Abstract
There is a global obesity epidemic that will continue to be a financial burden on healthcare systems around the world. Tackling obesity through diet and exercise should always be the first intervention, but this has not proved to be effective for a large number of patients. Pharmacotherapeutic options have been limited and many previously available drugs have been withdrawn due to safety concerns. Currently, only bariatric surgery has the capability to induce both substantial and durable weight loss. This article briefly reviews the history of pharmacotherapy for obesity before focusing on the clinical trial evidence for the use of the GLP-1 agonist liraglutide as a weight loss agent and comparing its efficacy with other emerging drug therapies for obesity.
Keywords: liraglutide, obesity, pharmacotherapy
Introduction
The trend towards reduced physical activity and increased calorific value of diets in the modern world has led to a pandemic of overweight and obesity. The World Health Organization (WHO) estimates a worldwide prevalence of overweight [a body mass index (BMI) of 25 kg/m2 or higher] of 38% of men and 40% of women [World Health Organization, 2015]. Overweight and obesity are associated with many negative health outcomes including type 2 diabetes mellitus [Colditz et al. 1995], hypertension [Must et al. 1999], cancers [Eheman et al. 2012], cardiovascular disease [Asia-Pacific Cohort Studies Collaboration, 2006], osteoarthritis [Davis et al. 1990], depression [Simon et al. 2008] and other comorbidities [Guh et al. 2009]. Importantly, obesity also carries a significant quality of life cost [Soltoft et al. 2009]. The mortality hazard for a person with BMI of 40 kg/m2 is approximately twice that for someone of normal BMI (BMI 18.5 to 25kg/m2), even after adjustment for the comorbidities associated with being overweight [Berrington et al. 2010]. In one prospective cohort study, an obese 40-year-old woman nonsmoker lost 7.1 years of life expectancy compared with their normal weight counterpart, with a loss of 5.8 years in men [Peeters et al. 2003]. Furthermore, the financial cost of obesity, both in terms of direct healthcare liabilities and indirect economic impact, is enormous and projected to be as much as £27 billion lost to the UK economy in 2015 [Morgan and Dent, 2010].
It is well established that weight loss in obese patients results in both improvement in comorbidities [Wing et al. 2011] and reduction in all-cause mortality [Kritchevsky et al. 2015]. The challenge is achieving this target in a durable way. Lifestyle modifications involving low-calorie diets and/or exercise are effective in inducing weight loss over the short term but the tendency is for much of the lost weight to be regained over time [Franz et al. 2007]. Thus, there is a need for effective additional interventions to provide long-term weight control. Bariatric surgical procedures have been shown to be efficacious [Sjostrom et al. 2012] and cost effective [Picot et al. 2009]. Perioperative morbidity and mortality is low [Aminian et al. 2015], but it is not always possible to avoid the sometimes persistent side effects of post-prandial nausea or vomiting, micronutrient deficiencies or hypoglycaemia related to dumping syndrome. Uptake of surgery in those eligible for this intervention is likely to be limited: the 2007 National Institute for Health and Clinical Excellence (NICE) guide for the commissioning of bariatric surgery assumed a take-up rate of 30–50% [National Institute for Clinical Excellence, 2007]; Since then, NICE has updated the guidelines for the management of obesity and in the absence of other effective measures, has widened the criteria for those eligible [National Institute for Health and Clinical Excellence, 2014a]. There are now estimated to be approximately 1 million people in the UK potentially eligible for surgery [National Institute for Health and Clinical Excellence, 2014b]. It is likely that relatively few of those in this new patient group will opt for surgery. Finding an effective, nonsurgical means of achieving an equivalent weight loss, and its associated health gain, for these patients is thus an urgent priority.
History of pharmacotherapy for obesity
The history of pharmacotherapy for obesity is a chequered one. The first licensed drugs, amphetamine derivatives, were approved by the nascent US Food and Drug Administration (FDA) in the 1940s. They remain available for use in the US but the addictive properties of amphetamines allow for short-term use only, limiting their efficacy for long-term weight loss. By the 1970s their use had dwindled. In an effort to reduce the side-effect burden but retain weight-loss efficacy, combination tablets of lower doses of amphetamine derivatives were explored. Phentermine–fenfluramine (or ‘phen-fen’) was marketed in the early 1990s after a successful trial of 121 patients showing a placebo-subtracted weight loss of 11.0% of baseline without frequent or serious adverse effects [Weintraub et al. 1992]. It became very popular and was heavily used, but had to be withdrawn from the market in 1997 due to evidence linking fenfluramine first with pulmonary hypertension [Brenot et al. 1993] and then with valvulopathies [Connolly et al. 1997]. The same year, sibutramine, a dual serotonin–noradrenaline reuptake inhibitor with anorectic and thermogenic effects [Luque and Rey, 2002], was licensed for use in the US and Europe. However, in 2010 evidence of an increased risk of myocardial infarction and stroke was published [James et al. 2010] and led to the European Medicines Agency (EMA) withdrawing approval.
Rimonabant, an inverse agonist of the cannabinoid receptor CB1, was approved by the EMA (although not the FDA) in 2006. The phase III RIO trial had shown a signal toward increased psychiatric adverse events despite those with psychiatric disorders being excluded from the trial [van Gaal et al. 2005]. A detailed review of the incidence of suicidality in clinical trials of rimonabant, undertaken by Sanofi-Aventis at the request of the FDA, found an odds ratio of 1.9 [95% confidence interval (CI) 1.1–3.1] on a dose of 20 mg compared with placebo [US Food and Drug Administration Advisory Committee, 2007] and it was withdrawn from the European market. Between 2010 and 2015 physicians in the EU had only one licensed drug available for use: orlistat, a pancreatic lipase inhibitor which reduces absorption of dietary lipids. Orlistat results in an approximate 3 kg excess weight loss over placebo per year [Padwal et al. 2003] and remains widely prescribed. The XENDOS trial found that the addition of orlistat to lifestyle measures reduced progression to type 2 diabetes in obese patients with impaired glucose tolerance by 45% over 4 years [Torgerson et al. 2004]. Unfortunately, its mechanism of action leads to gastrointestinal side effects which sometimes prove intolerable and which have somewhat limited its use.
New anti-obesity pharmaceuticals
On the background of this chequered history, several new drugs have recently emerged. A serotonin mimetic called lorcaserin (Belviq®), which possesses appetite suppressant properties through its action at 5-HT2c receptors on pro-opiomelanocortin (POMC) neurons in the appetite centres of the hypothalamus [Thomsen et al. 2008], received FDA approval in 2012. However, the application for authorisation from the EMA was withdrawn after concerns were raised regarding potential negative effects on psychiatric and cardiovascular risk. A phentermine/topiramate combination tablet (Qsymia®) was initially refused authorization by both the FDA and EMA on the grounds of uncertainties as to the cardiovascular safety of phentermine and psychiatric/neurocognitive effects of topiramate [European Medicines Agency, 2013], but after further safety data were provided it was approved in the US in 2012. The mechanism for the weight loss effect of topiramate is uncertain but is likely to involve both reduced calorific intake and increased energy expenditure [Allison et al. 2012].
Within the last year, two further medications for obesity have been approved. Bupropion/naltrexone (Contrave® in US, Mysimba® in EU), was authorized by the FDA in September 2014. Bupropion stimulates the same POMC neurons as lorcaserin through incompletely understood mechanisms and naltrexone blocks autocrine downregulation of this activity by antagonizing opioid receptors [Greenway et al. 2009]. Soon after, in December 2014, the FDA approved liraglutide (under the new name Saxenda®), a glucagon-like peptide-1 (GLP-1) receptor agonist, familiar as a treatment for type 2 diabetes mellitus, for use as a weight loss agent in patients without diabetes. EMA approval for bupropion/naltrexone and liraglutide arrived in March 2015 and the drugs now await member state authorization.
This article will focus on liraglutide, its effects on weight, and how physicians might use high-dose liraglutide in the treatment pathway for patients with obesity.
