Obesity is a growing global health concern, associated with a number of important comorbid conditions. It increases the risk of diabetes and contributes to development of cardiovascular disease. While the benefits of weight loss are well established, weight reduction remains a difficult-to-reach goal in overweight and obese individuals due to several metabolic and psychological factors. For many patients, lifestyle intervention is insufficient to achieve long-term weight loss, and additional options, such as pharmacotherapy, need to be considered. Besides the challenging enterprise of weight reduction, weight maintenance remains an even more crucial and outcome-determining aspect of weight management. This article focuses on the potential of currently available pharmacological strategies to support weight loss and maintenance goals in individuals at risk. Two pharmacotherapy types are considered: those developed primarily to induce weight loss and those developed primarily for blood glucose control that have a favorable effect on body weight. Finally, the potential of very low- and low-calorie diets combined with pharmacotherapy and pharmacological combination therapies are discussed, as well as emerging approaches in development.

Obesity—in particular, abdominal obesity—is a major driver in the development of diabetes and cardiovascular disease (1,2). Weight reduction and maintenance, therefore, are key therapeutic goals in both the management of obesity and its associated comorbidities, type 2 diabetes in particular (3). Unfortunately, several weight loss–induced hormonal changes encourage weight regain and counteract maintained weight loss (4). In the short-term, diet-induced weight loss increases orexigenic hormones ghrelin and gastric inhibitory polypeptide (GIP) and decreases anorexigenic hormones leptin, peptide YY, cholecystokinin, amylin, and glucagon-like peptide 1 (GLP-1) (4,5). As a result, patients feel an increased level of hunger, reduced satiety, and an increased desire to eat (4). These changes persist for a long time, even after weight has stabilized or increased again. In addition, resting metabolic rate and energy expenditure are reduced after diet-induced weight loss—an effect that is also mediated by leptin (6).

Moderate weight reduction of ∼4 kg combined with intensive lifestyle intervention has been shown to reduce the incidence of diabetes by 58% in obese patients in a prediabetes state (7). For individuals with diabetes, large-scale studies have shown that a loss of 5–10% of body weight with increased physical activity can improve fitness, improve cardiovascular disease risk factors, reduce HbA1c levels, and decrease use of diabetes, hypertension, and lipid-lowering medications (8,9). Additional benefits of weight loss might include reduction of depression symptoms and reduced severity of obstructive sleep apnea (10,11). Greater clinical improvements are observed with greater weight loss (9).

Guidelines recommend lifestyle modifications as the foundation of weight loss. Although intensive lifestyle intervention produces clinically beneficial weight loss for many patients, the reality is that intensive lifestyle intervention is difficult to achieve and maintain over the long-term for most patients. Even in an “optimal” clinical trial setting such as Look AHEAD (Action for Health in Diabetes), a third of all patients were unable to achieve at least 5% weight loss after 1 year (12). Most individuals tend to lose weight over a period of 4–6 months and lose 4–10% of baseline weight before experiencing a plateau in weight loss, generally followed by a weight regain (13). Long-term weight loss—in some studies defined as maintaining 75% of initial weight loss (14)—is still difficult to achieve for many patients, and additional options, such as pharmacotherapy with or without very low-calorie diet (VLCD), should be considered for patients who cannot attain sufficient weight loss with lifestyle modification only.

Antiobesity Pharmacotherapies

While several antiobesity agents have been withdrawn from the market due to safety concerns, five are now available in the U.S.—orlistat, lorcaserin, phentermine/topiramate, naltrexone/buprion, and liraglutide 3.0 mg (Table 1)—for chronic weight management. Only orlistat is currently available in Europe, with liraglutide 3.0 mg and naltrexone/buprion having recently received approval from the European Medicines Agency as well. These pharmacotherapies have been demonstrated to show efficacy in weight reduction both in obese patients with comorbidities and in obese patients without comorbidities (1518).

Table 1

Antiobesity therapies currently approved for chronic weight management

DrugMOAHow MOA leads to weight loss
Orlistat • Inhibits gastrointestinal and pancreatic lipases • Prevents absorption of dietary fat 
Lorcaserin • Selectively stimulates 5HT2C • Promotes feelings of satiety and regulates appetite 
Phentermine plus topiramate • Phentermine acts on hypothalamus to stimulate norepinephrine release from adrenal glands
• Topiramate acts on multiple cellular targets as an antiepileptic agent 
• Promotes feelings of satiety and regulates appetite
• The precise mechanisms by which phentermine plus topiramate produces weight loss are unknown 
Naltrexone sustained release plus bupropion sustained release • Increases levels of dopamine and POMC neuronal activity
• Blocks opioid receptors on POMC neurons, preventing feedback inhibition of these neurons and further increasing POMC activity 
• Suppresses appetite
• Increases secretion of melanocortins, which mediate anorectic effects and regulate energy balance 
Liraglutide (3.0 mg) • Binds and activates the human GLP-1 receptor
• Enhances glucose-dependent insulin secretion by the pancreatic β-cell
• Increases cAMP leading to insulin release in the presence of elevated glucose concentrations
• Increases satiety 
• Weight loss owing to inhibition of gastric emptying
• Decreased calorie ingestion through CNS
• Reduced acid secretion 
DrugMOAHow MOA leads to weight loss
Orlistat • Inhibits gastrointestinal and pancreatic lipases • Prevents absorption of dietary fat 
Lorcaserin • Selectively stimulates 5HT2C • Promotes feelings of satiety and regulates appetite 
Phentermine plus topiramate • Phentermine acts on hypothalamus to stimulate norepinephrine release from adrenal glands
• Topiramate acts on multiple cellular targets as an antiepileptic agent 
• Promotes feelings of satiety and regulates appetite
• The precise mechanisms by which phentermine plus topiramate produces weight loss are unknown 
Naltrexone sustained release plus bupropion sustained release • Increases levels of dopamine and POMC neuronal activity
• Blocks opioid receptors on POMC neurons, preventing feedback inhibition of these neurons and further increasing POMC activity 
• Suppresses appetite
• Increases secretion of melanocortins, which mediate anorectic effects and regulate energy balance 
Liraglutide (3.0 mg) • Binds and activates the human GLP-1 receptor
• Enhances glucose-dependent insulin secretion by the pancreatic β-cell
• Increases cAMP leading to insulin release in the presence of elevated glucose concentrations
• Increases satiety 
• Weight loss owing to inhibition of gastric emptying
• Decreased calorie ingestion through CNS
• Reduced acid secretion 

CNS, central nervous system; MOA, mechanism of action; POMC, proopiomelanocortin.

