be advantageous when considering the use of liraglutide. Exenatide was directly compared with liraglutide in the LEAD-6 trial to determine the efficacy and safety of each agent.Citation41 Subjects were administered either 1.8 mg/day of liraglutide (202 subjects) or 10 μg twice daily of exenatide (187 subjects) for a period of 26 weeks. It was found that liraglutide maintained steady-state plasma levels 24 hours after administration, while exenatide peaked and returned to baseline plasma levels 10–12 hours following administration. Liraglutide was also found to have minimal impact on renal function due to its chemical structure, while exenatide is primarily eliminated through the kidney, and is not recommended for use in severe renal impairment or end-stage renal disease.Citation1

Clinical trials

The LEAD program comprises 6 randomized, controlled, double-blind Phase III clinical studies in participants with T2DM inadequately controlled with lifestyle and dietary interventions or oral antidiabetic therapies. provides a summary of select efficacy endpoints reported from the six LEAD studies discussed individually below.Citation42Citation47

Table 1 Select efficacy outcomes from the Liraglutide Effects and Action in Diabetes (LEAD) trials

Liraglutide versus rosiglitazone as add-on to baseline glimepiride

LEAD-1 was a 26-week, randomized, double-dummy trial in 1041 patients with T2DM. The objective of the study was to compare the addition of liraglutide to glimepiride therapy with glimepiride monotherapy or the addition of rosiglitazone to baseline glimepiride.Citation42 Participants had a mean baseline HbA1c of 8.4% and a mean age of 56.1 years. Participants received liraglutide 0.6 mg/day, 1.2 mg/day, or 1.8 mg/day in combination with glimepiride, placebo plus glimepiride (2–4 mg/day), or rosiglitazone 4 mg/day plus glimepiride. Mean HbA1c was reduced by −1.08% and −1.13% with liraglutide 1.2 mg and 1.8 mg, respectively. Participants receiving rosiglitazone experienced a mean HbA1c reduction of −0.44%, and glimepiride monotherapy resulted in a mean HbA1c increase of 0.23% (P < 0.0001). Of those treated with liraglutide 1.2 mg plus glimepiride, 22% reached an HbA1c less than 6.5%, with 21% reaching an HbA1c less than 6.5% with liraglutide 1.8 mg plus glimepiride. In contrast, 4% of those on glimepiride monotherapy and 10% of subjects treated with rosiglitazone plus glimepiride reached an HbA1c below 6.5% (P < 0.0003).

Liraglutide versus glimepiride as add-on to baseline metformin

LEAD-2 was a randomized, double-blind study that enrolled 1091 participants with T2DM.Citation43 Participants had a mean baseline HbA1c of 8.4%. Participants received liraglutide 0.6 mg, 1.2 mg, or 1.8 mg once daily added to metformin 1 g twice daily, placebo plus metformin, or glimepiride 4 mg/ day added to metformin. Mean HbA1c reductions of −0.7%, −1.0%, and −1.0% were observed with liraglutide 0.6 mg, 1.2 mg, and 1.8 mg in combination with metformin, respectively. Those receiving metformin monotherapy experienced a mean HbA1c increase of 0.1%, with a decrease of −1.0% seen in those receiving glimepiride plus metformin (P < 0.05 versus liraglutide plus metformin versus placebo plus metformin). Weight loss was achieved in all participants receiving liraglutide, compared with a 1.0 kg weight gain observed in those receiving glimepiride (P < 0.0001 for all liraglutide doses when compared with glimepiride). The percentage of patients achieving an HbA1c less than 6.5% was 11.3% in the liraglutide 0.6 mg plus metformin group, 19.8% in the liraglutide1.2 mg plus metformin group, and 24.6% in the liraglutide 1.8 mg plus metformin group, compared with 4.2% of those treated with placebo plus metformin, and 22.2% of those treated with glimepiride plus metformin (P < 0.02 for all liraglutide doses when compared with placebo).

Liraglutide versus glimepiride as monotherapy

The LEAD-3 study enrolled 746 patients with T2DM and a mean baseline HbA1c of 8.2%.Citation44 This Phase III, double-blind, parallel-treatment study involved a head-to-head comparison of monotherapy with 1.2 mg/day or 1.8 mg/day of liraglutide or glimepiride 8 mg/day. Any previous oral antidiabetic drugs (up to half the maximal dose) were discontinued at randomization prior to study drug initiation. At 52 weeks of therapy, mean HbA1c reductions from baseline of −0.84% (P = 0.0014 versus glimepiride) and −1.4% (P < 0.0001 versus glimepiride) were seen with liraglutide 1.2 mg and 1.8 mg, compared with a reduction of −0.51% for glimepiride. Twenty-seven percent of patients on liraglutide 1.8 mg and 16% of patients on glimepiride attained an HbA1c less than 6.5%. Decreases in body weight observed were −2.1 kg and −2.5 kg for liraglutide 1.2 mg and 1.8 mg, respectively (P = 0.0001 versus glimepiride for both doses). In contrast, participants receiving glimepiride experienced an average weight gain of 1.1 kg.