Previous use of GLP-1 agonists
An oral glucose load results in a greater and more sustained insulin secretion than does an equivalent intravenous load [Elrick et al. 1964], a phenomenon known as the incretin effect. Endogenous GLP-1 was discovered in the early 1980s [Bell et al. 1983] and was shown to act as an incretin at physiological concentrations [Kreymann et al. 1987]. In vivo, GLP-1 is rapidly broken down by dipeptidyl peptidase-4 (DPP-4) and has a brief half-life of 2 minutes, making it unsuitable as a pharmacotherapy for diabetes. However, the potential for pharmacological manipulation of the GLP-1 receptor was evident. The emergence of longer-acting receptor agonists began with the discovery of exendin-4 from the venom of the gila monster, Heloderma suspectum [Eng et al. 1992]. Exendin-4 is a peptide with 53% amino acid identity to human GLP-1. It was shown potently to stimulate the GLP-1 receptor and induce proinsulin gene expression in islet beta cells [Goke et al. 1993]. It is resistant to DPP-4 degradation and possesses a much longer half-life than GLP-1 [Parkes et al. 2001], resulting in prolonged glucose-lowering activity in vivo [Young et al. 1999]. It was soon recognized that, in contrast to previously available insulin secretagogues such as sulfonylureas, administration of exendin-4 resulted in weight loss in animal models [Young et al. 1999], an effect that was later reproduced in phase III clinical trials [DeFronzo et al. 2005]. Exenatide, a synthetic peptide identical to exendin-4, was the first GLP-1 receptor agonist approved for the treatment of type 2 diabetes, authorized by the FDA in 2005 as monotherapy and then by the EMA in 2006 as add-on treatment to metformin and/or sulfonylurea where control was inadequate.
Parallel to the development of exenatide by Amylin Pharmaceuticals, Novo Nordisk worked on an alternative approach to designing a long acting GLP-1 receptor agonist. The GLP-1 peptide sequence was modified at the N-terminus to stabilise the molecule against degradation by DPP-4, and fatty acid side chains were added to enhance protein binding and prolong half-life. The length of the fatty acid chain was optimized for the balance between plasma half-life and GLP-1 receptor agonist potency [Knudsen et al. 2000]. The result was the molecule NN2211, named liraglutide, with one amino acid substitution from GLP-1 (Lys34Arg) and a 16-carbon palmitic acid side chain.
Liraglutide underwent phase III trials in the LEAD (Liraglutide Effect and Action in Diabetes) programme. As with exenatide, liraglutide was associated with a significantly greater weight loss compared with placebo in a dose-dependent manner. Liraglutide led to significant reductions in HbA1c over placebo in all the LEAD trials, amounting to between 1.2 and 1.6 HbA1c percentage points over 52 weeks [Blonde and Russell-Jones, 2009]. Liraglutide was tested at doses of 0.6, 1.2 and 1.8 mg. Weight loss was greatest in the higher 1.8 mg dose compared with lower doses across the LEAD studies. In terms of HbA1c reduction, whilst 1.8 mg outperformed the 1.2 mg dose when used as monotherapy (LEAD-3), no additional benefit of the higher dose was seen when used as add-on to oral hypoglycaemics (LEAD-1, -2 and -4). The 1.8 mg dose was, however, associated with an increased occurrence of predominantly gastro-intestinal side effects. Liraglutide became the second GLP-1 receptor agonist to be licensed for the treatment of type 2 diabetes, by the EMA in July 2009 and the FDA in January 2010. It was the subject of a NICE technology appraisal (TA203) in 2010 which recommended its use at a dose of 1.2 mg (but not 1.8 mg) for patients with T2DM and a BMI of ⩾35 kg/m2 [National Institute for Health and Clinical Excellence, 2010].
Mechanism of GLP-1 receptor agonist-induced weight loss
Endogenous GLP-1 is secreted by the L-cells of the intestine and acts via its G-protein linked membrane-bound receptor (GLP-1R). GLP-1R is expressed predominantly in the upper gastrointestinal tract, pancreatic islets [Bullock et al. 1996] and enteric visceral afferent nerves [Vahl et al. 2007]. In the central nervous system, GLP-1 is secreted as a neurotransmitter by neurons in areas of the brain including the hypothalamus. The GLP-1R is widely distributed throughout the brain [Merchenthaler et al. 1999] and densely expressed in hypothalamic nuclei involved in the regulation of appetite such as the paraventricular and arcuate nuclei [Shughrue et al. 1996].
GLP-1 has an appetite suppressant effect and the GLP-1 response to oral glucose is obtunded in obese patients [Færch et al. 2015]. The mechanism through which GLP-1 regulates appetite appears to be mediated through both peripheral and central nervous system (CNS) pathways. The action of GLP-1 on gut motility has been reviewed elsewhere [Marathe et al. 2011. An important effect is the slowing of gastric emptying which occurs as a result of GLP-1 activity [Delgado-Aros et al. 2002]. The resultant gastric stretch stimulates vagal afferent signals to the solitary nucleus of the medulla and onto the appetite centres of the hypothalamus to induce satiety or the area postrema to induce nausea. Early satiety and nausea are common side-effects of GLP-1 agonist drugs but are generally transient. Indeed, the retardation of gastric emptying by GLP-1 is subject to a rapid tachyphylaxis in experimental studies [Nauck et al. 2011]. Despite this, weight loss with GLP-1 agonists has been maintained in the absence of upper gastrointestinal side effects in clinical trials [Shyangdan et al. 2011], suggesting an alternative appetite suppressant pathway.
The CNS effects of GLP-1 have also been recently reviewed [van Bloemendaal et al. 2014]. It is estimated that 10–15% of endogenous GLP-1 secreted by intestinal L cells may reach the systemic circulation [Holst et al. 2005] where it is found in concentrations of approximately 10–40 pmol/l [Orskov et al. 1996]. From there, endogenous GLP-1 may be able to cross the blood–brain barrier and directly activate GLP-1R in the CNS [Kastin et al. 2002], while more substantial evidence attests to the ability of liraglutide and other GLP-1 analogues to do so [Secher et al. 2014; Hunter and Holscher, 2012]. Liraglutide has been shown to directly stimulate POMC neurons and inhibit neuropeptide-Y and Agouti-related peptide neurons of the arcuate nucleus resulting in appetite suppression [Secher et al. 2014]. These actions may also be accompanied by effects on other areas of the brain such as the mesolimbic system resulting in diminished food-induced reward signals and hence reduced food-seeking behaviour [Dickson et al. 2012; Dossat et al. 2011]. The efficacy of GLP-1 receptor agonists in maintaining weight loss may be explained by an attenuation of the fall in the anorexigenic hormone leptin that accompanies weight loss [Iepsen et al. 2014].
It is unclear whether GLP-1 influences energy expenditure. Some rodent models have demonstrated increased thermogenesis with administration of GLP-1 [Osaka et al. 2005]. Human studies, however, have yielded conflicting results [Harder et al. 2004; Horowitz et al. 2012].
In summary, while GLP-1 may increase energy expenditure in pharmacological concentrations, its predominant influence on body weight is through suppression of energy intake through direct and indirect actions on the appetite and food-reward centres of the brain and through local gastrointestinal effects.
Liraglutide: weight loss effects in diabetes: the LEAD and DURATION-6 trials
The LEAD and DURATION-6 trials are summarized in Table 1.
Table 1.