Orlistat

Orlistat functions as an antiobesity agent by inhibiting gastrointestinal lipases, thereby reducing absorption of dietary fat (19). Orlistat is indicated for obesity management, including weight loss and weight maintenance, when used in conjunction with a reduced-calorie diet, and for reduction of the risk of weight regain after prior weight loss (20). In the XENical in the Prevention of Diabetes in Obese Subjects (XENDOS) trial, obese subjects in the placebo group who adhered to a hypocaloric diet and increased physical exercise regimen lost an average of 6.2 kg after 1 year, whereas those in the orlistat group lost an average of 10.6 kg (21). Orlistat also provided weight loss benefits for patients with type 2 diabetes. After 52 weeks of orlistat treatment (120 mg t.i.d.), in combination with a hypocaloric diet and a weight-management program, obese patients with type 2 diabetes achieved –5.0% reduction in weight from baseline (vs. –1.8% with placebo; P < 0.0001) (15). Retrospective analysis of seven studies on orlistat (120 mg t.i.d.) confirms that orlistat-treated patients had significantly greater decreases in body weight than the placebo group (–3.77 vs. –1.42 kg, P < 0.0001). In addition, compared with placebo, more than twice as many orlistat 120 mg–treated patients lost ≥5% of baseline body weight (34.8 vs. 14.1%; P < 0.0001) or lost ≥10% of baseline body weight (9.7 vs. 3.7%; P < 0.0001) (22). Favorable initial response to orlistat has been shown to be a good prognostic indicator for long-term weight loss. Orlistat was generally well tolerated; gastrointestinal effects were the most commonly reported adverse events (AEs) in 20–30% of patients, but all events were considered either mild or moderate (21).

Lorcaserin

Lorcaserin is a selective small-molecule agonist of the 5-hydroxytryptamine 2C serotonin receptor (5HT2C), which regulates mechanisms related to satiety, ingestive behavior, glucose tolerance, and hepatic insulin sensitivity (16). Unlike previously available antiobesity agents, lorcaserin has low affinity for the 5HT2B receptor subtype, whose activation has been linked to the development of valvular heart disease (17). Lorcaserin is indicated as an adjunct to a reduced-calorie diet and increased physical activity for chronic weight management in adults with an initial BMI of ≥30 kg/m2 (obese) or ≥27 kg/m2 in the presence of at least one weight-related comorbid condition, such as hypertension, dyslipidemia, or type 2 diabetes (17). In overweight or obese patients without diabetes, lorcaserin treatment provided −5.81% reduction from baseline body weight after 1 year (vs. −2.16% with placebo; P < 0.001) (18). The Behavioral Modification and Lorcaserin Second Study for Obesity Management (BLOSSOM) compared lorcaserin 10 mg b.i.d. versus once daily (QD) versus placebo. Lorcaserin b.i.d. was associated with significantly greater weight loss than lorcaserin QD (−5.8 kg vs. −4.7 kg). Significantly more patients receiving lorcaserin BID and lorcaserin QD lost at least 5% body weight at 1 year (47.2% and 40.2%) than in the placebo group (25.0%). Similarly, significantly more patients achieved at least 10% weight loss in the lorcaserin groups compared with placebo (22.6% b.i.d. vs. 17.4% QD vs. 9.7% placebo). In a phase 3 study of subjects with type 2 diabetes (treated either with metformin or a sulfonylurea), lorcaserin treatment resulted in mean weight changes of −4.5% (b.i.d.) and −5.0% (QD) compared with −1.5% with placebo at week 52 (16). No evidence of increased depression, suicidal thoughts, or echocardiogram-detected valvular regurgitations was found in the lorcaserin treatment arms. Overall, the most common AEs with lorcaserin were headache, back pain, nasopharyngitis, and nausea.

Phentermine Plus Topiramate

Phentermine is a norepinephrine- and dopamine-releasing agent (with a lower effect with dopamine vs. norepinephrine) approved for the short-term treatment of obesity (23). Topiramate, a registered antiepileptic drug, has several pharmacological mechanisms of action. The exact mechanisms by which the combination phentermine plus topiramate produces weight loss still remain unknown. A phase 3 study examined the efficacy of the combination of phentermine 7.5 mg/topiramate 46.0 mg or phentermine 15.0 mg/topiramate 92.0 mg on weight loss after 56 weeks (24). Patients who completed 1 year of treatment had significantly (P < 0.0001) higher absolute body weight change of –9.9 kg (–9.6%) with phentermine 7.5 mg plus topiramate 46.0 mg vs. –12.9 kg (12.4%) with phentermine 15.0 mg plus topiramate 92.0 mg compared with –1.8 kg (1.6%) in the placebo group (24). Patients with type 2 diabetes (a subgroup of 388 subjects in this study) achieved weight reductions of −6.8% and −8.8% with phentermine 7.5 mg/topiramate 46.0 mg and phentermine 15.0 mg/topiramate 92.0 mg, respectively (vs. placebo, −1.9%). Phentermine/topiramate was well tolerated; constipation, paresthesia, and dry mouth were the most commonly reported treatment-emergent AEs (25). However, the U.S. Food and Drug Administration (FDA) has required a Risk Evaluation and Mitigation Strategy for phentermine/topiramate to educate prescribers and patients on the increased risk of orofacial clefts in infants exposed to phentermine/topiramate during the first trimester of pregnancy (26).

Naltrexone Sustained Release Plus Bupropion Sustained Release

Naltrexone is an opioid receptor antagonist, while bupropion is a norepinephrine and dopamine reuptake inhibitor (27); the combination increases proopiomelanocortin neuronal firing, which may have anorectic effects. The combination provides greater weight loss than monotherapy with either agent or placebo (27). Multiple 56-week multicenter, double-blind, placebo-controlled trials (CONTRAVE Obesity Research, or COR-I, COR-II, COR-BMOD, and COR-Diabetes) were conducted to evaluate the effect of bupropion/naltrexone in conjunction with lifestyle modification in a placebo-controlled cohort. In COR-I and COR-II, mean change in body weight after 1 year was –6.1% and −6.4% in the naltrexone 32 mg plus bupropion (NB) group, respectively, and –1.3% and −1.2% in the placebo group (28,29). In COR-I, 48% of participants assigned to NB had a decrease in body weight of ≥5% compared with 16% assigned to placebo (28). In COR-Diabetes, patients on bupropion/naltrexone lost significantly more weight compared with those on placebo (−5.0% vs. −1.8%). In COR-BMOD, intensive behavior modification was added to all treatment arms, resulting in better results, with a weight loss of 9.3% in the NB group vs. 5.1% in the placebo group. Compared with placebo, treatment with NB was associated with a higher incidence of nausea, constipation, and vomiting but was not associated with increased depression, suicidal thoughts, or hypoglycemia. While NB may represent a novel pharmacological approach for the treatment of obesity, further studies are required to assess its effects on cardiovascular outcomes, as systolic blood pressure and pulse rate have been found to be higher with NB than with placebo (17).

