ArticlePDF Available

Liraglutide (Saxenda®) as a Treatment for Obesity

Authors:

Abstract and Figures

Obesity is a significant concern in the United States, affecting approximately 35% of the popula- tion. Comorbidities, such as diabetes, hypertension, and hyperlipidemia, significantly increase one's risk of heart attack, stroke, and even death. Liraglutide, a medication originally used to treat diabetes, has been approved for the treatment of obesity. Clinical trials have shown significant improvements in body weight and body mass index (BMI) at a dose of up to 3.0 mg daily. The most common adverse effects are gastrointestinal in nature, however, these often subside with time. Safety concerns with regards to thyroid tumors and pancreatitis should be carefully considered prior to use of this agent. Liraglutide should be considered an additional tool in the treatment of obesity, especially in patients with concomitant diabetes.
No caption available
… 
Content may be subject to copyright.
Food and Nutrition Sciences, 2016, 7, 227-235
Published Online April 2016 in SciRes. http://www.scirp.org/journal/fns
http://dx.doi.org/10.4236/fns.2016.74024
How to cite this paper: St. Onge, E., Miller, S.A. and Motycka, C. (2016) Liraglutide (Saxenda®) as a Treatment for Obesity.
Food and Nutrition Sciences, 7, 227-235. http://dx.doi.org/10.4236/fns.2016.74024
Liraglutide (Saxenda®) as a Treatment for
Obesity
Erin St. Onge1, Shannon A. Miller1, Carol Motycka2
1University of Florida College of Pharmacy, Orlando, FL, USA
2University of Florida College of Pharmacy, Jacksonville, FL, USA
Received 7 March 2016; accepted 17 April 2016; published 20 April 2016
Copyright © 2016 by authors and Scientific Research Publishing Inc.
This work is licensed under the Creative Commons Attribution International License (CC BY).
http://creativecommons.org/licenses/by/4.0/
Abstract
Obesity is a significant concern in the United States, affecting approximately 35% of the popula-
tion. Comorbidities, such as diabetes, hypertension, and hyperlipidemia, significantly increase
one’s risk of heart attack, stroke, and even death. Liraglutide, a medication originally used to treat
diabetes, has been approved for the treatment of obesity. Clinical trials have shown significant
improvements in body weight and body mass index (BMI) at a dose of up to 3.0 mg daily. The most
common adverse effects are gastrointestinal in nature, however, these often subside with time.
Safety concerns with regards to thyroid tumors and pancreatitis should be carefully considered
prior to use of this agent. Liraglutide should be considered an additional tool in the treatment of
obesity, especially in patients with concomitant diabetes.
Keywords
Diabetes, Liraglutide, Obesity, Treatment
1. Introduction
The American Heart Association has adopted body mass index (BMI) as the accepted measurement for adiposity.
BMI is calculated by taking a patient’s weight in kilograms and dividing by their height in meters squared
(kg/m2). Individuals with a BMI of 30 or greater are considered obese [1]. As of 2014, 34.9% of the United
States (US) adult population was defined as obese. Although obesity rates do not appear to have significantly
increased overall in the past decade, the rate of obesity in women over the age of 60 has increased significantly
[2]. This increased rate of obesity in older women corresponds to the two-fold increase in diabetes in persons
aged 65 or older since 1980 [3]. The association between obesity and diabetes has been well studied, with
obese men having a seven times greater chance of developing diabetes and obese women having a twelve fold
E. St. Onge et al.
228
increased risk [4]. However, other comorbidities such as hypertension, hyperlipidemia, asthma, arthritis and
general poor health have also been associated with obesity [5] making it one of leading causes of preventable
death [6].