Trial name | Study population | Study groups | Primary outcome measure | Duration | Weight change with 0.6 mg | 1.2 mg | 1.8 mg | Statistical significance of weight loss |
---|---|---|---|---|---|---|---|---|
LEAD-1 (SU) |
|
OHGs discontinued and glimepiride (2–4 mg daily) introduced.Study arms:
|
Change in HbA1c from baseline at 26 weeks | 26 weeks | +0.7 kg | +0.3 kg | −0.2 kg | Nonsignificant compared with double placebo |
LEAD-2 (Met) |
|
OHGs discontinued and metformin (1000 mg BD) introduced.Study arms:
|
Change in HbA1c from baseline at 26 weeks | 26 weeks | −1.8 kg | −2.6 kg | −2.8 kg | Versus placebop < 0.01 for 1.2 and 1.8 mgVersus glimepiride 4 mg p < 0.0001 for 0.6, 1.2 and 1.8 mg |
LEAD-3 (Mono) |
|
OHG monotherapy discontinued.Study arms:
|
Change in HbA1c from baseline at 52 weeks | 52 weeks | N/A | −2.0 kg | −2.4 kg | Versus glimepiride 8 mg p = 0.0001 for 1.2 and 1.8 mg |
LEAD-4 (Met+TZD) |
|
OHG therapy discontinued. | Change in HbA1c from baseline at 26 weeks | 26 weeks | N/A | −1.0 kg | −2.0 kg | Versus placebo p < 0.0001 for 1.2 and 1.8 mg |
Metformin (1000 mg BD) and rosiglitazone (8 mg OD) introduced. | ||||||||
Study arms:
|
||||||||
LEAD-5 (Met + SU) |
|
OHG therapy discontinued. | Change in HbA1c at 26 weeks | 26 weeks | N/A | N/A | −1.8 kg |
Versus placebop = 0.0001 Versus lantusp < 0.0001 |
Metformin (1000 mg BD) and glimepiride 4 mg OD) introduced. | ||||||||
Study arms:
|
||||||||
LEAD-6 (versus exanatide BD head-to-head) |
|
Previous therapy continued. | Change in HbA1c at 26 weeks | 26 weeks | N/A | N/A | −3.2 kg | Nonsignificant |
Study arms:
|
||||||||
Open label | ||||||||
DURATION-6 (versus exenatide once weekly head-to-head) |
|
Previous therapy continued.Study arms:
|
Change in HbA1c at 26 weeks | 26 weeks | N/A | N/A | −3.6 kg | Liraglutide superior to exenatide, p = 0.0005 |
OHG, Oral hypoglycaemic; T2DM, type 2 diabetes mellitus; BMI, body mass index; OD, once daily; BD, twice daily; N/A, not available.
The 26-week, phase III, Liraglutide Effect and Action in Diabetes (LEAD) series of trials in patients with type 2 diabetes were powered to assess reduction in HbA1c as a primary endpoint, but also had sufficient statistical power to detect a 3% fall in weight. The results of LEAD trials 1 to 5 have previously been summarized by Blonde and Russell-Jones [Blonde and Russell-Jones, 2009].
In LEAD 1 (addition of liraglutide, rosiglitazone or placebo to sulfonylurea); participants in the active arm with a mean BMI of 30 kg/m2 did not lose significant weight compared with placebo [Marre et al. 2009].
In LEAD 2 (addition of liraglutide, glimepiride or placebo to metformin), participants with a mean BMI of 31 kg/m2 lost 2.8 kg at 1.8 mg dose over 26 weeks, significantly greater than the placebo arm (1.5 kg) (p < 0.0001) [Nauck et al. 2009].
In LEAD 3 (liraglutide versus glimepiride as monotherapy), participants with a mean BMI of 33 kg/m2 lost between 2.0 (1.2 mg dose, p < 0.0001) and 2.4 kg (1.8 mg dose, p < 0.0001), compared with a weight gain of 1.2 kg in the glimepiride arm [Garber et al. 2009].
In LEAD 4 (liraglutide or placebo added to combination of metformin and rosiglitazone), participants with a mean BMI of 33–34 kg/m2 lost 1.0 kg (1.2 mg dose, p < 0.0001) to 2.0 kg (1.8 mg dose, p < 0.0001) which was significantly different to the weight gain of 0.6 kg in the placebo arm [Zinman et al. 2009].
In LEAD 5 [addition of liraglutide (fixed-dose, 1.8 mg), insulin glargine (patient-titrated) or placebo to metformin and sulfonylurea], participants with a mean BMI of 30–31 kg/m2 lost 1.8 kg, significantly greater than placebo (−0.4 kg, p = 0.0001) and insulin glargine (+1.6 kg, p < 0.0001) [Russell-Jones et al. 2009].
Liraglutide was compared to exenatide in a head-to-head trial, LEAD-6 [Buse et al. 2009]. Patients on metformin, sulfonylurea or both were given either 1.8 mg liraglutide or 10 µg twice daily exenatide. After 26 weeks, participants in the liraglutide arm lost 3.24 kg compared with 2.87 kg in the exenatide arm, a nonsignificant difference (p = 0.2235), from a baseline BMI of 33 kg/m2. A degree of weight loss was seen in 78% of participants in the liraglutide arm and 76% on exenatide.
The DURATION-6 trial compared 1.8 mg liraglutide daily with 2 mg sustained-release exenatide (SR exenatide or Bydureon®) once weekly in patients with type 2 diabetes inadequately controlled on oral hypoglycaemic agents [Buse et al. 2013]. Mean BMI was 32.3 kg/m2 in both groups. Weight loss after 26 weeks (as a secondary endpoint) was significantly greater in the liraglutide arm (−3.57 liraglutide versus −2.68 kg SR exenatide, p = 0.0005).
Tolerability and safety of liraglutide in these trials was generally good. The most frequent side effects were gastrointestinal, predominantly nausea, which was most prevalent in the first few weeks then dissipated over time. The withdrawal rate in the liraglutide treatment arms was equivalent to or less than the comparator arms in all of the LEAD trials.
Liraglutide: weight loss effects in obesity (the SCALE trials)
Table 2 provides a summary of the SCALE trials. The first study to assess the effect of liraglutide on body weight and tolerability in obese individuals without type 2 diabetes was a double blind, placebo-controlled 20-week trial with open-label orlistat as a comparator and four different doses of liraglutide (1.2, 1.8, 2.4 and 3.0 mg) [Astrup et al. 2009]. All individuals received a 500 kcal per day energy-deficit diet and increased their activity throughout the trial which had an 84-week open-label extension that followed. It became clear from this dose-finding study that the 3 mg dose of liraglutide was the most effective dose for inducing weight loss at 20 weeks and beyond, and this dose was subsequently used for large scale phase III clinical trials. Mean weight loss after 20 weeks was 4.8 kg (1.2 mg dose), 5.5 kg (1.8 mg), 6.3 kg (2.4 mg) and 7.2 kg (3.0 mg). Orlistat resulted in a mean weight loss of 4.1 kg and placebo 2.1 kg.
Table 2.
Trial name | Study population | Study design | Duration | Weight loss | Statistical significance | ||
---|---|---|---|---|---|---|---|
Placebo | 1.8 mg | 3.0 mg | |||||
SCALE – diabetes |
|
|
56 weeks | −2.0% | −4.7% | −6.0% | p = <0.001 for 1.8 mg and 3.0 mg doses versus placebo |
Study arms:
|
|||||||
SCALE – pre-diabetes |
|
|
56 weeks | −2.6% | N/A | −8.0% | p = <0.001 versus placebo |
Study arms
|
|||||||
SCALE - Maintenance |
|
|
56 weeks | −0.2% (in addition to run-in weight loss) | N/A | −6.2% (in addition to run-in weight loss | p = <0.0001 versus placebo |
Study arms:
|
|||||||
SCALE – Sleep Apnoea |
|
|
36 weeks | −1.6% | N/A | −5.7% | p < 0.0001 versus placebo |
Study arms:
|
OHG, Oral hypoglycaemic; T2DM, type 2 diabetes mellitus; BMI, body mass index; CPAP, continuous positive airway pressure; N/A, not available.
The weight loss efficacy of the 3 mg dose of liraglutide in patients with and without diabetes has been the subject of the Satiety and Clinical Adiposity – Liraglutide Evidence (SCALE) series of four trials [Novo Nordisk, 2015]. The series comprised trials in four patient categories: (1) overweight/obesity and type 2 diabetes; (2) overweight/obesity and prediabetes; (3) the SCALE - Maintenance trial which assessed the ability of high-dose liraglutide to maintain weight lost following a low-calorie diet and exercise intervention; and (4) obesity and sleep apnoea. All trials tested the effect of liraglutide on weight loss as an adjunct to diet and physical exercise.
SCALE - Diabetes trial
The results of the SCALE - Diabetes trial were presented at the American Diabetes Association conference in 2014 [Davies et al. 2014]. A total of 846 participants with type 2 diabetes, BMI ⩾27 kg/m2 and HbA1c of 7.0–10.0% on up to three oral agents were randomised to receive liraglutide 3.0 mg, liraglutide 1.8 mg or placebo plus lifestyle intervention for 56 weeks. Participants had a mean BMI of 37 kg/m2 and body weight of 106 kg. After 56 weeks, both the 3.0 and 1.8 mg doses of liraglutide performed significantly better than placebo, with percentage body weight losses of 5.9%, 4.6% and 2.0%, respectively. A total of 49.9% of individuals on 3.0 mg liraglutide lost ⩾5.0% of body weight, compared with 35.6% at the lower dose and 13.8% on placebo. A total of 23.4%, 14.4% and 4.3% lost >10% body weight in the three arms, respectively. The reported incidence of nausea was similar in the 3.0 and 1.8 mg arms, peaking at 16% after 4 weeks and subsiding thereafter.