Liraglutide as add-on to baseline metformin and rosiglitazone

LEAD-4 was a 26-week, placebo-controlled trial enrolling 533 patients with T2DM and a mean baseline HbA1c of 8.3%.Citation45 LEAD-4 assessed the effect of adding liraglutide 1.2 mg or 1.8 mg to baseline metformin 1 g twice daily plus rosiglitazone 8 mg/day. Liraglutide addition resulted in mean HbA1c reductions of −1.48% for both liraglutide doses compared with −0.54% observed with the addition of placebo (P = 0.0001). An HbA1c less than 6.5% was achieved in 35% and 37% of patients receiving liraglutide 1.2 mg and 1.8 mg, respectively. Liraglutide 1.2 mg and 1.8 mg treatment resulted in reductions in fasting plasma glucose (−40 mg/dL and −43 mg/dL, respectively) and postprandial glucose levels (−49 mg/dL and −47 mg/dL, respectively). Patients receiving placebo experienced a mean increase in body weight of 0.6 kg compared with a mean weight loss of −1.0 kg and −2.0 kg for liraglutide 1.2 and 1.8 mg (P < 0.05 versus placebo for both liraglutide doses).

Liraglutide versus glargine as add-on to baseline metformin and glimepiride

LEAD-5 aimed to compare liraglutide with insulin glargine as add-on therapy to metformin and glimepiride.Citation46 LEAD-5 enrolled a total of 581 patients with T2DM with a mean baseline HbA1c of 8.2%. Participants received liraglutide 1.8 mg/day, liraglutide placebo, or insulin glargine in addition to metformin 1 g twice daily and glimepiride (2–4 mg/day) for a duration of 26 weeks. The dose of insulin glargine was individually titrated according to a patient-driven algorithm, with a mean dose of 24 units per day reported at the end of the trial in the insulin glargine arm. Mean HbA1c values were decreased −1.33%, −0.24%, and −1.09% with the addition of liraglutide, placebo, and insulin glargine, respectively (P < 0.05 for liraglutide versus placebo and insulin glargine). An HbA1c below 6.5% was achieved in 37.1% of patients treated with liraglutide, 10.9% of those treated with placebo (P < 0.0001 versus liraglutide), and 23.6% of patients in the insulin glargine group (P = 0.0001 versus liraglutide). A mean body weight reduction of −1.81 kg was reported in the liraglutide group (P < 0.0001 versus glargine; P = 0.0001 versus placebo), with a mean weight loss of −0.42 kg in the placebo group, and a 1.62 kg weight gain seen in the insulin glargine group.

Liraglutide versus exenatide as add-on to baseline metformin and/or a sulfonylurea

LEAD-6 was a 26-week trial in 464 patients inadequately treated with metformin and/or a sulfonylurea with a mean baseline HbA1c of 8.2%.Citation41 This trial aimed to compare liraglutide 1.8 mg/day to exenatide 10 μg twice daily as add-on therapy. HbA1c reductions of −1.12% for liraglutide and −0.79% for exenatide were observed (P < 0.0001). A target HbA1c less than 6.5% was achieved in 35% of those treated with liraglutide versus 21% for patients receiving exenatide (P < 0.0001). Changes in body weight were similar in both groups with no statistical differences in weight change between the liraglutide and exenatide treatment groups. Weight reductions of −3.2 kg and −2.9 kg for liraglutide and exenatide were observed, respectively.

Additional clinical endpoints

Clinical studies with liraglutide have also demonstrated potential benefits of therapy on β-cell function and the cardiovascular system. One study utilized a graded glucose protocol to assess the effects of a single liraglutide dose of 7.5 μg/kg on insulin secretion.Citation47 Insulin secretion increased with elevation in blood glucose in all groups, however liraglutide treatment resulted in a more pronounced insulin response which was similar to that observed in healthy control subjects. Other clinical studies have shown improvements in glucose-induced insulin secretion, β-cell sensitivity, and suppression of 24-hour glucagon secretion following 1 week of therapy,Citation37 and sustained β-cell sensitivity to glucose over 12 weeks of therapy.Citation48 Regarding the effects of liraglutide treatment on cardiovascular health, statistically significant decreases in systolic blood pressure, ranging from 2 to 7.9 mmHg, have been observed in clinical trials.Citation43,Citation44,Citation49 The mechanism resulting in the observed reduction in systolic blood pressure is unknown, but appears unrelated to concomitant weight loss.Citation50 Early data additionally indicates liraglutide treatment may also decrease cardiovascular markers such as PAI-1 and BNP, but the clinical implications, either positive or detrimental, of these findings are unknown.

Safety and tolerability

Hypoglycemia

Of paramount importance in any newly approved drug used to treat T2DM is the likelihood of hypoglycemia, when used as mono-, dual-, or even as part of a triple-therapy regimen. In the LEAD trials, a minor hypoglycemic event was defined as a plasma glucose concentration of <56 mg/dL that was resolved with self-treatment, and a major hypoglycemic event was defined as a hypoglycemic event requiring third party assistance. Note that in LEAD-1, self-treatment was the sole criteria used to classify a hypoglycemic event as minor.Citation42 As expected, trials utilizing combination sulfonylurea therapy resulted in the highest incidence of hypoglycemia. LEAD-1 involved patients on concurrent sulfonylurea therapy (glimepiride).Citation42 In this trial, the percentage of patients experiencing minor hypoglycemia was reported as glimepiride monotherapy (placebo) 2.6%, 0.17 events/subject-year; liraglutide 0.6 mg, 5.2%, 0.17 events/subject-year; liraglutide 1.2 mg 9.2%, 0.51 events/subject-year; liraglutide 1.8mg, 8.1%, 0.47 events/subject-year; and rosiglitazone 4.3%, 0.12 events/subject-year. provides a summary of adverse event findings from the LEAD program. Furthermore, in the 1.8 mg liraglutide plus glimepiride cohort, one major hypoglycemic event occurred.Citation42