Limited options exist for the treatment of obesity. This is due in part to weight loss medications being re-
moved from the market. For example, sibutramine was removed due to the increased risk of cardiovascular
events discovered in the SCOUT (Effect of Sibutramine on Cardiovascular Outcomes in Overweight and Obese
Subjects) trial [7]. Saxenda (liraglutide) was approved in December 2014 for the treatment of obesity. Prior to its
approval, liraglutide was used exclusively for diabetes at a lower dose (1.2 to 1.8 mg daily) and under a different
brand name, Victoza®. To gain approval for weight loss, medications must produce a weight loss of 5% from
baseline; this requirement was met in early clinical trials with liraglutide [8].
With the large number of individuals in the U.S. who are diabetic as well as obese, having a medication which
could potentially treat both conditions is crucial. This article will explore the use of liraglutide as a weight loss
medication including its mechanism of action, safety profile, and clinical evidence of efficacy.
2. Pharmacology
Research connecting the gastrointestinal system and insulin secretion dates back to the 1960s stemming from an
observation that oral glucose administration produced a greater insulin response compared to an intravenous
glucose infusion [9] [10]. This was later termed the incretin effect. The incretin effect is primarily mediated by
two insulinotropic gut hormones, glucagon-like peptide (GLP-1) and gastric inhibitory polypeptide (GIP), and
accounts for approximately 50% - 70% of the total insulin secreted following oral glucose administration [11].
GLP-1 is a peptide released from L-cells in the intestine in response to nutrient ingestion and subsequently en-
hances glucose-stimulated insulin secretion. Circulating GLP-1 and GIP are found within minutes post meal in-
gestion, suggesting both neuronal and endocrine signals are responsible for the release of the hormone [12]. The
incretin effect is greatly impaired in patients with type 2 diabetes (T2DM) and GLP-1 secretion is noticeably de-
ficient.
Liraglutide is a GLP-1 analogue, produced by recombinant DNA technology, which shares 97% amino acid
sequence homology to endogenous human GLP-1 [13] [14]. Native GLP-1 is rapidly degraded by endogenous
DPP-4 enzyme promoting a short half-life of 1.5 - 2 minutes. Liraglutide is stable against DPP-4 degradation
and has a prolonged plasma half-life of 13 hours [15] [16]. Similar to endogenous GLP-1, liraglutide binds to
and activates the GLP-1 receptor. GLP-1 receptors are found in the pancreas, stomach, intestine, heart, kidney,
peripheral and central nervous system [16] [17]. Once activated, several responses occur including glu-
cose-dependent stimulation of pancreatic insulin secretion and inhibition of inappropriately high glucagon secre-
tion.
In addition to its glucoregulatory mechanisms, activation of GLP-1 receptors regulates appetite and caloric
intake, slow gastric emptying and promote weight loss [13]. This has promoted further research with GLP-1 re-
ceptor agonists in the treatment of obesity. In clinical trials, subjects taking liraglutide with a BMI between 30 -
40 kg/m2 with or without diabetes, observed short term reduction in body weight by decreasing calorie intake
and improving eating behaviors without an increase in 24-h energy expenditure [18] [19]. Mean estimated ener-
gy intake during a meal was significantly reduced in liraglutide 1.8 and 3 mg subjects when compared to place-
bo (p < 0.003). This reduction translated into improved appetite rating scores consisting of reduced appetite, sa-
tiety, and fullness in both liraglutide arms compared with placebo. Gastric emptying rates were similar between
the three groups during a 5-h meal test. The 1-h gastric emptying rates, however, were 23% lower in the liraglu-
tide 3 mg arm and 13% lower in the 1.8 mg arm when compared to placebo (p = 0.007, p = 0.14, respectively).
The clinical significance of this finding is unknown [19].
Liraglutide’s effect on cardiac repolarization was tested in a QTc trial. A randomized, placebo-controlled,
cross-over study, found no clinically relevant prolongation in the QTc interval after daily doses up to 1.8 mg
were given [20]. The maximum plasma concentrations (Cmax) in overweight and obese subjects treated with li-
raglutide 3 mg was similar to the Cmax observed in healthy volunteers.