SCALE - Obesity and Pre-diabetes trial
The results of the SCALE - Obesity and Pre-diabetes trial were published in July 2015 [Pi-Sunyer et al. 2015]. Patients with type 2 diabetes were excluded from this trial. Participants had either no diabetes or prediabetes as defined by the American Diabetes Association 2010 criteria [American Diabetes Association, 2010]. A total of 3731 participants with BMI ⩾30 kg/m2 or BMI ⩾27 kg/m2 and a cardiovascular comorbidity were randomized 2:1 to receive 3.0 mg liraglutide or placebo for a period of 56 weeks. In those without prediabetes (n = 1446), mean baseline body weight was 104 kg and BMI 37.5 kg/m2. Participants with prediabetes (n = 2285) were slightly more obese with baseline weight 108 kg and BMI of 39 kg/m2.
Weight loss achieved by those with and without prediabetes was similar and equated to a mean weight loss of −8.0% (8.4 kg) from a baseline of 106 kg, significantly greater than that in the placebo group (−2.6% or 2.8 kg). A total of 64% of subjects in the liraglutide arm achieved ⩾5% body weight loss compared with 27% in the placebo arm. A total of 33% of subjects achieved more than 10% body weight loss compared with 10% in the placebo arm. Liraglutide was associated with a reduced progression to prediabetes (7.2% against 20.7%) and increased reversal of prediabetes (69.2% against 32.7%) as defined by American Diabetes Association (ADA) criteria.
SCALE - Maintenance trial
The SCALE - Maintenance trial results were published in 2013 [Wadden et al. 2013]. In this study, obese (or overweight with BMI ⩾27 with comorbidity) subjects undertook a 1200–1400 kcal/day diet and exercise programme during a 12-week run-in. Patients were entered into the trial if they managed to lose ⩾5% body weight at 12 weeks prior to randomization. A total of 422 participants were randomized to receive 3.0 mg liraglutide or placebo for a period of 56 weeks. Primary endpoint analyses were percentage weight change from randomisation, proportion maintaining the initial 5% weight loss and proportion achieving an additional 5% weight loss from randomization. The mean weight loss at randomization was −6.0% from an initial baseline of 105–107 kg (BMI 38 kg/m2). Significantly more additional weight was lost by those on liraglutide (−6.2%) than those on placebo (−0.2%). A total of 81.4% in the liraglutide arm maintained a >5% loss from baseline weight, 48.9% on placebo. An extra 5% weight loss was achieved by 50.5% versus 21.8% of participants in the liraglutide and placebo arms, respectively, >10% weight loss by 26.1% and 6.3%, respectively.
SCALE - Sleep Apnoea
This was the smallest of the four trials with 359 patients [Collier et al. 2014]. Patients with moderate or severe obstructive sleep apnoea (OSA) were randomized to liraglutide or placebo for 32 weeks and reduction in the apnoea–hypopnea index (AHI) was evaluated at the end of the trial. Results showed a significantly greater reduction in weight and reduction in AHI in the liraglutide arm compared to placebo, suggesting improvement of OSA on liraglutide treatment.
Additional beneficial effects
A consistent if moderate improvement in low-density lipoprotein (LDL), very low-density lipoprotein (VLDL), total cholesterol and triglyceride concentrations was seen across the SCALE trials, alongside a small reduction in both systolic and diastolic blood pressure. Urine albumin:creatinine ratio in SCALE - Diabetes was reduced by a mean average of 18.4% in the liraglutide 3.0 mg arm against −2.3% with placebo.
Adverse events
Adverse events across the trials occurred more frequently in the liraglutide arms than placebo with the expected gastrointestinal events predominating. Withdrawal due to adverse events ranged between 8.5% in SCALE – Maintenance (in which the placebo arm saw 8.6% withdraw for the same reason) to 9.9% in SCALE – Obesity and Pre-diabetes (versus 3.8% with placebo).
The association of GLP-1 agonists and DPP-4 antagonists with pancreatitis has been the source of much controversy [Butler et al. 2013; Nauck, 2013]. One putative mechanism involves a GLP-1-induced increase in pancreatic duct turnover and metaplasia [Matveyenko et al. 2009]. Recent meta-analyses [Li et al. 2014; Giorda et al. 2015; Wang et al. 2015] have failed to show increased risk of acute pancreatitis with incretin therapies. In the SCALE - Obesity and Pre-diabetes trial, 10 patients on liraglutide and 1 on placebo developed acute pancreatitis (an event rate of 0.4 per 100 patient-years of exposure against 0.1 on account of the 2:1 randomization). There were no pancreatitis episodes in the other three trials. Lipase activity monitoring from the SCALE - Diabetes trial showed an early increase in activity which was sustained throughout the trial in those on liraglutide which was not seen with placebo. This did not rise above the upper limit of normal and no pancreatitis episodes were reported. Gallbladder-related complications, including cholelithiasis and cholecystitis, were more common in the liraglutide arm of each of these trials but serious adverse events involving gallbladder disease were rare.
Concern regarding the effect of liraglutide on thyroid C cells and risk of medullary thyroid cancer stems from evidence that rodents exposed to long-term liraglutide have an increased incidence of thyroid C-cell hyperplasia and medullary thyroid cancer [Bjerre Knudsen et al. 2010]. As a result, liraglutide’s drug information leaflet in the United States contains a black-box warning to this effect. In one large human study, no evidence of an increase in circulating calcitonin in association with liraglutide use was found [Hegedus et al. 2011]. Calcitonin concentrations were monitored during the SCALE trials. A small number of patients were found to have persistent elevations of calcitonin across the trials, more frequently in the liraglutide arms, but overall no statistical association between liraglutide and elevated calcitonin was seen. No incidents of medullary thyroid cancer were reported. The FDA has mandated the keeping of a 15-year medullary thyroid cancer registry for those prescribed 3.0 mg liraglutide.
Liraglutide treatment was associated with a small increase in mean heart rate of between two and four beats per minute across the four trials, although adverse event reports of palpitations and tachycardia occurred in less than 1% of participants in the treatment arms of each trial and were not significantly more frequent than for placebo. The increase in pulse rate returned to normal rapidly on cessation of treatment. Whether or not this heart rate increase is clinically meaningful will be addressed by currently ongoing post-marketing cardiovascular outcomes trials.
In summary, these trials point towards a moderate weight loss effect of liraglutide without evidence of severe toxicity and good tolerability.
Regulatory guidelines for obesity drug approval
Both the FDA and EMA have guideline thresholds for efficacy for any drug seeking marketing authorization as a weight-loss agent. Both agencies require weight loss to be the primary endpoint of the trial. The FDA efficacy benchmarks are (after one year) either (1) a placebo-subtracted mean weight loss of 5% body weight with statistically significant difference from placebo or (2) a proportion of patients in the active treatment group achieving 5% body weight loss of ⩾35% and significantly higher than placebo [Food and Drug Administration, 2007]. The EMA target (after 1 year) is of a mean weight loss of ⩾10% which in turn is ⩾5% more than placebo. If <5% more than placebo, the proportion of each group who respond can be considered as an alternative qualification, although a specified target for this difference is not defined [European Medicines Agency, 2007].
In composite data analysis of the SCALE trials, 3.0 mg liraglutide led to a 7.5% weight loss over 1 year compared with 2.3% for placebo which, although not quite meeting the EMA target, was considered to be sufficiently beneficial for approval [European Medicines Agency, 2015a]. The summary of evidence used by the Committee for Medicinal Products for Human Use in forming a favourable opinion of liraglutide for weight loss was published in February 2015 [European Medicines Agency, 2015b]. The FDA briefing document providing the basis for its approval was published in September 2014 [Food and Drug Administration, 2014].