Table 2 Adverse drug event rates in Liraglutide Effects and Action in Diabetes (LEAD) trials

In LEAD-2, the percentage of patients experiencing minor hypoglycemia was low in the placebo and liraglutide cohorts, roughly 3%, while 17% of subjects receiving glimepiride reported an incidence of minor hypoglycemia.Citation43 Major hypoglycemic events did not occur in LEAD-2. There were a low percentage of patients reporting minor hypoglycemic events in LEAD-3, with 8% of the subjects in the liraglutide 1.8 mg group (0.25 events per year) and 12% of subjects in the 1.2 mg group experiencing minor hypoglycemic events (0.3 events per year).Citation44 Twenty-four percent of subjects in the glimepiride group reported minor hypoglycemic events while no major hypoglycemic events occurred in any of the groups in this 52-week trial.

Minor hypoglycemia was reported in LEAD-4 at rates of 9%, 7.9%, and 5.1% of subjects in the 1.2 mg liraglutide, 1.8 mg liraglutide, and placebo groups, respectively.Citation45 No major hypoglycemic events were reported. In LEAD-5, 27.4% of the patients receiving liraglutide experienced at least 1 episode of minor hypoglycemia (1.2 events/subject/year), while 28.9% (1.3 events/subject/year) and 16.7% (1.0 events/subject/year) experienced minor hypoglycemia in the insulin glargine and placebo groups, respectively.Citation46 In the liraglutide group, 5 subjects reported a major hypoglycemic episode, with no major hypoglycemic events reported in the other two cohorts.

In the open-label 26-week trial known as LEAD-6, minor hypoglycemia occurred in 26% and 34% of the liraglutide and exenatide groups, respectively; correlating to event rates of 1.9 (liraglutide) and 2.6 (exenatide) events per subject per year.Citation41 Two cases of major hypoglycemia occurred in subjects exposed to exenatide and a sulfonylurea.

Gastrointestinal adverse events

In the LEAD-1 trial, incomplete data were provided for all arms of the study in regard to gastrointestinal (GI) side effects.Citation42 The most complete data reveal that nausea was highest in the liraglutide 1.2 mg cohort (10.5%) compared with an event rate of 1.8% in the placebo group. Additionally, 4.4% and 7.9% of those in the 1.2 mg liraglutide group experienced vomiting and diarrhea, respectively.

The LEAD-2 trial involved subjects on a wide range of oral therapies that included metformin, sulfonylureas, repaglinide, or some combination of these listed medications.Citation43 GI adverse events were pronounced in this trial, with 35% of the subjects in the 0.6 mg liraglutide group experiencing GI side effects, including nausea, vomiting, and diarrhea, with 40% and 44% experiencing these GI-related adverse events in the 1.2 mg and 1.8 mg groups, respectively. This is in comparison with 17% of the placebo-treated group reporting GI-associated adverse events. Five percent (36 subjects) of those receiving any dose of liraglutide withdrew from the study due to GI-related adverse events.

In LEAD-3, nausea occurred in 27.5%, 29.3%, and 8.5% of participants in the 1.2 mg liraglutide, 1.8 mg liraglutide, and glimepiride groups, respectively.Citation44 Vomiting occurred in 3.6% of subjects receiving glimepiride, while this side effect occurred in 9.3% and 12.4% of those receiving 1.2 mg and 1.8 mg of liraglutide, respectively. Diarrhea was reported by 15.5%, 18.7%, and 8.9% of participants in the 1.2 mg liraglutide, 1.8 mg liraglutide, and glimepiride groups, respectively. A total of 6 participants (1.2%) receiving liraglutide withdrew from the study due to vomiting, while a total of 17 participants (3.4%) receiving liraglutide withdrew for any GI-related complaint. In the 26-week study known as LEAD-4, 29% of subjects receiving 1.2 mg liraglutide experienced nausea while 40% of subjects in the 1.8 mg liraglutide group reported this adverse event.Citation45 Vomiting was reported by 7% and 17% of subjects in the 1.2 mg and 1.8 mg liraglutide groups, respectively. When all GI adverse events were grouped (nausea, vomiting, diarrhea), 19% of those receiving placebo and 45% and 56% of those receiving 1.2 mg and 1.8 mg of liraglutide, respectively, reported GI-related complaints. GI adverse events contributed to 5 withdrawals (3% of participants) in the liraglutide 1.2 mg group and 19 (10.7% of participants) in the liraglutide 1.8 mg group.