3. Pharmacokinetics
The pharmacokinetic properties of liraglutide (outlined in Table 1) do not differ to a clinically relevant extent
when comparing various subcutaneous injection sites (abdomen, upper arm, and thigh) [13]. Liraglutide is highly
E. St. Onge et al.
229
Table 1. Pharmacokinetic properties of liraglutide [13]-[16].
Absorption Max plasma concentration in 9 - 14h
Distribution
Bioavalability 55%
Volume of distribution 20 - 25 L for a 100 kg person
Plasma protein binding (>98%)
Metabolism Similar to that of large proteins
In vitro studies suggest metabolism by DPP-4 and neutral endopeptidase
Excretion
No specific organ is major route of elimination (drug is metabolized)
o Some metabolites excreted in feces (5%) and urine (6%)
Mean apparent clearance 0.9 - 1.4 L/h
Half-life ≈13 h
protein bound (98%), and has reduced susceptibility from DPP4 degradation [21]. Liraglutide demonstrates a
relatively slow rate of absorption, with maximum concentrations observed at 9 - 14 hours [22]. Absolute bio-
availability is approximately 55% upon subcutaneous administration with a mean apparent volume of distribu-
tion of 20 - 25 L (100 kg person) [13].
Liraglutide is metabolized in a manner similar to large proteins; no specific route has been identified as a ma-
jor route of elimination [13]. Intact liraglutide is not found in feces or urine and only low levels of liraglutide
related metabolites are detected during the first 6 - 8 days [17] [23]. The elimination half-life is approximately
13 hours, allowing for once daily administration. Although renal elimination does not appear significant, liraglu-
tide area under the curve (AUC) was lower in patients with mild to severe renal impairment. There have been
reports of acute renal failure with GLP-1 receptor agonists including liraglutide, however some of these reports
were in patients with underlying renal disease and the majority occurred in volume depleted patients [13]. Data
in hepatic impairment is limited. Caution should be utilized in patients with renal or hepatic impairment.
Race, ethnicity and gender have no effect on the pharmacokinetics of liraglutide and no dose adjustment is
necessary [13]. This drug demonstrated little to no inhibition of cytochrome P450 enzymes thus indicating low
potential for pharmacokinetic cytochrome P450 mediated and plasma protein binding drug-drug interactions
[24]. No clinically significant drug interaction has been identified with oral contraceptives, digoxin, lisinopril,
atorvastatin, acetaminophen, griseofulvin, and insulin detemir with coadministration of steady state liraglutide
1.8 mg/day [13].
4. Clinical Trials
Several clinical trials have investigated the efficacy of liraglutide on weight loss in non-diabetic as well as di-
abetic patients (Table 2).
The efficacy of liraglutide as a weight loss agent was evaluated in a 56-week randomized, controlled clinical
trial [25]. Patients without diabetes were enrolled in the trial if BMI was 30 (or 27 with other comorbidities).
In a 2:1 ratio, patients were randomized to receive liraglutide (n = 2487) or placebo (n = 1244). The dose of li-
raglutide was initiated at 0.6 mg subcutaneously daily and titrated up 0.6 mg weekly to the target dose of 3.0 mg
daily. Both groups received counseling on lifestyle modifications. The primary endpoints in this trial were
weight change from baseline, the proportion of patients who lost at least 5% of their weight from baseline, and
the proportion of patients who lost more than 10% of their baseline body weight. After 56 weeks, patients in the
liraglutide group lost 8.4 kg ± 7.3 kg while patients in the placebo group lost 2.8 kg ± 6.5 kg from baseline (p <
0.001 vs placebo). The percentage of patients who lost at least 5% of their body weight from baseline was 63.2% in
the liraglutide group vs. 27.1% in the placebo group (p < 0.001 vs placebo). Likewise, 33.1% of patients in the
liraglutide group vs 10.6% of patients in the placebo group lost at least 10% of their body weight from baseline
(p < 0.001). Patients in the liraglutide group most commonly reported gastrointestinal related adverse events;
with nausea and vomiting reported primarily within the first 4 - 8 weeks of treatment. Based on the results of
this trial, the authors concluded liraglutide 3.0 mg once daily, in combination with diet and exercise, produced
clinically meaningful weight loss in obese patients without diabetes.