How does liraglutide 3.0 mg compare with other new weight-loss pharmaceuticals
A comparison of liraglutide with other new weight-loss pharmaceuticals is given in Table 3. The efficacy of phentermine/topiramate was evaluated in three phase III clinical trials (EQUIP, CONQUER and SEQUEL). The EQUIP trial [Allison et al. 2012] included patients without significant obesity-associated comorbidities. Subjects received a dose of 15/92, 3.75/23 mg or placebo. After 56 weeks, mean weight loss was 10.9%, 5.1% and 1.6%, respectively. The CONQUER trial [Gadde et al. 2011] enrolled patients with obesity-related comorbidities: hypertension, prediabetes or type 2 diabetes on diet control or metformin. Three arms received 15/92, 7.5/46 mg or placebo and had a mean weight loss of 9.8%, 7.8% and 1.2%, respectively. A total of 78% of trial completers elected to continue on their randomized arms for a 52-week extension study, SEQUEL [Garvey et al. 2012]. They finished the study with mean weight losses of 10.5%, 9.3% and 1.8%.
Table 3.
Drug | Trial | Particulars of trial | Duration | Dose | Weight loss | Reference | Licensed in EU? |
---|---|---|---|---|---|---|---|
Phentermine / topiramate | EQUIP | Uncomplicated obesity | 56 weeks | 3.75/23 mg | −5.1% | Allison et al. [2012] | No |
15/92 mg | −10.9% | ||||||
CONQUER | Obesity plus metabolic syndrome comorbidities | 56 weeks | 7.5/46 | −7.8% | Gadde et al. [2011] | ||
15/92 | −9.8% | ||||||
SEQUEL | See above | 108 weeks | 7.5/46 | −9.3% | Garvey et al. [2012] | ||
15/92 | −10.5% | ||||||
Lorcaserin | BLOOM | Obesity with nondiabetes metabolic syndrome comorbidity | 52 weeks | 10 mg BD | −5.8% | Smith et al. [2010] | No |
BLOOM-DM | Obesity and T2DM | 52 weeks | 10 mg OD | −5.0% | O’Neill et al. [2011] | ||
10 mg BD | −4.5% | ||||||
BLOSSOM | Obesity with nondiabetes metabolic syndrome comorbidity | 52 weeks | 10 mg OD | −4.7% | Fidler et al. [2011] | ||
10 mg BD | −5.8% | ||||||
Naltrexone / bupropion | COR-I | Obesity without diabetes | 56 weeks | 16/360 mg | −5.0% | Greenwayet al. [2010] | Yes |
32/360 mg | −6.1% | ||||||
COR-II | Obesity and nondiabetes metabolic syndrome comorbidity | 56 weeks | 32/360 mg | −6.4% | Apovianet al. [2013] | ||
COR-DM | Obesity and T2DM | 56 weeks | 32/360 mg | −5.0% | Hollanderet al. [2013] |
T2DM, type 2 diabetes mellitus; OD, once daily; BD, twice daily.
Lorcaserin was also the subject of three phase III trials: BLOOM (10 mg BD versus placebo), BLOSSOM (10 mg BD, OD or placebo) and BLOOM-DM (patients with type 2 diabetes, 10 mg BD, OD or placebo). In the BLOOM trial [Smith et al. 2010], patients in the lorcaserin arm lost a mean of 5.81% body weight compared with 2.16% on placebo after 1 year. At the end of 1 year, the active treatment arm was re-randomized to continue on lorcaserin or receive placebo for a further year. Both groups saw weight regain, the lorcaserin arm regained significantly less weight than the placebo arm. In the BLOSSOM trial [Fidler et al. 2011], 10 mg lorcaserin BD led to significantly more weight loss (5.8%) than 10 mg OD (4.7%) and placebo (2.8%). The same dose–response effect was not seen in the BLOOM-DM study [O’Neill et al. 2012]: participants on 10 mg BD lost and equivalent mean weight (4.5%) to those on OD (5.0%), both significantly greater than placebo (1.5%).
Four phase III trials were undertaken assessing naltrexone/bupropion, named: COR-I, COR-II, COR-BMOD and COR-DM. COR-I [Greenway et al. 2010] compared a dose of 16/360 mg with a dose of 32/360 mg and placebo. Both active treatment arms outperformed placebo but no significant dose–response effect of naltrexone was shown. COR-II [Apovian et al. 2013] examined the efficacy of the higher 32/360 mg dose against placebo with a placebo subtracted mean weight loss of 5.2% after 52 weeks (6.4% and 1.2%), with 50.5% versus 17.1% achieving ⩾5% weight loss at 1 year. COR-BMOD [Wadden et al. 2010] demonstrated that naltrexone/bupropion could significantly improve the weight loss achieved with an intensive behaviour modification intervention. Participants in COR-DM [Hollander et al. 2013] with type 2 diabetes, half of whom were treated with a sulfonylurea and a third with a thiazolidinedione, had a lesser weight loss than in COR-I and COR-II but which remained statistically higher than placebo (5.0% versus 1.8%).
Thus, in phase III pharmaceutical industry trials, 3.0 mg liraglutide resulted in slightly greater weight loss than naltrexone/bupropion and lorcaserin, and somewhat less weight loss than phentermine/topiramate. All of the newer agents’ weight loss effect was greater than that commonly observed with orlistat, in the region of 3% [Padwal et al. 2003] (Table 3).
Conclusion
For decades the world has lacked effective pharmacotherapy for the treatment of obesity. The few available drugs have been withdrawn over time due to unfavourable side-effect profiles. In Europe, with the exception of orlistat, there has been a void in the pharmacotherapy armamentarium, with few weight-reducing options available between diet and exercise and bariatric surgery. The SCALE clinical programme has shown that liraglutide, via manipulation of the physiological satiety hormone GLP-1, is an effective and well-tolerated anti-obesity drug with additional beneficial effects on diabetes, prediabetes and dyslipidaemia. It is a welcome addition to the treatment options available to the obesity physician. Further research studies will be useful to determine which patients are likely to derive the greatest benefit from high-dose liraglutide and to further define its place in the management of obesity.
Footnotes
Funding: This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
Conflict of interest statement: The authors declare that there is no conflict of interest.
Contributor Information
James Crane, Guy’s and St Thomas’ NHS Foundation Trust - Diabetes and Endocrinology, St Thomas’ Hospital Westminster Bridge Road, London SE1 7EH, UK.
Barbara McGowan, Guy’s and St Thomas’ NHS Foundation Trust - Diabetes and Endocrinology, and King’s College London - Diabetes and Nutritional Sciences, London, UK.