Liraglutide (3.0 mg)

The underlying mechanisms that mediate the effects of weight reduction of liraglutide are most probably a combination of effects on the gastrointestinal tract and the brain. Native GLP-1 suppresses appetite and energy intake in both normal-weight and obese individuals, as well as in people with type 2 diabetes, and delays gastric emptying. GLP-1 receptors are expressed in several brain stem nuclei involved in appetite regulation, and subcutaneously administered liraglutide might also reach these sites (30).The weight-reducing effect of liraglutide was already noted in studies using the lower doses of 1.2 and 1.8 mg, indicated for the treatment of type 2 diabetes. Liraglutide (3.0 mg QD) has been shown to provide weight-reduction benefits for obese patients; after 20 weeks, the placebo-subtracted reduction in weight from baseline with liraglutide (3.0 mg) treatment was –4.4 kg (P = 0.003) (30,31). The SCALE maintenance trial assessed the efficacity of liraglutide in maintaining weight loss achieved with a low-calorie diet after 1 year. After a diet-induced weight loss, subjects on liraglutide lost an additional 6.2% vs. 0.2% with placebo. Fifty percent of subjects on liraglutide achieved ≥5% weight loss vs. 21.8% in the placebo group. Results were similar in patients with type 2 diabetes (SCALE Diabetes trial) and prediabetes (SCALE Obesity and Prediabetes) (32,33). The weight reductions observed with liraglutide (3.0 mg QD) primarily result from reductions in fat mass and body fat percentage (including visceral fat) rather than lean tissue mass (31,34). In the SCALE Obesity and Prediabetes trial, there seemed to be a slightly reduced weight loss response in patients with a BMI >40 kg/m2, but this was not confirmed in the SCALE Diabetes trial (32,33). Similar to liraglutide 1.8 mg treatment in patients with diabetes, the most common AEs with liraglutide 3.0 mg treatment were gastrointestinal, which is also consistent with the known physiological effects of GLP-1 receptor agonists (31). Of note, heart rate was slightly but significantly increased in liraglutide-treated patients versus placebo by 4 bpm (31). In the liraglutide group, three individuals were diagnosed with a malignancy (breast, colon, and prostate) versus no individuals in the placebo group.

Safety Aspects

In the selection of the best antiobesity drug for a specific patient, the aspects of possible side effects, contraindications, and drug interactions should be taken into consideration. An overview is provided in Table 2. Recent safety concerns about an increased risk of major adverse cardiac events have led either to definitive withdrawal of existing antiobesity medications or a lack of new treatments being approved (35). Assessment of cardiovascular safety has now emerged as a major consideration for all new antiobesity and glucose-lowering agents under current review by the FDA (36). So far, none of the current FDA-approved medications have had a cardiovascular outcome trial. Given the significant need for effective and safe weight loss medication, it is perhaps not surprising that many more antiobesity therapies are in development (see the recent article by Rodgers et al. [20]; these newer therapies are also overviewed below). The potential of these therapies in patients with type 2 diabetes, as well as their cardiovascular safety, will need to be established.

Table 2

Overview of common side effects, contraindications, and drug interactions

Common side effectsContraindicationsDrug interactions
Orlistat Steatorrhea, oily rectal leakage, abdominal pain, headache Pregnancy, malabsorption, cholestasis, deficiency in fat soluble vitamins Cyclosporine, L-thyroxine, amiodarone, warfarine 
Lorcaserin Headache, back pain, nasopharyngitis, nausea Pregnancy, renal insufficiency, valvular heart disease, depression Serotonergic drugs (serotonin syndrome), cabergoline 
Phentermine/topiramate Increased heart rate, paresthesia, headache, insomnia, xerostomia, constipation, nasopharyngitis Pregnancy, hyperthyroidism, glaucoma Monoamine oxidase inhibitors, SSRI inhibitors, amitriptyline, lithium, opioids, contraceptives, alcohol 
Naltrexone/bupropion Headache, sleep disorder, nausea, constipation, vomiting Pregnancy; use of opioid, opiate agonist, or partial agonist; acute withdrawal of opioid alcohol, benzodiazepines, barbiturates, or antiepileptics; seizure disorder; bulimia nervosa Alcohol, anti-Parkinson drugs, monoamine oxidase inhibitors, tamoxifen, pimozide, thioridazine 
Liraglutide Increased heart rate, headache, nausea/vomiting, diarrhea/constipation Pregnancy, multiple endocrine neoplasia syndrome type 2  
Common side effectsContraindicationsDrug interactions
Orlistat Steatorrhea, oily rectal leakage, abdominal pain, headache Pregnancy, malabsorption, cholestasis, deficiency in fat soluble vitamins Cyclosporine, L-thyroxine, amiodarone, warfarine 
Lorcaserin Headache, back pain, nasopharyngitis, nausea Pregnancy, renal insufficiency, valvular heart disease, depression Serotonergic drugs (serotonin syndrome), cabergoline 
Phentermine/topiramate Increased heart rate, paresthesia, headache, insomnia, xerostomia, constipation, nasopharyngitis Pregnancy, hyperthyroidism, glaucoma Monoamine oxidase inhibitors, SSRI inhibitors, amitriptyline, lithium, opioids, contraceptives, alcohol 
Naltrexone/bupropion Headache, sleep disorder, nausea, constipation, vomiting Pregnancy; use of opioid, opiate agonist, or partial agonist; acute withdrawal of opioid alcohol, benzodiazepines, barbiturates, or antiepileptics; seizure disorder; bulimia nervosa Alcohol, anti-Parkinson drugs, monoamine oxidase inhibitors, tamoxifen, pimozide, thioridazine 
Liraglutide Increased heart rate, headache, nausea/vomiting, diarrhea/constipation Pregnancy, multiple endocrine neoplasia syndrome type 2  

SSRI, selective serotonin reuptake inhibitors.

Weight Maintenance

As obesity is considered to be a chronic disease, its treatment necessitates enduring efforts in calorie restriction and increased physical activity. After diet-induced weight loss, several counterregulatory neurohormonal mechanisms aim at maintaining and even reexpanding fat mass (37). Given the importance of obesity and its associated risks as a public health problem, the limited evidence about long-term weight maintenance still remains problematic. Most patients unfortunately regain weight after a weight-management program, whereas individuals continuing in a clinical trial can maintain a weight loss of ∼3% of initial body weight up to 5 years. As identified in the Look AHEAD study, not only greater initial weight losses but also the sustained weight loss during the first year are predictive factors for successful weight loss after 4 years (38). Other predictors of weight loss maintenance include the use of low-calorie diets and increased physical activity and self-monitoring (14).

Maintaining a considerable weight loss requires substantial behavioral efforts. Among cognitive strategies for weight maintenance that Voils et al. (39) listed in a review are the following items: self-regulatory focus, self-efficacy to maintain weight loss, self-efficacy to recover from a relapse, self-monitoring, and social support derived from social networks. Also, eating frequency has been reported to be higher in weight loss maintainers. In a systematic review, meal-replacement strategies, high-protein diets, and antiobesity drugs were associated with weight loss maintenance, mainly after VLCD/low-calorie diet (37).