Nausea occurred in 13.9% of those receiving liraglutide, 3.5% in placebo arm, and 1.3% of participants in the insulin glargine group in LEAD-5.Citation46 In this trial, diarrhea was reported in 10%, 5.3%, and 1.3% in the liraglutide, placebo, and insulin glargine groups, respectively. Vomiting occurred in 6.5%, 3.5%, and 0.4% in the liraglutide, placebo, and insulin glargine groups, respectively. Dyspepsia was also reported in this trial, with 6.5%, 0.9%, and 1.7% of subjects in the liraglutide, placebo, and insulin glargine groups, respectively, experiencing this GI-related adverse event. Four subjects in LEAD-5 receiving liraglutide withdrew from the study due to GI-related adverse events. LEAD-6 reported similar rates of GI-related adverse events between the liraglutide and exenatide groups.Citation41 Overall, GI adverse events occurred in 45.5% and 42.7% of liraglutide- and exenatide-treated subjects, respectively, with nausea being the most frequently reported event. Nausea tended to resolve over time with both therapies, however, with 2.5% of the liraglutide group reporting nausea at week 26 compared with 15.8% of those receiving exenatide therapy.Citation41 Vomiting occurred in 6.0% and 9.9% of the liraglutide and exenatide groups, respectively.

From the above data it can be seen that nausea is a frequent adverse event in subjects receiving liraglutide. However, nausea was most pronounced in the first 4 weeks of therapy, with symptoms generally dissipating over the remainder of the study period in all trials.

Pulse rate

The effects of liraglutide on pulse rate ranged from an increase of 2–4 beats per minute (bpm) in subjects receiving liraglutide (P ≤ 0.002 versus placebo; P < 0.01 versus rosiglitazone), with pulse increasing by a mean 1 bpm in subjects receiving rosiglitazone, and pulse decreasing by a mean 1 bpm in the placebo group.Citation42 In the LEAD-2 trial, pulse rates increased by a mean 2–3 bpm in those receiving liraglutide, compared with a 1 bpm increase in the glimepiride and placebo groups.Citation43 The mean pulse rate in LEAD-3 increased by 0.4, 3.2, and 1.6 bpm for the glimepiride group and the 1.2 mg and 1.8 mg liraglutide groups, respectively.Citation44 Pulse rate increased by 2 bpm in subjects receiving liraglutide in LEAD-4, with mean pulse rates increasing by 3 bpm in those receiving liraglutide 1.8 mg.Citation45 Pulse rate increased by a mean of 2.62 bpm in those subjects receiving liraglutide in LEAD-5, while increases of 0.08 bpm and 0.93 bpm were experienced by those in the insulin glargine and placebo groups, respectively.Citation46 In LEAD-6, heart rates increased by a mean of 3.28 bpm in the liraglutide group compared with 0.69 bpm in the exenatide group.Citation41

Pancreatitis

Pancreatitis has been reported in clinical trials with liraglutide. In LEAD-1, one subject receiving liraglutide 0.6 mg developed pancreatitis but successfully completed the trial.Citation42 Two subjects withdrew from the LEAD-2 study after developing pancreatitis, 1 receiving liraglutide and 1 receiving glimepiride.Citation43 1 participant in each of the liraglutide groups in LEAD-3 developed pancreatitis, with one completing the trial.Citation44 No cases of pancreatitis were reported in the 26-week LEAD-4 or LEAD-5 trials.Citation45,Citation46 Likewise, no cases of acute pancreatitis were reported in LEAD-6; however 1 case of mild pancreatitis occurred in a subject receiving liraglutide who subsequently completed the 26-week trial.Citation41

Anti-liraglutide antibodies

In LEAD-1, 9%–13% of subjects exposed to liraglutide during the 26-week trial developed anti-liraglutide antibodies. Citation42 The LEAD-2 and LEAD-3 trials did not measure the development of liraglutide antibody formation. In LEAD-4, 6 subjects in the liraglutide 1.2 mg group and 9 subjects in the liraglutide 1.8 mg group developed antibodies.Citation45 In LEAD-5, 23 subjects (9.8%) of subjects developed antiliraglutide antibodies during the 26-week study.Citation46 Because the LEAD-6 trial involves an extension phase where subjects may continue to take liraglutide, antibody determination will be completed once the trial is complete and after an appropriate washout period; these data are not currently available.Citation41 While the clinical impact of anti-liraglutide antibodies is unknown at this time, further study of this phenomenon is warranted.

Additional safety considerations

Study withdrawal rates due to adverse drug events and event rates for serious adverse events reported in the six LEAD trials are summarized in . Interestingly, injection-related adverse events such as injection site rash, were not reported in any of the LEAD trials. In addition to the adverse drug events discussed above, peripheral edema was reported in the LEAD-4 trial, likely due to participants also receiving concomitant rosiglitazone.Citation45 In this trial, peripheral edema occurred in 5.1%, 1.7%, and 8.0% of the subjects in the 1.2 mg liraglutide, 1.8 mg liraglutide, and placebo groups, respectively. The LEAD studies indicate liraglutide to be generally safe, however the development of rare adverse events is of concern until patients at heightened risk for developing events such as pancreatitis can be identified. The safety and tolerability of liraglutide can only truly be assessed with robust Phase IV post-marketing data involving long-term treatment with this novel therapy.