References
- Allison D., Gadde K., Garvey W., Petersen C., Schwiers M., Najarian T., et al. (2012) Controlled-release phentermine/topiramate in severely obese adults: a randomized controlled trial (EQUIP). Obesity 20: 330–342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- American Diabetes Association (2010) Diagnosis and classification of diabetes mellitus. Diabetes Care 33: S62–S69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aminian A., Brethaue S., Kirwan J., Kashyap S., Burguera B., Schauer P. (2015) How safe is metabolic/diabetes surgery? Diabetes Obes Metab 17: 198–201. [DOI] [PubMed] [Google Scholar]
- Apovian C., Aronne L., Rubino D., Still C., Wyatt H., Burns C., et al. (2013) A randomized, phase 3 trial of naltrexone SR/bupropion SR on weight and obesity-related risk factors COR-II. Obesity 21: 935–943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Asia-Pacific Cohort Studies Collaboration (2006) Central obesity and risk of cardiovascular disease in the Asia Pacific region. Asia Pac J Clin Nutr 15: 287–292. [PubMed] [Google Scholar]
- Astrup A., Rossner S., Van Gaal L., Rissanen A., Niskanen L., Al Hakim M., et al. (2009) Effects of liraglutide in the treatment of obesity: a randomised, double-blind, placebo controlled study. Lancet 374: 1606–1616. [DOI] [PubMed] [Google Scholar]
- Bell G., Santerre R., Mullenbach G. (1983) Hamster preproglucagon contains the sequence of glucagon and two related peptides. Nature 302: 716–718. [DOI] [PubMed] [Google Scholar]
- Berrington de, Gonzalez A., Hartge P., Cerhan J., Flint A., Hannan L., MacInnis R., et al. (2010) Body mass index and mortality among 1.46 million white adults. N Eng J Med 363: 2211–2219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bjerre Knudsen L., Madsen L., Andersen S., Almholt K., de Boer A., Drucker D., et al. (2010) Glucagon-like Peptide-1 receptor agonists activate rodent thyroid C-cells causing calcitonin release and C-cell proliferation. Endocrinology 151: 1473–1486. [DOI] [PubMed] [Google Scholar]
- Blonde L., Russell-Jones D. (2009) The safety and efficacy of liraglutide with or without oral antidiabetic drug therapy in type 2 diabetes: an overview of the LEAD 1–5 studies. Diabetes Obes Metab 11: 26–34. [DOI] [PubMed] [Google Scholar]
- Brenot F., Herve P., Petitpretz P., Parent F., Duroux P., Simonneau G. (1993) Primary pulmonary hypertension and fenfluramine use. Br Heart J 70: 537–541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bullock B., Heller R., Habener J. (1996) Tissue distribution of messenger ribonucleic acid encoding the rat glucagon-like peptide-1 receptor. Endocrinology 137: 2968–2978. [DOI] [PubMed] [Google Scholar]
- Buse J., Rosenstock J., Sesti G., Schmidt W., Montanya E., Brett J., et al. (2009) 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 374: 39–47. [DOI] [PubMed] [Google Scholar]
- Butler P., Elashoff M., Elashoff R., Gale E. (2013) A critical analysis of the clinical use of incretin-based therapies: are the GLP-1 therapies safe? Diabetes Care 36: 2118–2125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colditz G., Willett W., Rotnitzky A., Manson J. (1995) Weight gain as a risk factor for clinical diabetes mellitus in women. Ann Intern Med 122: 481–486. [DOI] [PubMed] [Google Scholar]
- Collier A., Blackman A., Foster G., Zammit G., Rosenberg R., Wadden T., et al. (2014) Liraglutide 3.0mg reduces severity of obstructive sleep apnoea and body weight in obese individuals with moderate or severe disease: SCALE sleep apnoea trial. Thorax 69: A16–A17. [Google Scholar]
- Connolly H., Crary J., McGoon M., Hensrud D., Edwards B., Edwards W., et al. (1997) Valvular heart disease associated with fenfluramine–phentermine. N Eng J Med 337: 581–588. [DOI] [PubMed] [Google Scholar]
- Davies M., Bode B., Kushner R., Lewin A., Skjoth T., Jensen C., et al. (2014) Liraglutide 3.0 mg for weight management in obese/overweight adults with type 2 diabetes: Results from the SCALE™ Diabetes 56-week randomized, double-blind, placebo-controlled trial. Diabetes 63(Suppl. 1): Abstract 97-OR. [Google Scholar]
- Davis M., Ettinger W., Neuhaus J. (1990) Obesity and osteoarthritis of the knee: evidence from the National Health and Nutrition Examination Survey (NHANES I). Semin Arthritis Rheum 20: S34–S41. [DOI] [PubMed] [Google Scholar]
- DeFronzo R., Ratner R., Han J., Kim D., Fineman M., Baron A. (2005) Effects of exenatide (exendin-4) on glycaemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetes. Diabetes Care 28: 1092–1100. [DOI] [PubMed] [Google Scholar]
- Delgado-Aros S., Kim D., Burton D., Thomforde G., Stephens D., Brinkmann B., et al. (2002) Effect of GLP-1 on gastric volume, emptying, maximum volume ingested, and postprandial symptoms in humans. Am J Physiol Gastrointest Liver Physiol 282: G424–G431. [DOI] [PubMed] [Google Scholar]
- Dickson S., Shirazi R., Hansson C., Bergquist F., Nissbrandt H., Skibicka K. (2012) The glucagon-like peptide 1 (GLP-1) analogue, exendin-4, decreases the rewarding value of food: a new role for mesolimbic GLP-1 receptors. J Neurosci 32: 4812–4820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dossat A., Lilly N., Kay K., Williams D. (2011) Glucagon-like peptide-1 receptors in nucleus accumbens affect food intake. J Neurosci 31: 14453–14457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eheman C., Henley S., Ballard-Barbash R., Jacobs E., Schymura M., Noone A., et al. (2012) Annual Report to the Nation on the status of cancer, 1975–2008, featuring cancers associated with excess weight and lack of sufficient physical activity. Cancer 118: 2338–2366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elrick H., Stimmler L., Hlad C., Jr, Arai Y. (1964) Plasma insulin response to oral and intravenous glucose administration. J Clin Endocrinol Metab 24: 1076–1082. [DOI] [PubMed] [Google Scholar]
- Eng J., Kleinman W., Singh L., Singh G., Raufman J. (1992) Isolation and characterization of exendin-4, an exendin-3 analogue, from Heloderma suspectum venom. Further evidence for an exendin receptor on dispersed acini from guinea pig pancreas. J Biol Chem 267: 7402–7405. [PubMed] [Google Scholar]
- European Medicines Agency (2007) Guideline on Clinical Evaluation of Medicinal Products Used in Weight Control. Committee for Medicinal Products for Human Use; Available at: http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC500003264.pdf (accessed 13 May 2015). [Google Scholar]
- European Medicines Agency (2013) Refusal of the Marketing Authorisation of Qsiva (Phentermine/Topiramate). Available at: http://www.ema.europa.eu/docs/en_GB/document_library/Summary_of_opinion_-_Initial_authorisation/human/002350/WC500139215.pdf (accessed 29 April 2015).
- European Medicines Agency (2015a) EPAR Summary for the Public: Saxenda (Liraglutide). Available at: http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Summary_for_the_public/human/003780/WC500185789.pdf (accessed 13 May 2015).
- European Medicines Agency (2015b) Assessment Report: Saxenda (International Non-Propietary Name: Liraglutide). Committee for Medicinal Products for Human Use; Available at: http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Public_assessment_report/human/003780/WC500185788.pdf (accessed 15 May 2015). [Google Scholar]
- Færch K., Torekov S., Vistisen D., Johansen N., Witte D., Jonsson A., et al. (2015) GLP-1 response to oral glucose is reduced in pre-diabetes, screen-detected type 2 diabetes and obesity and influenced by sex. Diabetes 64: 2513–2525. [DOI] [PubMed] [Google Scholar]
- Fidler M., Sanchez M., Raether B., Weissman N., Smith S., Shanahan W., et al. (2011) A one-year randomized trial of lorcaserin for weight loss in obese and overweight adults: the BLOSSOM trial. J Clin Endocrinol Metab 96: 3067–3077. [DOI] [PubMed] [Google Scholar]
- Food and Drug Administration (2007) Guidance for Industry: Developing Products for Weight Management. Available at: http://www.fda.gov/downloads/Drugs/Guidances/ucm071612.pdf (accessed 13 May 2015).