Long-term medical therapy as an adjunct to sustain weight loss should be taken into consideration (National Institutes of Health, National Heart, Lung, and Blood Institute). Several of the aforementioned agents have been tested as part of a long-term weight-management plan.

Experience With Former Withdrawn Drugs: Sibutramine and Rimonabant

Apart from the short-term efficacy of sibutramine (38), the Sibutramine Trial of Obesity Reduction and Maintenance (STORM) study indicated a superiority of sibutramine over placebo to maintain at least 80% of initially achieved weight loss (43% in sibutramine group vs. 16% in placebo group) (40). Eventually, sibutramine was withdrawn by the FDA owing to risk of serious cardiovascular events, as shown in the Sibutramine Cardiovascular Outcomes Trial (SCOUT) report.

Rimonabant, a cannabinoid receptor antagonist, significantly increased weight loss at 1 year of treatment (−3.4 kg for 5 mg and −6.6 kg for 20 mg vs. −1.8 kg for placebo) (41), resulting in a significant increase in patients achieving 5% and 10% weight loss. At 2 years of treatment, the results were comparable with a mean 5.5 kg and 2.9 kg weight reduction for the rimonabant 20 mg and 5 mg group vs. 1.2 kg in the placebo group (42). After 2 years, the advantageous cardiovascular effects were maintained. Due to concerns about psychiatric side effects (mood disturbances and suicidality), rimonabant was withdrawn by the European Medicines Agency in 2009.

Orlistat

Several placebo-controlled randomized trials have investigated the long-term effectiveness of orlistat in weight reduction. After 2 years, 57% of subjects on orlistat 120 mg maintained a weight loss of ≥5% vs. 37.4% in the placebo group (43). In another 2-year study, use of orlistat induced a mean weight loss of 7.6% (orlistat 120 mg) and 6.8% (orlistat 60 mg) vs. 4.5% for placebo after 2 years of treatment (44). In another study, in which orlistat was added after a VLCD-induced weight loss of ≥5%, use of orlistat was associated with maintenance of an extra 2.4-kg weight loss for up to 3 years in obese subjects (45). In a 4-year study, in patients who completed 4 full years of treatment, 52.8% in the treated group and 37.3% in the placebo group lost ≥5% of baseline body weight and 26.2% and 15.6%, respectively, lost ≥10% of baseline body weight (21). This resulted in a greater reduction in the incidence of type 2 diabetes over lifestyle changes alone (6.2% vs. 9.0% [–37.3% reduction], P = 0.0032) (21).

Lorcaserin

In the BLOOM trial, 3,182 adults who were overweight or obese received lorcaserin 10 mg b.i.d. or placebo for 52 weeks in conjunction with diet and exercise. At week 52, all subjects were rerandomized to either placebo or lorcaserin for an additional year. At 1 year, the average weight loss was 5.8 ± 0.2 kg with lorcaserin and 2.2 ± 0.1 kg with placebo, resulting in 47.5% of patients in the lorcaserin group and 20.3% in the placebo group losing 5% or more body weight. Among patients in the lorcaserin group who had weight loss of 5% or more at year 1, the loss was maintained in a greater proportion of patients who continued to receive lorcaserin in year 2 than in those who were reassigned to receive placebo (67.9% vs. 50.3%) (46).

Phentermine/Topiramate

The 52-week CONQUER trial randomized obese patients to placebo, midtreatment dose (7.5/46 mg), or maximum treatment dose (15/92 mg). At 56 weeks, change in body weight was −10.2 kg, –8.1 kg, and –1.4 kg in the patients assigned to maximum, midtreatment, and placebo, respectively. Five percent and 10% weight loss was achieved in 70% and 48% in the maximum dose group, 62% and 37% in the midtreatment group, and 21% and 7% in the placebo group (47), respectively. An extension of this trial (SEQUEL) showed persistent favorable effects after 2 years, with −10.7%, −9.3%, and −2.2% for the maximum dose, midtreatment, and placebo group, respectively (25). Phentermine/topiramate was clearly effective in all BMI categories. However, a significant treatment effect by baseline BMI was observed. Among all subjects with class II obesity or greater at baseline (i.e., BMI ≥35 kg/m2), mean weight losses after 2 years were significantly greater for phentermine/topiramate compared with placebo. In a prespecified subgroup analysis of weight loss in subjects with type 2 diabetes at baseline, the phentermine/topiramate–treated subjects also showed greater weight loss compared with placebo-treated subjects. At week 108, subjects with type 2 diabetes in the placebo group (n = 55) lost 2.0% of body weight, whereas subjects with type 2 diabetes in both the 7.5/46 mg (n = 26) and 15/92 mg (n = 64) groups lost 9.0% of body weight.

Liraglutide

Liraglutide, when given in a dose of 2.4 or 3.0 mg daily during 2 years, results in a mean weight loss of 5.8 kg, which was significantly greater than with orlistat and placebo. At year 2, significantly more subjects on liraglutide 2.4/3.0 mg than on orlistat lost ≥5 and ≥10% weight. Of the 64% who achieved ≥5% weight loss with liraglutide 2.4/3.0 mg at year 1, >85% maintained this at year 2 (31). The use of a low-calorie diet in a recent liraglutide approach indicated how effective this injectable GLP-1 receptor agonist can be (48).

VLCD and Low-Calorie Diets

Effects of different maintenance strategies after VLCDs have been evaluated in several trials, including the use of antiobesity agents.

VLCD can be considered a critical enhancer of weight management with antiobesity agents. Given the average weight loss of ∼8–10 kg over 1 year with the majority of antiobesity agents, other additional approaches for more appropriate and optimized weight loss and maintenance have been studied. The use of a VLCD (500–800 kcal/day) is an interesting consideration as initial start-up therapy for such an approach. The use of a VLCD prior to pharmacological treatment has shown excellent results, with successes beyond 10 kg weight loss and with a maintenance lasting for at least 1 year (38).

In the Apfelbaum study, >90% of patients could maintain >50% of the weight loss that was achieved (38). Astrup and colleagues (49) could report a similar successful 52-week outcome, using topiramate after an initial low-calorie approach.

Also with orlistat, in an analysis on maintenance in subjects with abdominal obesity, Richelsen et al. (45) could demonstrate that an extra weight loss of 2.4 kg could be achieved after VLCD and maintained for up to 3 years.