A small trial involving 44 obese binge-eaters aimed to evaluate the efficacy of liraglutide 1.8 mg daily for 12
weeks [26]. Subjects were randomized to receive liraglutide 1.8 mg plus diet and exercise or diet and exercise
alone. Subjects were excluded if they were taking medications which affect weight or appetite and if they had
diabetes, impaired glucose tolerance, or cardiovascular disease. Among the primary endpoints were changes in
E. St. Onge et al.
230
E. St. Onge et al.
231
Binge Eating Scale (BES) scores, BMI, and waist circumference from baseline. Liraglutide treatment resulted in
significant decreases in BES score, BMI, and waist circumference from baseline; while subjects in the control
group only saw a significant decrease in BES from baseline. The investigators concluded that 12 weeks of lirag-
lutide treatment in non-diabetic binge-eating patients resulted in significant improvements in BES and body
weight.
An observational study reported the results from 84 overweight or obese women with polycystic ovary syn-
drome (PCOS) who were treated with up to 1.8 mg of liraglutide daily for at least 4 weeks [27]. Patients were
included in the study if they had failed to lose any weight despite therapy with metformin and lifestyle interven-
tions for 6 months. The primary endpoints were change in body weight and BMI from baseline. Mean body
weight at baseline was 98.9 kg and the mean duration of treatment with liraglutide was 27.8 weeks. Results of
this study showed a mean weight loss of 9kg (95% CI 7.8 - 10.1; p < 0.0001) and a mean change in BMI of 3.2
kg/m2 (95% CI 2.8 - 3.6; p < 0.0001) from baseline. When evaluating these parameters in patients who were
treated for 20 weeks, the mean weight loss and change in BMI compared to baseline were even greater. Based
on the results of this study, the authors concluded liraglutide may be an effective adjunct to metformin, diet, and
exercise in overweight and obese women with PCOS.
The efficacy of liraglutide was evaluated in another trial of 84 overweight or obese women who had a diagno-
sis of PCOS [28]. In this 12 week trial, 32 obese women with newly diagnosed PCOS were randomized to re-
ceive metformin 1000 mg twice daily or liraglutide 1.2 mg daily. Changes in BMI, body weight, waist circum-
ference, and body fat mass were the primary endpoints. After 12 weeks, significant changes in BMI, body
weight, waist circumference, and body fat mass were experienced by patients in both groups compared to base-
line. Adverse effects reported by both groups were gastrointestinal in nature with nausea and diarrhea most
commonly reported. Based on the results of this study, the investigators concluded short term treatment with li-
raglutide was associated with significant weight loss in obese women with PCOS.
The SCALE trial investigated the safety and efficacy of liraglutide for weight loss in 846 overweight or obese
patients with T2DM over a period of 56 weeks [29]. Patients were included in this trial if they had a BMI 27
with a stable body weight over the past 3 months, were treated with 0 to 3 diabetic agents, and had a hemoglobin
A1c between 7% - 10%. In a 2:1:1 ratio, patients were randomly assigned to receive liraglutide 3 mg, liraglutide
1.8mg, or placebo. The primary endpoints of this trial were relative change in body weight, proportion of pa-
tients losing 5% of their baseline body weight, and the proportion of patients losing >10% of their body weight
from baseline. At week 56, the mean weight loss was 6.4 kg, 5 kg, and 2.2 kg for the liraglutide 3 mg, liraglutide
1.8 mg, and placebo groups, respectively. These results were statistically significant for both liraglutide groups
compared to placebo. The proportion of patients losing 5% of their body weight from baseline was 54.3% in
the liraglutide 3 mg group, 40.4% in the liraglutide 1.8 mg group, and 21.4% in the placebo group. Again, these
results were statistically significant for both liraglutide groups when compared to placebo. Finally, the propor-
tion of patients who lost >10% of their body weight from baseline was 25.2%, 15.9%, and 6.7% in the liraglu-
tide 3 mg, liraglutide 1.8 mg, and placebo groups, respectively. Based on these results, the study investigators
concluded liraglutide 3 mg daily leads to significant weight loss over 56 weeks of treatment compared to place-
bo.