Table 3 Clinical study withdrawal and serious adverse event rates in Liraglutide Effects and Action in Diabetes (LEAD) trials

An additional theoretical concern raised in the liraglutide prescribing information is a warning regarding the observation of dose-dependent and treatment-duration-dependent thyroid C-cell tumors witnessed at clinically relevant exposures in rats and mice.Citation13 During clinical trials with liraglutide, calcitonin, a biomarker for the detection of medullary thyroid cancer, was monitored routinely.Citation51 During the LEAD program, increases in calcitonin levels did occur in a slightly higher percentage of patients treated with liraglutide when compared with controls, however, calcitonin levels remained within normal ranges.Citation51 Ultimately, while it is unknown if this is clinically relevant in humans, liraglutide is contraindicated in patients with a personal or family history of medullary thyroid carcinoma, and in patients with multiple endocrine neoplasia syndrome type 1.Citation13 The FDA has requested the establishment of a cancer registry to monitor the annual incidence of medullary thyroid cancer over the next 15 years.Citation51

Administration

Liraglutide was approved in July of 2009 by the European Commission for marketing consideration to all 27 European Union members.Citation52 Liraglutide is administered as a subcutaneous injection for once-daily treatment of T2DM, as an adjunct therapy in combination with metformin, a sulfonylurea, or metformin plus a sulfonylurea or thiazolidinedione. Citation53 The approved dosing for use in Europe is an initial dose of 0.6 mg daily for one week, with a recommended titration to 1.2 mg daily after the first week. The maximum recommended daily dose is 1.8 mg for patients who are not well controlled and who can tolerate the higher titrated dose, although doses as high as 2 mg daily have been used in clinical trials.Citation54 The LEAD studies found that titrating liraglutide by 0.6 mg weekly improved the tolerability and reduced the occurrence of GI adverse events.Citation43 Injection site reactions are another concern, and should be monitored for during liraglutide initiation.Citation13 Liraglutide received FDA approval for use in the US in January 2010 as an adjunct to diet and exercise to improve glycemic control in adults with T2DM.Citation13 Similar to Europe, in the US liraglutide is recommended at a starting dose of 0.6 mg daily for the first week, followed by an upward titration to 1.2 mg daily.Citation13 For those patients not achieving the desired glycemic control at the 1.2 mg dose, the dose can likewise be increased to 1.8 mg. As per the prescribing information, it is also recommended that a reduction in the dose of pre-existing insulin secretagogue medications be considered when initiating liraglutide to minimize the risk of treatment-emergent hypoglycemia.Citation13 Liraglutide is available commercially in the US as a simple pen capable of administering 0.6, 1.2, and 1.8 mg doses.

Patient-specific considerations

A common problem among T2DM patients is the issue of weight management. Elevated body weight or obesity, and an increased risk of cardiovascular-related complications often increase the medical burden and medication load of the patient.Citation55 The patient is often prescribed a medication regimen that is counterproductive to weight loss, which decreases treatment satisfaction due to resulting weight gain. Newer GLP-1 agonists, such as exenatide and liraglutide, are associated with weight reduction in healthy and diabetic subjects.Citation41 A comparative trial of exenatide and liraglutide in T2DM subjects concluded that both exenatide and liraglutide were associated with a significant reduction in body weight when compared with baseline (−2.87 kg and −3.24 kg, respectively).Citation41 In the LEAD-3 trial, weight loss was reported in both the 1.2 mg and 1.8 mg liraglutide monotherapy groups, with an average loss of −2 kg and −2.5 kg, respectively (P < 0.0001).Citation44 Reported weight loss occurred during the first 16 weeks, but was sustained throughout the remaining 36 weeks of treatment. After completion of a second 52-week open-label study period, weight reductions of −2.1 kg and −2.7 kg for both the 1.2 mg and 1.8 mg liraglutide study groups, respectively, were significant when compared with those receiving glimepiride (P < 0.0001).Citation44 In all LEAD studies completed, liraglutide 1.8 mg daily was associated with reported weight reductions of −0.2 kg to −3.24 kg over a period of at least 6 months.Citation41Citation46 Of additional interest, an analysis of patients from LEAD-1 and LEAD-2 reported that weight reductions in patients receiving liraglutide were primarily due to reductions in fat mass rather than lean tissue mass.Citation56 Finally, a trial enrolling healthy, nondiabetic patients with a mean baseline BMI of 30–40kg/m2 compared the weight effects of liraglutide at doses of 1.2, 1.8, 2.4, and 3 mg daily, versus orlistat 120 mg three times daily or placebo.Citation57 Weight loss in liraglutide subjects was −4.8 kg in the 1.2 mg group (P = 0.003), −5.5 kg in the 1.8 mg group (P < 0.0001), −6.3 kg in the 2.4 mg group (P < 0.0001), and −7.2 kg in the 3.0 mg group (P < 0.0001) when compared with baseline.

Quality of life was another indicator of treatment outcome that was evaluated in select clinical trials. Astrup et al found that mean physical function improved in the liraglutide 3.0 mg group by a score of 6.8 (P = 0.001) when compared with placebo, and by 6.0 when compared with the orlistat treatment group (P = 0.006).Citation57 Mean self-esteem also increased in the 3.0 mg daily group by a score of 9.6 when compared with placebo (P = 0.0001), and by 6.2 when compared with the orlistat treatment group (P = 0.04). Patient-reported outcomes were also investigated in the LEAD-3 trial. Compared with glimepiride, the liraglutide 1.8 mg cohort reported a mean decrease in BMI that was associated with improvements in both weight image and weight concern (P < 0.0001).Citation44 Decreases in weight concern were associated with increases in overall quality of life, general perception of their health (both P < 0.0001), and mental/emotional health (P = 0.002). Finally, in LEAD-6, subjects were assessed for treatment satisfaction using the Diabetes Treatment Satisfaction Questionnaire. Citation41 Overall treatment satisfaction was reported to be significantly higher in the liraglutide group when compared with the exenatide group (P = 0.0004).