In a crossover study with the lipase inhibitor orlistat, the use of this pharmacological agent on top of a low-calorie diet after an initial 52 weeks of hypocaloric dieting could maintain body weight ∼7–8% up to 120 weeks (42). As to changes in metabolic risk factors, the orlistat patients tended to show better outcomes (45). Using sibutramine (STORM trial), James et al. (40) found a significant drug effect on weight loss maintenance after a 600 kcal–deficit weight loss program. In that same STORM trial, van Baak et al. (50) could demonstrate that on top of a pharmacological treatment, leisure activity could contribute to weight maintenance as well. Similarly, the use of topiramate has shown results comparable with those of sibutramine, confirming the useful strategy of VLCD followed by pharmacotherapy, irrespective of the mechanism of action of the respective agent studied. Also, with lorcaserin similar results could be confirmed in a crossover design.

Using an injectable approach with liraglutide 3 mg, Wadden et al. (48) confirmed the previous findings that treatment with a GLP-1 receptor agonist liraglutide at high doses with lifestyle support could significantly maintain weight loss (>5% of initial body weight) achieved by a low-calorie diet. An almost 6-kg placebo-subtracted weight loss could be observed at week 56, resulting in a total weight loss of ∼12 kg after 1 year.

As was previously consistently shown with other pharmacological agents (20), liraglutide could enhance the benefit of lifestyle intervention on body weight—beyond 10 kg—and additional reduction in cardiovascular disease risk factors.

Future Prospects in Clinical and Preclinical Development

As our knowledge of the physiology of appetite and energy homeostasis improves, so too will our ability to understand how therapies might be combined to provide effective weight management. This is particularly necessary in patients with type 2 diabetes (51,52).

A new class of antidiabetes therapies that show potential for weight loss (although they are not indicated for weight loss per se) are the sodium–glucose cotransporter (SGLT)-2 inhibitors (53). In individuals with type 2 diabetes, SGLT-2 expression is increased in the renal proximal tubular cells, leading to increased renal glucose reabsorption, which ultimately aggravates hyperglycemia. SGLT-2 inhibitors reduce blood glucose, mainly through increased glycosuria, although indirect mechanisms have also been reported (51,54). Dapagliflozin therapy is associated with modest reduction in body weight and shows a favorable weight profile in comparison with both placebo and metformin. Dapagliflozin-induced body weight has been shown to occur through reductions in fat mass, visceral fat, and subcutaneous fat (55). Also, canagliflozin has demonstrated dose-related reductions in baseline body weight in individuals with diabetes (56,57). Canagliflozin (50–300 mg) has also been reported to have beneficial weight effects in overweight or obese individuals without diabetes (58). While increased glucose excretion contributes to the weight loss observed with SGLT-2 inhibitor treatment, weight reduction is often limited to <4 kg after even 52 weeks of treatment (54). This attenuation of weight loss may occur because SGLT-2 inhibitor–induced glycosuria is accompanied by compensatory hyperphagia, as demonstrated in animal studies (53) and suggested by human studies (59). A potential future option is a SGLT-2 inhibitor/GLP-1 receptor agonist combination (59). The effect of GLP-1 receptor agonists on satiety may weaken the compensatory “overeating” mechanism observed with SGLT-2 inhibition and enhance weight loss by SGLT-2 inhibitors (60). In the CANagliflozin cardioVascular Assessment Study (CANVAS), addition of canagliflozin 100 mg or 300 mg to a background therapy of dipeptidyl peptidase-4 inhibitor or GLP-1 receptor agonist resulted in a weight loss varying between 2.3 and 3.2% after 18 weeks (61). Other combination trials with SGLT-2 inhibitors as background therapy are also underway.

Given the role of leptin and amylin in controlling food intake and energy expenditure and the role of incretins (GLP-1) in glucose and weight control (62,63), it is no surprise that many of the therapies in preclinical development involve these different hormones. Therapies that are currently being studied include the following.

Peptide Hormone Combination Therapies

Because results with recombinant human leptin or metreleptin (human leptin analog) have been disappointing in reducing weight for obese patients with type 2 diabetes (62), approaches are now focused on leptin-related synthetic peptides, such as leptin receptor antagonists or leptin-related synthetic peptide analogs or mimetics, and leptin combination therapies (64). Initial preclinical as well as clinical data suggest that leptin and amylin—two hormones involved in the control of satiety—have additive effects (64). In a proof-of-concept randomized controlled trial in overweight/obese subjects, it was shown that combination treatment with pramlintide/metreleptin led to a significant earlier, sustained, and greater weight loss than treatment with pramlintide or metreleptin alone (65). However, a subsequent trial was recently halted because of safety concerns.

PEGylated (PEG) leptin, along with PEG–GLP-1/glucagon, may be another potential combination therapy option (66). This combination is an intriguing potential option, as preclinical data indicate that PEG-leptin and PEG–GLP-1/glucagon coagonism can restore leptin responsiveness, which is often reduced when leptin is used alone. Responsiveness to leptin is associated with decreased food intake, improved glucose tolerance, and insulin sensitivity, as well as decreased triglycerides and lower plasma cholesterol. These may be the contributing factors that lead to the weight loss observed with leptin/GLP-1/glucagon coagonism (66).

Another potential therapy is the combination of amylin analogs and GLP-1 receptor agonists. As both agents can slow gastric emptying, it is possible that these two agents combined may have synergistic effects, but the gastrointestinal tolerance should be evaluated.

Unimolecular Dual or Triple Incretin Receptor Agonists

Another incretin pathway compound in early-stage development is a peptide that acts as an agonist at both the GLP-1 and GIP receptors (67). Although GIP is orexogenic, GIP-overexpressing mice even show a reduced tendency to develop diet-induced obesity, contradicting the notion that the orexogenic effect of GIP results in an obesogenic effect (68). Dipeptidyl peptidase 4 inhibitors increase endogenous levels of GLP-1 and GIP, resulting in better glycemic control without weight gain. A preclinical study indicates that dual agonism of GLP-1 and GIP has the potential to enhance the antiobesity effects observed with monoagonism, as it affects both adiposity-induced insulin resistance and pancreatic insulin deficiency. Simultaneous administration of a GLP-1 agonist and a peptide YY agonist resulted in decreased food intake in humans that exceeded the effects of the individual compounds (69). Likewise, coadministration of cholecystokinin with either a GLP-1 agonist or amylin produced greater than additive weight loss in rats (70); in a recent study in rodents, a new monomeric peptide triagonist simultaneously acting at three key metabolically related peptide hormone receptors (GLP-1, GIP, and glucagon) was found to provide additional glucose control and weight-reducing benefits over dual coagonism (71). Extensive clinical investigation into the efficacy and safety of coagonist therapy for the treatment of patients with obesity and type 2 diabetes is warranted.

Conclusions

Benefits of weight reduction are irrefutable, and lifestyle interventions aimed at prompting weight loss remain the cornerstone of obesity treatment, but the majority of patients are unable to achieve 10-kg weight loss targets and/or maintenance. Clearly, a shift in paradigm of weight loss management is needed, with the acceptance of pharmacotherapy as a valuable addition. Several interesting drugs are currently approved, but long-term data on efficacy and safety, in particular, cardiovascular safety, remain warranted. Additionally, health care practitioners should consider the weight effects of pharmacotherapy in the management of obesity-related comorbidities and consider weight-neutral or weight-reducing medications that can complement the patient’s desire for a healthier lifestyle (60).