- Food and Drug Administration (2014) FDA Briefing Document NDA 206321: Liraglutide Injection, 3mg. Endocrinologic and Metabolic Drugs Advisory Committee; Available at: http://www.fda.gov/downloads/advisorycommittees/committeesmeetingmaterials/drugs/endocrinologicandmetabolicdrugsadvisorycommittee/ucm413317.pdf (accessed 15 May 2015). [Google Scholar]
- Franz M., van Wormer J., Crain A., Boucher J., Histon T., Caplan W., et al. (2007) Weight-loss outcomes: a systematic review and meta-analysis of weight-loss clinical trials with a minimum 1-year follow-up. J Am Diet Assoc 107: 1755–1767. [DOI] [PubMed] [Google Scholar]
- Gadde K., Allison D., Ryan D., Peterson C., Troupin B., Schwiers M., et al. (2011) Effects of low-dose, controlled release, phentermine plus topiramate combination on weight and associated comorbidities in overweight and obese adults (CONQUER): a randomised, placebo-controlled, phase-3 trial. Lancet 377: 1341–1352. [DOI] [PubMed] [Google Scholar]
- Garber A., Henry R., Ratner R., Garcia-Hernandez P., Rodriguez-Pattzi H., Olvera-Alvarez I., et al. (2009) Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a randomised, 52-week, phase III, double-blind, parallel-treatment trial. Lancet 373: 473–481. [DOI] [PubMed] [Google Scholar]
- Garvey W.T., Ryan D.H., Look M., Gadde K.M., Allison D.B., Peterson C.A., et al. (2012) Two-year sustained weight loss and metabolic benefits with controlled-release phentermine/topiramate in obese and overweight adults (SEQUEL): a randomized, placebo-controlled, phase 3 extension study. Am J Clin Nutr 95(2): 297–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Giorda C., Sacerdote C., Nada E., Marafetti L., Baldi I., Gnavi R. (2015) Incretin-based therapies and acute pancreatitis risk: a systematic review and meta-analysis of observational studies. Endocrine 48: 461–471. [DOI] [PubMed] [Google Scholar]
- Goke R., Fehmann H., Linn T., Schidt H., Krause M., Eng J., et al. (1993) Exendin-4 is a high potency agonist and truncated exendin-(9–39)-amide an antagonist at the glucagon-like peptide 1-(7–36)-amide receptor of insulin-secreting beta-cells. J Biol Chem 268: 19650–19655. [PubMed] [Google Scholar]
- Greenway F., Fujioka K., Plodkowski R., Mudaliar S., Guttadauria M., Erickson J., et al. (2010) Effect of naltrexone plus bupropion on weight loss in overweight and obese adults (COR-I): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 376: 595–605. [DOI] [PubMed] [Google Scholar]
- Greenway F., Whitehouse M., Guttadauria M., Anderson J., Atkinson R., Fujioka K., et al. (2009) Rational design of a combination medication for the treatment of obesity. Obesity 17: 30–39. [DOI] [PubMed] [Google Scholar]
- Guh D., Zhang W., Bansback N., Amarsi Z., Birmingham C., Anis A. (2009) The incidence of co-morbidities related to obesity and overweight: a systematic review and meta-analysis. BMC Public Health 9: 88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harder H., Nielsen L., Thi T.D.T., Astrup A. (2004) The effect of liraglutide, a long-acting glucagon-like peptide 1 derivative, on glycaemic control, body composition, and 24-h energy expenditure in people with type 2 diabetes. Diabetes Care 27(8): 1915–1921. [DOI] [PubMed] [Google Scholar]
- Hegedus L., Moses A., Zdravkovic M., Le Thi T., Daniels G. (2011) GLP-1 and calcitonin concentration in humans: lack of evidence of calcitonin release from sequential screening in over 5000 subjects with type 2 diabetes or non-diabetic obese subjects treated with the human GLP-1 analog, liraglutide. J Clin Endocrinol Metab 96: 853–860. [DOI] [PubMed] [Google Scholar]
- Hollander P., Gupta A., Plodkowski R., Greenway F., Bays H., Burns C., et al. (2013) Effects of naltrexone sustained-release/bupropion sustained-release combination therapy on body weight and glycemic parameters in overweight and obese patients with type 2 diabetes. Diabetes Care 36: 4022–4029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holst J.J., Deacon C.F. (2005) Glucagon-like peptide-1 mediates the therapeutic actions of DPP-IV inhibitors. Diabetologia 48(4): 612–615. [DOI] [PubMed] [Google Scholar]
- Horowitz M., Flint A., Jones K., Hindsberger C., Rasmussen M., Kapitza C., et al. (2012) Effect of the once-daily human GLP-1 analogue liraglutide on appetite, energy intake, energy expenditure and gastric emptying in type 2 diabetes. Diabetes Res Clin Pract 97: 258–266. [DOI] [PubMed] [Google Scholar]
- Hunter K., Holscher C. (2012) Drugs developed to treat diabetes, liraglutide and lixisenatide, cross the blood–brain barrier and enhance neurogenesis. BMC Neurosci 13: 33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iepsen E., Lundgren J., Dirksen C., Jensen J., Pedersen O., Hansen T., et al. (2014) Treatment with a GLP-1 receptor agonist diminishes the decrease in free plasma leptin during maintenance of weight loss. Int J Obes 39: 834–841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- James W., Caterson I., Coutinho W., Finer N., van Gaal L., Maggioni A., et al. (2010) Effect of sibutramine on cardiovascular outcomes in overweight and obese subjects. N Eng J Med 363: 905–917. [DOI] [PubMed] [Google Scholar]
- Kastin A., Akerstrom V., Pan W. (2002) Interactions of glucagon-like peptide-1 (GLP-1) with the blood-brain barrier. J Mol Neurosci 18: 7–14. [DOI] [PubMed] [Google Scholar]
- Knudsen L., Nielsen P., Huusfeldt P., Johansen N., Madsen K., Pedersen F., et al. (2000) Potent derivatives of glucagon-like peptide-1 with pharmacokinetic properties suitable for once daily administration. J Med Chem 43: 1664–1669. [DOI] [PubMed] [Google Scholar]
- Kreymann B., Williams G., Ghatei M., Bloom S. (1987) Glucagon-like peptide-1 7–36: a physiological incretin in man. Lancet 330: 1300–1304. [DOI] [PubMed] [Google Scholar]
- Kritchevsky S., Beavers K., Miller M., Shea M., Houston D., Kitzman D., et al. (2015) Intentional weight loss and all-cause mortality: a meta-analysis of randomized clinical trials. PLoS One 10: e0121993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li L., Shen J., Bala M., Busse J., Ebrahim S., Vandvik P., et al. (2014) Incretin treatment and risk of pancreatitis in patients with type 2 diabetes mellitus: systematic review and meta-analysis of randomised and non-randomised studies. BMJ 348: g2366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luque C., Rey J. (2002) The discovery and status of sibutramine as an anti-obesity drug. Eur J Pharmacol 440: 119–128. [DOI] [PubMed] [Google Scholar]
- Marathe C.S., Rayner C.K., Jones K.L., Horowitz M. (2011) Effects of GLP-1 and incretin-based therapies on gastrointestinal motor function. Exp Diabetes Res 2011: 279530. doi: 10.1155/2011/279530 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marre M., Shaw J., Brandle M., Bebakar W., Kamaruddin N., Strand J., et al. (2009) 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 26: 268–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matveyenko A., Dry S., Cox H., Moshtaghian A., Gurlo T., Galasso R., et al. (2009) Beneficial endocrine but adverse exocrine effects of sitagliptin in the human islet amyloid polypeptide transgenic rat model of type 2 diabetes. Diabetes 58: 1604–1615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Merchenthaler I., Lane M., Shughrue P. (1999) Distribution of pre-pro-glucagon and glucagon-like peptide-1 receptor messenger RNAs in the rat central nervous system. J Comp Neurol 403: 261–280. [DOI] [PubMed] [Google Scholar]
- Morgan E., Dent M. (2010) The Economic Burden of Obesity. Oxford, UK: National Obesity Observatory. [Google Scholar]
- Must A., Spadano J., Coakley E., Field A., Colditz G., Dietz W. (1999) The disease burden associated with overweight and obesity. JAMA 282: 1523–1529. [DOI] [PubMed] [Google Scholar]
- National Institute for Health and Clinical Excellence (2007) Bariatric Surgical Service for the Treatment of People with Severe Obesity. Commissioning Guide. Available at: http://www.nice.org.uk%2Fmedia%2F87F%2F65%2FBariatricSurgeryFINALPlusNewToolUpdates.pdf (accessed 26 November 2015).
- National Institute for Health and Clinical Excellence (2010) NICE Technology Appraisal Guidance [TA203]. Liraglutide for the Treatment of Type 2 Diabetes Mellitus. Available at: http://www.nice.org.uk/guidance/ta203 (accessed 1 May 2015).
- National Institute for Health and Clinical Excellence (2014a) CG189 - Obesity: Identification, Assessment and Management of Overweight and Obesity in Children, Young People and Adults. Available at: http://www.nice.org.uk/guidance/cg189 (accessed 15 April 2015).
- National Institute for Health and Clinical Excellence (2014b) Costing Report: Obesity. Implementing the NICE guideline on obesity (CG189). Available at: https://www.nice.org.uk/guidance/cg189/resources/costing-report-193304845 (accessed 5 December 2015).
- Nauck M. (2013) A critical analysis of the clinical use of incretin-based therapies: the benefits by far outweigh the potential risks. Diabetes Care 36: 2126–2132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nauck M., Frid A., Hermansen K., Shah N., Tankova T., Mitha I., et al. (2009) 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 32: 84–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nauck M., Kemmeries G., Holst J., Meier J. (2011) Rapid tachyphylaxis of the glucagon-like peptide-1 induced deceleration of gastric emptying in humans. Diabetes 60: 1561–1565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Novo Nordisk (2015) Novo Nordisk SCALE Trials Homepage. Available at: http://www.novonordisk-trials.com/WebSite/Search/trialSearchResult.aspx?Command=Search&SortBy=StartDate&SearchTerms=scale (accessed 11 May 2015).