A smaller study involving 328 patients, aimed to assess the effect of liraglutide on body weight and waist cir-
cumference in overweight and obese Chinese patients with T2DM [30]. In this open-label study, patients re-
ceived up to 1.8 mg daily of liraglutide over 24 weeks. The primary endpoints were defined as changes in body
weight, BMI, and waist circumference to height ratio (WHR) from baseline. After 24 weeks of treatment, sig-
nificant reductions in all primary outcomes were observed. The authors compared their results to clinical trials
conducted in Western countries and concluded that liraglutide is more effective in Chinese patients than Western
patient populations. It is important to note here, the trials being compared were not designed to evaluate weight
loss as a primary outcome and thus this conclusion warrants further investigation.
The efficacy of liraglutide in reducing A1c and weight was investigated in a prospective, observational study
of Arab patients with T2DM [31]. This study was conducted at 3 centers in Dubai, and included all adult pa-
tients with T2DM between 18 - 70 years of age who received a prescription for liraglutide. The dose of liraglu-
tide was initiated at 0.6 mg and titrated to 1.2 mg or 1.8 mg daily as tolerated. The primary endpoints were de-
fined as change in weight and A1c from baseline to 6 months. The mean change in body weight from baseline to
6 months was 2.5% (p < 0.001). In addition, the mean A1c decreased from 8.3% at baseline to 7.6% after 6
months (p < 0.001). Based on these results, the investigators concluded liraglutide as add on therapy for diabetes,
E. St. Onge et al.
232
produced significant reductions in weight and A1c in this Arab population.
5. Adverse Effects
The most common adverse effects reported by patients using liraglutide include nausea, diarrhea, constipation,
vomiting, headache, decreased appetite, dyspepsia, fatigue, dizziness, and abdominal pain [13]. Hypoglycemia is
a concern when using liraglutide with other antidiabetic agents especially insulin or sulfonylureas. The pre-
scribing information for liraglutide recommends against the use of liraglutide in combination with insulin [13].
However, combination therapy with long acting insulin has been evaluated in two studies [32] [33]. The results
of these trials showed low rates of hypoglycemia with combination therapy. Little information is available re-
garding the use of liraglutide with other types of insulin, therefore, providers should encourage regular glucose
monitoring with initiation of therapy or dose changes and make adjustments as necessary. The prescribing in-
formation for liraglutide states there is an increased risk of serious hypoglycemia in combination with insulin
secretagogues (ex: sulfonylureas, meglitinides) [13]. Thus, a decreased dose of the insulin secretagogue should
be considered when initiating therapy with liraglutide.
Thyroid C-cell tumors and acute pancreatitis are among the most serious warnings and precautions with li-
raglutide use. The package insert for liraglutide contains a black box warning for the risk of thyroid C-cell tu-
mors [13]. These tumors have only been seen in mice and rats; the ability of liraglutide to cause such tumors in
humans is unknown. The use of liraglutide is contraindicated in patients with a personal or family history of
medullary thyroid carcinoma (MTC) or in patients with multiple endocrine neoplasia syndrome type 2 (MEN 2).