This publication is based on the presentations at the 5th World Congress on Controversies to Consensus in Diabetes, Obesity and Hypertension (CODHy). The Congress and the publication of this supplement were made possible in part by unrestricted educational grants from AstraZeneca.

Funding. L.V.G. received grant support from National Research Funds, Belgium, and also received grant support for hepatic research from the European Union consortium (Hepadip and Resolve consortia).

Duality of Interest. L.V.G. is, or has been, a member of the advisory boards and speakers bureaus of AstraZeneca/Bristol-Myers Squibb, Boehringer Ingelheim, Eli Lilly, Janssen, Johnson & Johnson, Merck Sharp & Dohme, Novo Nordisk, and Sanofi (in the period 2010–2014). No other potential conflicts of interest relevant to this article were reported.

1.
Van Gaal
LF
,
Mertens
IL
,
De Block
CE
.
Mechanisms linking obesity with cardiovascular disease
.
Nature
2006
;
444
:
875
880
[PubMed]
2.
Van Gaal
LF
,
De Block
CE
.
Bariatric surgery to treat type 2 diabetes: what is the recent evidence?
Curr Opin Endocrinol Diabetes Obes
2012
;
19
:
352
358
[PubMed]
3.
Inzucchi
SE
,
Bergenstal
RM
,
Buse
JB
, et al
.
Management of hyperglycemia in type 2 diabetes, 2015: a patient-centered approach: update to a position statement of the American Diabetes Association and the European Association for the Study of Diabetes
.
Diabetes Care
2015
;
38
:
140
149
[PubMed]
4.
Sumithran
P
,
Prendergast
LA
,
Delbridge
E
, et al
.
Long-term persistence of hormonal adaptations to weight loss
.
N Engl J Med
2011
;
365
:
1597
1604
[PubMed]
5.
de Luis
DA
,
Gonzalez Sagrado
M
,
Conde
R
,
Aller
R
,
Izaola
O
.
Decreased basal levels of glucagon-like peptide-1 after weight loss in obese subjects
.
Ann Nutr Metab
2007
;
51
:
134
138
[PubMed]
6.
Rosenbaum
M
,
Leibel
RL
.
20 years of leptin: role of leptin in energy homeostasis in humans
.
J Endocrinol
2014
;
223
:
T83
T96
[PubMed]
7.
Knowler
WC
,
Barrett-Connor
E
,
Fowler
SE
, et al.;
Diabetes Prevention Program Research Group
.
Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin
.
N Engl J Med
2002
;
346
:
393
403
[PubMed]
8.
Wing
RR
;
Look AHEAD Research Group
.
Long-term effects of a lifestyle intervention on weight and cardiovascular risk factors in individuals with type 2 diabetes mellitus: four-year results of the Look AHEAD trial
.
Arch Intern Med
2010
;
170
:
1566
1575
[PubMed]
9.
Wing
RR
,
Lang
W
,
Wadden
TA
, et al.;
Look AHEAD Research Group
.
Benefits of modest weight loss in improving cardiovascular risk factors in overweight and obese individuals with type 2 diabetes
.
Diabetes Care
2011
;
34
:
1481
1486
[PubMed]
10.
Rubin
RR
,
Wadden
TA
,
Bahnson
JL
, et al.;
Look AHEAD Research Group
.
Impact of intensive lifestyle intervention on depression and health-related quality of life in type 2 diabetes: the Look AHEAD Trial
.
Diabetes Care
2014
;
37
:
1544
1553
[PubMed]
11.
Foster
GD
,
Borradaile
KE
,
Sanders
MH
, et al.;
Sleep AHEAD Research Group of Look AHEAD Research Group
.
A randomized study on the effect of weight loss on obstructive sleep apnea among obese patients with type 2 diabetes: the Sleep AHEAD study
.
Arch Intern Med
2009
;
169
:
1619
1626
[PubMed]
12.
Look AHEAD Research Group
.
Eight-year weight losses with an intensive lifestyle intervention: the Look AHEAD study
.
Obesity (Silver Spring)
2014
;
22
:
5
13
[PubMed]
13.
Pi-Sunyer
FX
.
Weight loss in type 2 diabetic patients
.
Diabetes Care
2005
;
28
:
1526
1527
[PubMed]
14.
Phelan
S
,
Wing
RR
,
Loria
CM
,
Kim
Y
,
Lewis
CE
.
Prevalence and predictors of weight-loss maintenance in a biracial cohort: results from the coronary artery risk development in young adults study
.
Am J Prev Med
2010
;
39
:
546
554
[PubMed]
15.
Berne
C
;
Orlistat Swedish Type 2 diabetes Study Group
.
A randomized study of orlistat in combination with a weight management programme in obese patients with type 2 diabetes treated with metformin
.
Diabet Med
2005
;
22
:
612
618
[PubMed]
16.
O’Neil
PM
,
Smith
SR
,
Weissman
NJ
, et al
.
Randomized placebo-controlled clinical trial of lorcaserin for weight loss in type 2 diabetes mellitus: the BLOOM-DM study
.
Obesity (Silver Spring)
2012
;
20
:
1426
1436
[PubMed]
17.
Garvey
WT
,
Ryan
DH
,
Bohannon
NJ
, et al
.
Weight-loss therapy in type 2 diabetes: effects of phentermine and topiramate extended release
.
Diabetes Care
2014
;
37
:
3309
3316
[PubMed]
18.
Smith
SR
,
Weissman
NJ
,
Anderson
CM
, et al.;
Behavioral Modification and Lorcaserin for Overweight and Obesity Management (BLOOM) Study Group
.
Multicenter, placebo-controlled trial of lorcaserin for weight management
.
N Engl J Med
2010
;
363
:
245
256
[PubMed]
19.
Derosa
G
,
Cicero
AF
,
D’Angelo
A
,
Fogari
E
,
Maffioli
P
.
Effects of 1-year orlistat treatment compared to placebo on insulin resistance parameters in patients with type 2 diabetes
.
J Clin Pharm Ther
2012
;
37
:
187
195
[PubMed]
20.
Rodgers
RJ
,
Tschöp
MH
,
Wilding
JP
.
Anti-obesity drugs: past, present and future
.
Dis Model Mech
2012
;
5
:
621
626
[PubMed]
21.
Torgerson
JS
,
Hauptman
J
,
Boldrin
MN
,
Sjöström
L
.
XENical in the prevention of Diabetes in Obese Subjects (XENDOS) study: a randomized study of orlistat as an adjunct to lifestyle changes for the prevention of type 2 diabetes in obese patients
.
Diabetes Care
2004
;
27
:
155
161
[PubMed]
22.
Jacob
S
,
Rabbia
M
,
Meier