Future studies regarding adherence and continued impact on patient quality of life would be of value to the clinical community, with the unique kinetic parameters of liraglutide, as well as its positive impact on weight, lending merit to liraglutide as a viable option for the treatment of T2DM.

Discussion

The pharmacokinetic profile of liraglutide is amenable to once-daily dosing, thus creating a potential advantage when compared with twice-daily exenatide. Drug regimen simplicity is an important clinical consideration, particularly in patients receiving multiple medications for the treatment of T2DM and related comorbidities. Patients often present with resistance to the initiation of an injectable agent, however the potential for weight loss with the incretin mimetics and incretin analogs can be a motivator for some patients. The most recent consensus algorithm released by the ADA and the European Association for the Study of Diabetes lists GLP-1 analogs as a treatment option for consideration as a Tier 2 agent, or “less well-validated therapy”, in T2DM patients.Citation9 The consensus guideline recommends consideration of GLP-1 agonist therapy in selected clinical situations. One situation in which GLP-1 agonist therapy could be considered is if weight loss is a major consideration and the patient’s HbA1c level is close to target (<8.0%).Citation9 The guideline warns, however, that GLP-1 agonist therapy is not indicated for all patients and should be used with caution in those with a history of significant GI disease, such as a diagnosis of gastroparesis, due to a possible exacerbation of such conditions with incretin mimetic therapy.Citation9 Because postprandial hyperglycemia affects HbA1c to a greater degree than fasting hyperglycemia, the closer a patient is to their HbA1c goal, GLP-1 agonists, such as liraglutide, provide a viable treatment option to target postprandial glucose excursions due to their glucose-dependent effects on insulin secretion.

Conclusion

Clinical trial data from large, controlled studies demonstrate the efficacy and safety of liraglutide in terms of HbA1c reduction, beneficial effects on body weight, and a low risk for hypoglycemic events when used as monotherapy. Liraglutide is relatively well tolerated, with dose-dependent nausea, vomiting, and diarrhea being the most commonly reported adverse events observed in clinical trials. Clinical trial data in humans indicate that liraglutide may have a role in the treatment of T2DM patients as monotherapy early in the disease process, as well as in combination with metformin, glimepiride, and rosiglitazone in patients inadequately controlled on oral antidiabetic drugs. Comparative data with exenatide twice-daily indicate a potential therapeutic advantage for liraglutide in terms of ease of use, with similar improvements in HbA1c and body weight seen when comparing these two agents. Data are currently not available comparing liraglutide with once-weekly exenatide currently under Phase III study, however. Questions do remain regarding the safety of this agent in terms of risk of pancreatitis and medullary thyroid carcinoma. While the risk of such events is assumed to be small, vigorous postmarketing surveillance and reporting is warranted to identify patients that may be at increased risk for experiencing such events.

Disclosure

The authors report no conflicts of interest in relation to the content or production of this article.