Patients should be educated about symptoms of potential thyroid tumors including a mass in the neck, dysphagia,
dyspnea, and persistent hoarseness [34]. Postmarketing reports of acute pancreatitis have surfaced with liraglu-
tide use. Providers should monitor patients for signs and symptoms of pancreatitis after initiation of liraglutide
and with any dose increases. In addition, patients should be educated to seek medical help immediately if they
experience persistent, severe abdominal pain with liraglutide use [13] [34]. The use of liraglutide is currently
being monitored under a FDA REMS (Risk Evaluation and Mitigation Strategy) program. A fact sheet for this
program can be found at www.SAXENDA.com/REMS.
6. Dosage and Administration
Saxenda® (liraglutide) is available as multi-dose, prefilled syringes. Each pen contains 18 mg of drug (6 mg/mL,
3 mL) and can deliver doses of 0.6 mg, 1.2 mg, 2.4 mg, or 3 mg. Saxenda comes in packs of 3 or 5 pens de-
pending on the patient’s need. The pens should be stored in the refrigerator prior to use but can be kept at room
temperature for up to 30 days once the pen has been used [13].
In an effort to minimize gastrointestinal adverse effects, liraglutide should be initiated at 0.6 mg daily for one
week. The dose should be titrated weekly by 0.6 mg until the target dose of 3 mg daily has been reached. If a pa-
tient is not able to tolerate the dose increase, delay titration for one additional week. Liraglutide should be in-
jected subcutaneously once daily in the abdomen, thigh, or upper arm. Patients should be educated to rotate be-
tween all three sites if needed as absorption has been shown to be equivalent in clinical studies [14]. Liraglutide
can be administered without regard to meals, however, patients should be encouraged to inject themselves at the
same time each day [13].
7. Therapeutic Considerations
In addition to the adverse effects and precautions previously mentioned, there are other factors practitioners
should take into consideration when considering the use of Saxenda®. Cost is a serious concern as very few in-
surance companies cover weight loss medications [35]. This leaves patients to decide whether or not they will
pay for the medication out of pocket [36]. Studies have shown only 2/3 of all medications that are prescribed are
actually filled [37]. Therefore, it is pertinent that clinicians discuss a patient’s financial situation prior to provid-
ing a prescription for any weight loss medication. Saxenda® costs approximately $1200 per month for cash pay-
ing patients. This is substantially more expensive than many of the other agents available for weight loss. To
avoid this, some clinicians have considered placing patients on Victoza® for weight loss. However, it is impor-
tant to keep in mind that the highest dose recommended for Victoza® is 1.8 mg of liraglutide which has not been
shown to have the same efficacy for weight loss as Saxenda® at 3.0 mg of liraglutide.
E. St. Onge et al.
233
Another factor to take into consideration is dosage form. Saxenda® is a subcutaneous injection which may be
undesirable to some patients. For diabetic patients who may already be injecting insulin, this may not be a con-
cern. However, patients may be unwilling to inject themselves or may have a fear of injections and/or needles.
Because of this, it is important that clinicians discuss these fears with patients before prescribing a medication
like Saxenda®.
Lastly, with a substantial number of patients experiencing nausea and vomiting with this medication, careful
consideration should be given to individuals who may be more sensitive to these adverse effects. In addition,
providers should use caution in those whom vomiting could exacerbate an underlying condition such as recent
abdominal surgery or respiratory problems. .
8. Conclusion
Rate of obesity worldwide continues to climb and presents as a major public health concern. Obesity is fre-
quently coupled with other debilitating diseases such as heart disease and diabetes. Reduced caloric intake and
increased physical activity are the cornerstones of obesity treatment. However, in some patients pharmacological
treatment may be warranted. High dose liraglutide has been shown to reduce food intake and promote weight
loss in patients with or without diabetes, for up to a year. Liraglutide may also have a role in obese patients with
PCOS. Overall, liraglutide is well tolerated, with gastrointestinal complaints reported most commonly. Moni-
toring for safety, efficacy and tolerability are important. Liraglutide is an attractive option for chronic weight
management in overweight or obese patients. Additional research is warranted for its role in combination weight
loss treatments.