MK
,
Hauptman
J
.
Orlistat 120 mg improves glycaemic control in type 2 diabetic patients with or without concurrent weight loss
.
Diabetes Obes Metab
2009
;
11
:
361
371
[PubMed]
23.
Wilding
J
,
Van Gaal
L
,
Rissanen
A
,
Vercruysse
F
,
Fitchet
M
;
OBES-002 Study Group
.
A randomized double-blind placebo-controlled study of the long-term efficacy and safety of topiramate in the treatment of obese subjects
.
Int J Obes Relat Metab Disord
2004
;
28
:
1399
1410
[PubMed]
24.
Gadde
KM
,
Allison
DB
,
Ryan
DH
, et al
.
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
2011
;
377
:
1341
1352
[PubMed]
25.
Garvey
WT
,
Ryan
DH
,
Look
M
, et al
.
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
2012
;
95
:
297
308
[PubMed]
26.
U.S. Food and Drug Administration. QSYMIA (phentermine and topiramate extended-release) capsules: risk evaluation and mitigation strategy [article online], 2013. Available from http://www.fda.gov/downloads/drugs/drugsafety/postmarketdrugsafetyinformationforpatientsandproviders/ucm312598.pdf. Accessed 8 September 2014
27.
Katsiki
N
,
Hatzitolios
AI
,
Mikhailidis
DP
.
Naltrexone sustained-release (SR) + bupropion SR combination therapy for the treatment of obesity: ‘a new kid on the block’
?
Ann Med
2011
;
43
:
249
258
[PubMed]
28.
Greenway FL, Fujioka K, Plodkowski RA, Mudaliar S, Guttadauria M, Erickson J, Kim DD, Dunayevich E; COR-I Study Group. 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 2010 21;376:595–605
29.
Apovian
CM
,
Aronne
L
,
Rubino
D
, et al.;
COR-II Study Group
.
A randomized, phase 3 trial of naltrexone SR/bupropion SR on weight and obesity-related risk factors (COR-II)
.
Obesity (Silver Spring)
2013
;
21
:
935
943
[PubMed]
30.
Astrup
A
,
Rössner
S
,
Van Gaal
L
, et al.;
NN8022-1807 Study Group
.
Effects of liraglutide in the treatment of obesity: a randomised, double-blind, placebo-controlled study
.
Lancet
2009
;
374
:
1606
1616
[PubMed]
31.
Astrup
A
,
Carraro
R
,
Finer
N
, et al.;
NN8022-1807 Investigators
.
Safety, tolerability and sustained weight loss over 2 years with the once-daily human GLP-1 analog, liraglutide
.
Int J Obes
2012
;
36
:
843
854
[PubMed]
32.
Davies
MJ
,
Bergenstal
R
,
Bode
B
, et al.;
NN8022-1922 Study Group
.
Efficacy of liraglutide for weight loss among patients with type 2 diabetes: the SCALE diabetes randomized clinical trial
.
JAMA
2015
;
314
:
687
699
33.
Pi-Sunyer
X
,
Astrup
A
,
Fujioka
K
, et al.;
SCALE Obesity and Prediabetes NN8022-1839 Study Group
.
A randomized, controlled trial of 3.0 mg of liraglutide in weight management
.
N Engl J Med
2015
;
373
:
11
22
34.
Jendle
J
,
Nauck
MA
,
Matthews
DR
, et al.;
LEAD-2 and LEAD-3 Study Groups
.
Weight loss with liraglutide, a once-daily human glucagon-like peptide-1 analogue for type 2 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
[PubMed]
35.
Hiatt
WR
,
Goldfine
AB
,
Kaul
S
.
Cardiovascular risk assessment in the development of new drugs for obesity
.
JAMA
2012
;
308
:
1099
1100
[PubMed]
36.
U.S. Food and Drug Administration. Guidance for industry: diabetes mellitus—evaluating cardiovascular risk in new antidiabetic therapies to treat type 2 diabetes [article online], 2008. Available from http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm071627.pdf. Accessed 30 April 2014
37.
Wilson
JL
,
Enriori
PJ
.
A talk between fat tissue, gut, pancreas and brain to control body weight
.
Mol Cell Endocrinol
2015
;
418
:
108
119
[PubMed]
38.
Apfelbaum M, Vague P, Ziegler O, Hanotin C, Thomas F, Leutenegger E. Long-term maintenance of weight loss after a very-low-calorie diet: a randomised blinded trial of the efficacy and tolerability of sibutramine. Am J Med 1999;106:179–184
39.
Voils
CI
,
Gierisch
JM
,
Yancy
WS
 Jr
, et al
.
Differentiating behavior initiation and maintenance: theoretical framework and proof of concept
.
Health Educ Behav
. 2014 Jun;41:325–336
[PubMed]
40.
James
WP
,
Astrup
A
,
Finer
N
, et al
.
Effect of sibutramine on weight maintenance after weight loss: a randomised trial. STORM Study Group. Sibutramine Trial of Obesity Reduction and Maintenance
.
Lancet
2000
;
356
:
2119
2125
[PubMed]
41.
Van Gaal LF, Rissanen AM, Scheen AJ, Ziegler O, Rössner S; RIO-Europe Study Group. 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 2005;365:1389–1397
42.
Van Gaal
LF
,
Scheen
AJ
,
Rissanen
AM
,
Rössner
S
,
Hanotin
C
,
Ziegler
O
;
RIO-Europe Study Group
.
Long-term effect of CB1 blockade with rimonabant on cardiometabolic risk factors: two year results from the RIO-Europe Study
.
Eur Heart J
2008
;
29
:
1761
1771
43.
Sjöström
L
,
Rissanen
A
,
Andersen
T
, et al.;
European Multicentre Orlistat Study Group
.
Randomised placebo-controlled trial of orlistat for weight loss and prevention of weight regain in obese patients
.
Lancet
1998
;
352
:
167
172
44.
Rössner
S
,
Sjöström
L
,
Noack
R
,
Meinders
AE
,
Noseda
G
;
European Orlistat Obesity Study Group
.
Weight loss, weight maintenance, and improved cardiovascular risk factors after 2 years treatment with orlistat for obesity
.
Obes Res
2000
;
8
:
49
61
45.
Richelsen
B
,
Tonstad
S
,
Rössner
S
, et al
.
Effect of orlistat on weight regain and cardiovascular risk factors following a very-low-energy diet in abdominally obese patients: a 3-year randomized, placebo-controlled study
.
Diabetes Care
2007
;
30
:
27
32
46.
Smith
SR
,
Weissman
NJ
,
Anderson
CM
, et al.;