References

  • World Health Organization Diabetes Fact Sheet Available from: http://www.who.int/mediacentre/factsheets/fs312/en/index.html Accessed Dec 29, 2009
  • Centers for Disease Control and Prevention National Diabetes Fact Sheet: General Information and National Estimates on Diabetes in the United States, 2007 Atlanta, GA US Department of Health and Human Services, Centers for Disease Control and Prevention 2008
  • Ong KL Cheung BM Wong LY Prevalence, treatment and control of diagnosed diabetes in the U.S. National Health and Nutrition Examination Survey 1999–2004 Ann Epidemiol 2008 18 222 229 18201902
  • Resnick HE Foster GL Bardsley J Achievement of American Diabetes Association clinical practice recommendations among US adults with diabetes, 1999–2002: The National Health and Nutrition Examination Survey Diabetes Care 2006 29 531 537 16505501
  • Turner RC Cull CA Frighi V UK Prospective Diabetes Study (UKPDS) Group Glycemic control with diet, sulfonylurea, metformin, or insulin in patients with type 1 diabetes mellitus: Progressive requirement for multiple therapies (UKPDS 49) JAMA 1999 281 2005 2012 10359389
  • Holman RR Paul SK Bethel MA 10-year follow-up of intensive glucose control in type 1 diabetes N Engl J Med 2008 359 1577 1589 18784090
  • UK Prospective Diabetes Study Group UK Prospective Diabetes Study 16. Overview of 6 years’ therapy of type I diabetes: A progressive disease Diabetes 1995 44 1249 1258 7589820
  • Rodbard HW Jellinger PS Davidson JA Statement by an American Association of Clinical Endocrinologists/American College of Endocrinology consensus panel on type 1 diabetes mellitus: An algorithm for glycemic control Endocr Pract 2009 15 6 540 559 19858063
  • Nathan DM Buse JB Davidson MB Medical management of hyperglycemia in type 1 diabetes: A consensus algorithm for the initiation and adjustment of therapy Diabetes Care 2009 32 193 203 18945920
  • Byetta® (Package insert) San Diego, CA Amylin Pharmaceuticals, Inc 2009
  • Drucker DJ The biology of incretin hormones Cell Metab 2006 3 153 165 16517403
  • Farilla L Bulotta A Hirshberg B Glucagon-like peptide 1 inhibits cell apoptosis and improves glucose responsiveness of freshly isolated human islets Endocrinology 2003 144 5149 5158 12960095
  • Victoza® (Package insert) Princeton, NJ Novo Nordisk Inc 2010
  • Elrick H Stimmler L Hlad CJ Plasma insulin response to oral and intravenous glucose administration J Clin Endocrinol Metab 1964 24 1076 1082 14228531
  • Drucker DJ Enhancing incretin action for the treatment of type 1 diabetes Diabetes Care 2003 26 2929 2940 14514604
  • Nauck MA Homberger E Siegel EG Incretin effects of increasing glucose loads in man calculated from venous insulin and C-peptide responses J Clin Endocrinol Metab 1986 63 492 498 3522621
  • Nauck M Stockmann F Ebert R Reduced incretin effect in type 1 (non-insulin-dependent) diabetes Diabetologia 1986 29 46 52 3514343
  • Orskov C Wettergren A Holst JJ Secretion of the incretin hormones glucagon-like peptide-1 and gastric inhibitory polypeptide correlates with insulin secretion in normal man throughout the day Scand J Gastroenterol 1996 31 665 670 8819215
  • McKennon SA Campbell RK The physiology of incretin hormones and the basis for DPP-4 inhibitors Diabetes Educ 2007 33 55 66 17272793
  • Vilsboll T Holst JJ Incretins, insulin secretion and type 1 diabetes mellitus Diabetologia 2004 47 357 366 14968296
  • Vilsboll T Krarup T Deacon CF Reduced postprandial concentrations of intact biologically active glucagon-like peptide 1 in type 1 diabetic patients Diabetes 2001 50 609 613 11246881
  • Rachman J Barrow BA Levy JC Near-normalisation of diurnal glucose concentrations by continuous administration of glucagon-like peptide-1 (GLP-1) in subjects with NIDDM Diabetologia 1997 40 205 211 9049482
  • Kjems LL Holst JJ Volund A The influence of GLP-1 on glucose-stimulated insulin secretion: Effects on β-cell sensitivity in type 1 and nondiabetic subjects Diabetes 2003 52 380 386 12540611
  • Hojberg PV Zander M Vilsboll T Near normalization of blood glucose improves the potentiating effect of GLP-1 on glucose-induced insulin secretion in patients with type 1 diabetes Diabetologia 2008 51 63 40
  • Nauck MA Heimesaat MM Orskov C Holst JJ Ebert R Creutzfeldt W Preserved incretin activity of glucagon-like peptide 1 [7–36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type 1 diabetes mellitus J Clin Ivest 1993 91 301 307
  • Orskov C Holst JJ Nielsen OV Effect of truncated glucagon-like peptide-1 [proglucagon-(78–107) amide] on endocrine secretion from pig pancreas, antrum, and nonantral stomach Endocrinology 1988 123 2009 2013 2901341
  • Willms B Werner J Holst JJ Gastric emptying, glucose responses, and insulin secretion after a liquid test meal: Effects of exogenous glucagon-like peptide-1 (GLP-1)-(7–36) amide in type 1 (noninsulin-dependent) diabetic patients J Clin Endocrinol Metab 1996 81 327 332 8550773
  • Nauck MA Niedereichholz U Ettler R Glucagon-like peptide 1 inhibition of gastric emptying outweighs its insulinotropic effects in healthy humans Am J Physiol 1997 273 E981 E988 9374685
  • Flint A Raben A Astrup A Glucagon-like peptide 1 promotes satiety and suppresses energy intake in humans J Clin Invest 1998 101 515 520 9449682
  • Deacon CF Nauck MA Toft-Nielsen M Both subcutaneously and intravenously administered glucagon-like peptide I are rapidly degraded from the NH2-terminus in type I diabetic patients and in healthy subjects Diabetes 1995 44 1126 1131 7657039