References
[1] Jensen, M.D., Ryan, D.H., Apovian, C.M., et al. (2014) 2013 AHA/ACC/TOS Guideline for the Management of
Overweight and Obesity in Adults: A Report of the American College of Cardiology/American Heart Association Task
Force on Practice Guidelines and The Obesity Society. JACC, 63, 2985-3023.
http://dx.doi.org/10.1016/j.jacc.2013.11.004
[2] Ogden, C.L., Carroll, M.D., Kit, B.K. and Flegal, K.M. (2014) Prevalence of Childhood and Adult Obesity in the
United States, 2011-2012. JAMA, 311, 806-814. http://dx.doi.org/10.1001/jama.2014.732
[3] Centers for Disease Control and Prevention. Rates of Diagnosed Diabetes per 100 Civilian, Non-Institutionalized Pop-
ulation, by Age, United States, 1980-2014. http://www.cdc.gov/diabetes/statistics/prev/national/figbyage.htm
[4] Guh, D.P., Zhang, W., Bansback, N., et al. (2009) The Incidence of Co-Morbidities Related to Obesity and Overweight.
A Systematic Review and Meta-Analysis. BMC Public Health, 9, 1-20. http://dx.doi.org/10.1186/1471-2458-9-88
[5] Mokdad, A.H., Ford, E.S., Bownman, B.A., et al. (2003) Prevalence of Obesity, Diabetes, and Obesity-Related Health
Risk Factors, 2001. JAMA, 289, 76-79. http://dx.doi.org/10.1001/jama.289.1.76
[6] Centers for Disease Control and Prevention. Potentially Preventable Deaths from the Five Leading Causes of Death-
United States, 2008-2010. http://www.cdc.gov/mmwr/pdf/wk/mm6317.pdf
[7] James, W.P., Caterson, I.D., Coutinho, W., et al. (2010) Effect of Sibutramine on Cardiovascular Outcomes in Over-
weight and Obese Subjects. NEJM, 363, 905-917. http://dx.doi.org/10.1056/NEJMoa1003114
[8] US Food and Drug Administration. Guidance for Industry Developing Products for Weight Management.
http://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ucm071612.pdf
[9] Erick, H., Stimmler, L., Hlad, C.J., et al. (1964) Plasma Insulin Response to Oral and Intravenous Glucose Administra-
tion. The Journal of Clinical Endocrinology & Metabolism, 24, 1076-1082. http://dx.doi.org/10.1210/jcem-24-10-1076
[10] Kazafeos, K. (2011) Incretin Effect: GLP-1, GIP, DPP4. Diabetes Research and Clinical Practice, 93, S32-s36.
http://dx.doi.org/10.1016/S0168-8227(11)70011-0
[11] McIntyre, N., Holdsworth, C.D. and Turner, D.S. (1964) New Interpretations of Oral Glucose Tolerance. The Lancet, 2,
20-21. http://dx.doi.org/10.1016/S0140-6736(64)90011-X
[12] Chaikomin, R., Doran, S. and Jones, K.L. (2005) Initially More Rapid Small Intestinal Glucose Delivery Increases
Plasma Insulin, GIP, and GLP-1 but Does Not Improve Overall Glycemia in Healthy Subjects. American Journal of
PhysiologyEndocrinology and Metabolism, 289, E504-E507. http://dx.doi.org/10.1152/ajpendo.00099.2005
[13] (2014) Liraglutide [Package Insert]. Novo Nordisk Inc., Plainsboro, NJ.