Behavioral Modification and Lorcaserin for Overweight and Obesity Management (BLOOM) Study Group
.
Multicenter, placebo-controlled trial of lorcaserin for weight management
.
N Engl J Med
2010
;
363
:
245
256
47.
Gadde
KM
,
Allison
DB
,
Ryan
DH
, et al
.
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
2011
;
377
:
1341
1352
48.
Wadden TA, Hollander P, Klein S, et al.; NN8022-1923 Investigators. Weight maintenance and additional weight loss with liraglutide after low-calorie-diet-induced weight loss: the SCALE maintenance randomized study.
Int J Obes
2013
;
17
:
1443
1451
49.
Astrup A, Caterson I, Zelissen P, et al. Topiramate: long-term maintenance of weight loss induced by a low-calorie diet in obese subjects.
Obes Res
2004
;
12
:
1658
1669
50.
van Baak
MA
,
van Mil
E
,
Astrup
AV
, et al
.;
STORM Study Group. Leisure-time activity is an important determinant of long-term weight maintenance after weight loss in the Sibutramine Trial on Obesity Reduction and Maintenance (STORM trial)
.
Am J Clin Nutr
2003
;
78
:
209
214
[PubMed]
51.
Scheen
AJ
,
Van Gaal
LF
.
Combating the dual burden: therapeutic targeting of common pathways in obesity and type 2 diabetes
.
Lancet Diabetes Endocrinol
2014
;
2
:
911
922
[PubMed]
52.
Cefalu
WT
,
Bray
GA
,
Home
PD
, et al
. Advances in the science treatment and prevention of the disease of obesity: reflections from a Diabetes Care Editors’ Expert Forum.
Diabetes Care
2015
;
38
:
1567
1582
[PubMed]
53.
Devenny
JJ
,
Godonis
HE
,
Harvey
SJ
,
Rooney
S
,
Cullen
MJ
,
Pelleymounter
MA
.
Weight loss induced by chronic dapagliflozin treatment is attenuated by compensatory hyperphagia in diet-induced obese (DIO) rats
.
Obesity (Silver Spring)
2012
;
20
:
1645
1652
[PubMed]
54.
Scheen
AJ
.
Pharmacodynamics, efficacy and safety of sodium-glucose co-transporter type 2 (SGLT2) inhibitors for the treatment of type 2 diabetes mellitus
.
Drugs
2015
;
75
:
33
59
[PubMed]
55.
Bolinder
J
,
Ljunggren
Ö
,
Kullberg
J
, et al
.
Effects of dapagliflozin on body weight, total fat mass, and regional adipose tissue distribution in patients with type 2 diabetes mellitus with inadequate glycemic control on metformin
.
J Clin Endocrinol Metab
2012
;
97
:
1020
1031
[PubMed]
56.
Stenlöf
K
,
Cefalu
WT
,
Kim
KA
, et al
.
Efficacy and safety of canagliflozin monotherapy in subjects with type 2 diabetes mellitus inadequately controlled with diet and exercise
.
Diabetes Obes Metab
2013
;
15
:
372
382
[PubMed]
57.
Stenlöf
K
,
Cefalu
WT
,
Kim
KA
, et al
.
Long-term efficacy and safety of canagliflozin monotherapy in patients with type 2 diabetes inadequately controlled with diet and exercise: findings from the 52-week CANTATA-M study
.
Curr Med Res Opin
2014
;
30
:
163
175
[PubMed]
58.
Bays
HE
,
Weinstein
R
,
Law
G
,
Canovatchel
W
.
Canagliflozin: effects in overweight and obese subjects without diabetes mellitus
.
Obesity (Silver Spring)
2014
;
22
:
1042
1049
[PubMed]
59.
Ferrannini
G
,
Hach
T
,
Crowe
S
,
Sanghvi
A
,
Hall
KD
,
Ferrannini
E
.
Energy balance after sodium-glucose cotransporter 2 inhibition
.
Diabetes Care
2015
;
38
:
1730
1735
[PubMed]
60.
Van Gaal
L
,
Scheen
A
.
Weight management in type 2 diabetes: current and emerging approaches to treatment
.
Diabetes Care
2015
;
38
:
1161
1172
[PubMed]
61.
Fulcher
G
,
Matthews
DR
,
Perkovic
V
, et al.;
CANVAS trial collaborative group
.
Efficacy and safety of canagliflozin when used in conjunction with incretin-mimetic therapy in patients with type 2 diabetes
.
Diabetes Obes Metab
2016
;
18
:
82
91
[PubMed]
62.
Cummings
BP
.
Leptin therapy in type 2 diabetes
.
Diabetes Obes Metab
2013
;
15
:
607
612
[PubMed]
63.
Lovshin
JA
,
Drucker
DJ
.
Incretin-based therapies for type 2 diabetes mellitus
.
Nat Rev Endocrinol
2009
;
5
:
262
269
[PubMed]
64.
Grasso
P
.
Novel approaches to the treatment of obesity and type 2 diabetes mellitus: bioactive leptin-related synthetic peptide analogs
.
Recent Pat Endocr Metab Immune Drug Discov
2011
;
5
:
163
175
[PubMed]
65.
Moon
HS
,
Chamberland
JP
,
Mantzoros
CS
.
Amylin and leptin activate overlapping signalling pathways in an additive manner in mouse GT1-7 hypothalamic, C₂C₁₂ muscle and AML12 liver cell lines
.
Diabetologia
2012
;
55
:
215
225
[PubMed]
66.
Clemmensen
C
,
Chabenne
J
,
Finan
B
, et al
.
GLP-1/glucagon coagonism restores leptin responsiveness in obese mice chronically maintained on an obesogenic diet
.
Diabetes
2014
;
63
:
1422
1427
[PubMed]
67.
Finan
B
,
Ma
T
,
Ottaway
N
, et al
.
Unimolecular dual incretins maximize metabolic benefits in rodents, monkeys, and humans
.
Sci Transl Med
2013
;
5
:
209ra151
[PubMed]
68.
Kim
SJ
,
Nian
C
,
Karunakaran
S
,
Clee
SM
,
Isales
CM
,
McIntosh
CH
.
GIP-overexpressing mice demonstrate reduced diet-induced obesity and steatosis, and improved glucose homeostasis
.
PLoS One
2012
;
7
:
e40156
[PubMed]
69.
Field
BC
,
Wren
AM
,
Peters
V
, et al
.
PYY3-36 and oxyntomodulin can be additive in their effect on food intake in overweight and obese humans
.
Diabetes
2010
;
59
:
1635
1639
[PubMed]
70.
Trevaskis
JL
,
Sun
C
,
Athanacio
J
, et al
.
Synergistic metabolic benefits of an exenatide analogue and cholecystokinin in diet-induced obese and leptin-deficient rodents
.
Diabetes Obes Metab
2015
;
17
:
61
73
[PubMed]
71.
Finan
B
,
Yang
B
,
Ottaway
N
, et al
.
A rationally designed monomeric peptide triagonist corrects obesity and diabetes in rodents
.
Nat Med
2015
;
21
:
27
36
[PubMed]