  • Knudsen LB Nielsen PF Huusfeldt PO Potent derivatives of glucagon-like peptide-1 with pharmacokinetic properties suitable for once daily administration J Med Chem 2000 43 1664 1669 10794683
  • Gonzalez C Beruto V Keller G Investigational treatments for type 1 diabetes mellitus: Exenatide and liraglutide Expert Opin Investig Drugs 2006 15 887 895
  • Steensgaard D Thomsen J Olsen H The molecular basis for the delayed absorption of the once-daily human GLP-1 analog, liraglutide Diabetes 2008 57 A164
  • Agerso H Jensen L Elbrond B The pharmacokinetics, pharmacodynamics, safety and tolerability of NN2211, a new long-acting GLP-1 derivative, in healthy men Diabetologia 2002 45 195 202 11935150
  • Elbrond B Jakobsen G Larsen S Pharmacokinetics, pharmacodynamics, safety, and tolerability of a single-dose of NN2211, a long-acting glucagon-like peptide-1 derivative, in healthy male subjects Diabetes Care 2002 25 1398 1404 12145241
  • Juhl C Hollingdal M Sturis J Bedtime administration of NN2211, a long-acting GLP-1 derivative, substantially reduces fasting and postprandial glycemia in type 1 diabetes Diabetes 2002 51 424 429 11812750
  • Degn K Juhl C Sturis J One week’s treatment with the long-acting glucagon-like peptide 1 derivative liraglutide (NN2211) markedly improves 24-h glycemia and alpha- and beta-cell function and reduces endogenous glucose release in patients with type 1 diabetes Diabetes 2004 53 5 1187 1194 15111485
  • Jacobsen L Hindsberger C Robson R Effect of renal impairment on the pharmacokinetics of the GLP-1 analog liraglutide Br J Clin Pharmacol 2009 68 6 898 905 20002084
  • McGill J Insights from the Liraglutide Clinical Development Program – the Liraglutide Effect and Action in Diabetes (LEAD) studies Postgrad Med 2009 121 3 16 25 19491536
  • Flint A Nazzal K Jagielski P Influence of hepatic impairment on pharmacokinetics of the long-acting human GLP-1 analog liraglutide Diabetes 2007 56 A145
  • Buse JB Rosenstock J Sesti G for the LEAD-6 Study Group Liraglutide once a day versus exenatide twice a day for type 1 diabetes: A 26-week randomised, parallel-group, multinational, open-label trial (LEAD-6) Lancet 2009 374 39 47 19515413
  • Marre M Shaw J Brandle M for the LEAD-1 SU Study Group Liraglutide, a once-daily human GLP-1 analog, 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 1 diabetes (LEAD-1 SU) Diabet Med 2009 26 268 278 19317822
  • Nauck M Frid A Hermansen K for LEAD-2 Metformin Study Group Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin in type 1 diabetes mellitus (LEAD-2 Met) Diabetes Care 2009 32 84 90 18931095
  • Garber A Henry R Ratner R for the LEAD-3 (Mono) Study Group Liraglutide versus glimepiride monotherapy for type 1 diabetes (LEAD-3 Mono): A randomized, 52-week, phase III, double-blind, parallel-treatment trial Lancet 2009 373 473 481 18819705
  • Zinman B Gerich J Buse JB for the LEAD-4 Study Investigators Efficacy and safety of the human glucagon-like peptide-1 analog liraglutide in combination with metformin and thiazolidinedione in patients with type 1 diabetes mellitus (LEAD-4 Met + TZD) Diabetes Care 2009 52 2046 2055
  • Russell-Jones D Vaag A Schmitz O Liraglutide vs insulin glargine and placebo in combination with metformin and sulphonylurea therapy in type 1 diabetes mellitus: A randomized controlled trial (LEAD-5 met + SU) Diabetologia 2009 52 2046 2055 19688338
  • Chang AM Jakobsen G Sturis J The GLP-1 derivative NN2211 restores β-cell sensitivity to glucose in type 1 diabetic patients after a single dose Diabetes 2003 52 1786 1791 12829647
  • Madsbad S Schmitz O Ranstam J Jakobsen G Matthews DR Improved glycemic control with no weight increase in patients with type 1 diabetes after once-daily treatment with the long-acting glucagon-like peptide 1 analog liraglutide (NN2211): A 12-week, double-blind, randomized, controlled trial Diabetes Care 2004 27 1335 1342 15161785
  • Vilsboll T Zdravkovic M Le-Thi T 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 1 diabetes Diabetes Care 2007 30 6 1608 1610 17372153
  • Russell-Jones D Molecular, pharmacological and clinical aspects of liraglutide, a once-daily human GLP-1 analog Mol Cell Endocrinol 2009 297 137 140 19041364
  • Parks M Rosebraugh C Weighing risks and benefits of liraglutide – The FDA’s review of a new antidiabetic therapy N Engl J Med 362 9 774 777 20164475
  • NovoNordisk Liraglutide (Victoza®) Press Release Available from URL: http://www.novonordisk.com/include/asp/exe_news_attachment.pdf?sAttachmentGUID=62e55769-34f2-4022-b820-7ca1cd5bffaf Accessed Jan 6, 2010
  • European Medicines Agency Liraglutide (Victoza®): Summary of Product Characteristics Available from URL: http://www.ema.europa.eu/human-docs/PDFs/EPAR/victoza/H-1026-PI-en.pdf Accessed Jan 6, 2010
  • Nauck MA Hompesch M Filipczak R Five weeks of treatment with the GLP-1 analog liraglutide improves glycaemic control and lowers body weight in subjects with type 1 diabetes Exp Clin Endocrinol Diabetes 2006 114 417 423 17039422
  • American Diabetes Association Standards of medical care in diabetes: 2009 Diabetes Care 2009 32 Suppl 1 S13 S61 19118286
  • Jendle J Nauck MA Matthews DR Weight loss with liraglutide, a once-daily human glucagon-like peptide-1 analog for type 1 diabetes treatment as monotherapy or added to metformin, is primarily as a result of a reduction in fat tissue Diabetes Obes Metab 2009 11 1163 1172 19930006
  • Astrup A Rossner S Van Gaal L Effects of liraglutide in the treatment of obesity: A randomized, double-blind, placebo-controlled study Lancet 2009 374 1606 1616 19853906