- O’Neill P., Smith S., Weissman N., Fidler M., Sanchez M., Zhang J., et al. (2012) Randomized placebo-controlled clinical trial of Lorcaserin for weight loss in type 2 diabetes mellitus: the BLOOM-DM Study. Obesity 20: 1426–1436. [DOI] [PubMed] [Google Scholar]
- Orskov C., Wettergren A., Holst J. (1996) 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 31: 665–670. [DOI] [PubMed] [Google Scholar]
- Osaka T., Endo M., Yamakawa M., Inoue S. (2005) Energy expenditure by intravenous administration of glucagon-like peptide-1 mediated by the lower brainstem and sympathoadrenal system. Peptides 26: 1623–1631. [DOI] [PubMed] [Google Scholar]
- Padwal R., Rucker D., Li S., Curioni C., Lau D. (2003) Long-term pharmacotherapy for obesity and overweight. Cochrane Database Syst Rev 4: CD004094. [DOI] [PubMed] [Google Scholar]
- Parkes D., Jodka C., Smith P., Nayak S., Rinehart L., Gingerich R., et al. (2001) Pharmacokinetic actions of exendin-4 in the rat: comparison with glucagon-like peptide-1. Drug Dev Res 53: 260–267. [Google Scholar]
- Peeters A., Barendregt J., Willekens F., Mackenbach J., Al Mamun A., Bonneux L. (2003) Obesity in adulthood and its consequences for life expectancy: a life-table analysis. Ann Intern Med 138: 24–32. [DOI] [PubMed] [Google Scholar]
- Picot J., Jones J., Colquitt J., Gospodarevskaya E., Loveman E., Baxter L., et al. (2009) The clinical effectiveness and cost-effectiveness of bariatric (weight loss) surgery for obesity: a systematic review and economic evaluation. Health Technol Assess 13(41): 1–190, 215–357. [DOI] [PubMed] [Google Scholar]
- Pi-Sunyer X., Astrup A., Fujioka K., Greenway F., Halpern A., Krempf M., et al. (2015) A randomized, controlled trial of 3.0mg of liraglutide in weight management. N Eng J Med 373: 11–22. [DOI] [PubMed] [Google Scholar]
- Russell-Jones D., Vaag A., Schmitz O., Sethi B., Lalic N., Antic S., et al. (2009) Liraglutide vs insulin glargine and placebo in combination with metformin and sulfonylurea therapy in type 2 diabetes mellitus (LEAD-5 met+SU): a randomised controlled trial. Diabetologia 52: 2046–2055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Secher A., Jelsing J., Baquero A., Hecksher-Sorensen J., Cowley M., Dalboge L., et al. (2014) The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss. J Clin Invest 124: 4473–4488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shughrue P., Lane M., Mercenthaler I. (1996) Glucagon-like peptide-1 receptor (GLP1-R) mRNA in the rat hypothalamus. Endocrinology 137: 5159–5162. [DOI] [PubMed] [Google Scholar]
- Shyangdan D., Royle P., Clar C., Sharma P., Waugh N., Snaith A. (2011) Glucagon-like peptide analogues for type 2 diabetes mellitus. Cochrane Database Syst Rev CD006423. doi: 10.1002/14651858.CD006423.pub2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simon G., Ludman E., Linde J., Operskalski B., Ichikawa L., Rohde P., et al. (2008) Association between obesity and depression in middle-aged women. Gen Hosp Psychiatry 30: 32–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sjostrom L., Peltonen M., Jacobson P., Sjostrom C., Karason K., Wedel H., et al. (2012) Bariatric surgery and long-term cardiovascular events. JAMA 307: 56–65. [DOI] [PubMed] [Google Scholar]
- Smith S., Weissman N., Anderson C., Sanchez M., Chuang E., Stubbe S., et al. (2010) Multicenter, placebo-controlled trial of lorcaserin for weight management. N Eng J Med 363: 245–256. [DOI] [PubMed] [Google Scholar]
- Soltoft F., Hammer M., Kragh N. (2009) The association of body mass index and health-related quality of life in the general population: data from the 2003 Health Survey of England. Qual Life Res 18: 1293–1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thomsen W., Grottick A., Menzaghi F., Reyes-Saldana H., Espitia S., Yuskin D., et al. (2008) Lorcaserin, a novel selective human 5-hydroxytryptamine2c agonist: in vitro and in vivo pharmacological characterization. J Pharmacol Exp Ther 325: 577–587. [DOI] [PubMed] [Google Scholar]
- Torgerson J., Hauptman J., Boldrin M., Sjostrom L. (2004) A randomized study of orlistat as an adjunct to lifestyle changes for the prevention of type 2 diabetes in obese patients. Diabetes Care 27: 155–161. [DOI] [PubMed] [Google Scholar]
- US Food and Drug Administration Advisory Committee (2007) FDA Briefing Document: Zimulti (Rimonabant) Tablets, 20 mg. Rockville, MD: FDA; Available at: www.fda.gov/ohrms/dockets/ac/07/briefing/2007-4306b1-fda-backgrounder.pdf (accessed 5 December 2015). [Google Scholar]
- Vahl T., Tauchi M., Durler T., Elfers E., Fernandes T., Bitner R., et al. (2007) Glucagon-like peptide-1 (GLP-1) receptors expressed on nerve terminals in the portal vein mediate the effects of endogenous GLP-1 on glucose tolerance in rats. Endocrinology 148: 4965–4973. [DOI] [PubMed] [Google Scholar]
- van Bloemendaal L., Ten Kulve J., la Fleur S., Ijzerman R., Diamant M. (2014) Effects of glucagon-like peptide 1 on appetite and body weight: focus on the CNS. J Endocrinol 221: T1–T16. [DOI] [PubMed] [Google Scholar]
- van Gaal L., Rissanen A., Scheen A., Ziegler O., Rossner S. (2005) Effects of the cannabinoid-1 receptor blocker rimonabant on weight reduction and cardiovascular risk factors in overweight patients: 1-year experience from the RIO-Europe study. Lancet 365: 1389–1397. [DOI] [PubMed] [Google Scholar]
- Wadden T., Foreyt J., Foster G., Hill J., Klein S., O’Neill P., et al. (2010) Weight loss with naltrexone SR/bupropion SR combination therapy as an adjunct to behaviour modification: the COR-BMOD trial. Obesity 19: 110–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wadden T., Hollander P., Klein S., Niswender K., Woo V., Hale P., et al. (2013) Weight maintenance and additional weight loss with liraglutide after low-calorie-diet-induced weight loss: the SCALE Mainte-nance randomized study. Int J Obes 37: 1443–1451. [DOI] [PubMed] [Google Scholar]
- Wang T., Wang F., Gou Z., Tang H., Li C., Shi L., et al. (2015) Using real-world data to evaluate the association of incretin-based therapies with risk of acute pancreatitis: a meta-analysis of 1,324,515 patients from observational studies. Diabetes Obes Metab 17: 32–41. [DOI] [PubMed] [Google Scholar]
- Weintraub M., Sundaresan P., Madan M., Schuster B., Balder A., Lasagna L., et al. (1992) Long-term weight control study I (weeks 0 to 34): the enhancement of behaviour modification, caloric restriction and fenfluramine plus phentermine versus placebo. Clin Pharmacol Ther 51: 586–594. [DOI] [PubMed] [Google Scholar]
- Wing R., Lang W., Wadden T., Safford M., Knowler W., Bertoni A., et al. (2011) Benefits of modest weight loss in improving cardiovascular risk factors in overweight and obese individuals with type 2 diabetes. Diabetes Care 34: 1481–1486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- World Health Organization (2015) World Health Organization Factsheet No. 311. Available at: http://www.who.int/mediacentre/factsheets/fs311/en (accessed 16 September 2015).
- Young A., Gedulin B., Bhavsar S., Bodkin N., Jodka C., Hansen B., et al. (1999) Glucose-lowering and insulin-sensitizing actions of exendin-4: studies in obese diabetic (ob/ob, db/db) mice, diabetic fatty Zucker rats, and diabetic rhesus monkeys (Macaca mulatta). Diabetes 48: 1026–1034. [DOI] [PubMed] [Google Scholar]
- Zinman B., Gerich J., Buse J., Lewin A., Schwartz S., Raskin P., et al. (2009) 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 32: 1224–1230. [DOI] [PMC free article] [PubMed] [Google Scholar]