[14] Jacobsen, L.V., Flint, A., Olsen, A.K., et al. (2015) Liraglutide in Type 2 Diabetes Mellitus: Clinical Pharmacokinetic
and Pharmacodynamics. Clinical Pharmacokinetics, 1-16. http://dx.doi.org/10.1007/s40262-015-0343-6
E. St. Onge et al.
234
[15] Scott, L.J. (2015) Liraglutide: A Review of Its Use in the Management of Obesity. Drugs, 75, 899-910.
http://dx.doi.org/10.1007/s40265-015-0408-8
[16] Perry, C.M. (2011) Liraglutide: A Review of Its Use in the Management of Type 2 Diabetes Mellitus. Drugs, 71, 2547-
2573. http://dx.doi.org/10.2165/11208110-000000000-00000
[17] Scott, L.J. (2014) Liraglutide: A Review of Its Use in Adult Patients with Type 2 Diabetes Mellitus. Drugs, 74, 2161-
2174. http://dx.doi.org/10.1007/s40265-014-0321-6
[18] Horowitz, M., Flint, A., Jones, K.L., 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 Research and Clini-
cal Practice, 97, 258-266. http://dx.doi.org/10.1016/j.diabres.2012.02.016
[19] Van Can, J., Sloth, B., Jensen, C.B., et al. (2014) Effects of the Once Daily GLP-1 Analogue Liraglutide on Gastric
Emptying, Glycemic Parameters, Appetite and Energy Metabolism in Obese, Non-Diabetic Adults. International
Journal of Obesity, 38, 784-793. http://dx.doi.org/10.1038/ijo.2013.162
[20] Chatterjee, D.J., Khutoryansky, N., Zdravkovic, M., et al. (2009) Absence of QTc Prolongation in a Thorough QT Stud
with Subcutaneous Liraglutide, a Once-Daily Human GLP-1 Analog for Treatment of Type 2 Diabetes. The Journal of
Clinical Pharmacology, 49, 1353-1362. http://dx.doi.org/10.1177/0091270009339189
[21] Nuffer, W.A. and Trujillo, J.M. (2015) Liraglutide: A New Option for the Treatment of Obesity. Pharmacotherapy, 35,
926-934. http://dx.doi.org/10.1002/phar.1639
[22] Agerso, H., Jensen, L.B., Elbrond, B., et al. (2002) The Pharmacokinetics, Pharmacodynamics, Safety and Tolerability
of NN211, a New Long-Acting GLP-1 Derivative, in Healthy Men. Diabetologia, 45, 195-202.
http://dx.doi.org/10.1007/s00125-001-0719-z
[23] Neumiller, J.J. and Campbell, R.K. (2009) Liraglutide: A Once Daily Incretin Mimetic for the Treatment of Type 2
Diabetes Mellitus. Annals of Pharmacotherapy, 43, 1433-1444. http://dx.doi.org/10.1345/aph.1M134
[24] Gonzalez, C., Beruto, V., Keller, G., et al. (2006) Investigational Treatments for Type 2 Diabetes Mellitus: Exenatide
and Liraglutide. Expert Opinion on Investigational Drugs, 15, 887-895. http://dx.doi.org/10.1517/13543784.15.8.887
[25] Pi-Sunyer, X., Astrup, A., Fujioka, K., et al. (2015) A Randomized, Controlled Trial of 3.0mg of Liraglutide in Weight
Management. NEJM, 373, 11-22. http://dx.doi.org/10.1056/NEJMoa1411892
[26] Robert, S.A., Rohana, A.G., Shah, S.A., et al. (2015) Improvement in Binge Eating in Non-Diabetic Obese Individuals
after 3 Months of Treatment with Liraglutide—A Pilot Study. Obesity Research & Clinical Practice, 9, 301-304.
http://dx.doi.org/10.1016/j.orcp.2015.03.005
[27] Rasmussen, C.B. and Lindenberg, S. (2014) The Effect of Liraglutide on Weight Loss in Women with Polycystic
Ovary Syndrome: An Observational Study. Frontiers in Endocrinology, 5, 1-6.
http://dx.doi.org/10.3389/fendo.2014.00140
[28] Jensterle, M., Kravos, N.A., Pfeifer, M., Kocjan, T. and Janez, A. (2015) A 12-Week Treatment with the Long-Acting
Glucagon-Like Peptide 1 Receptor Agonist Liraglutide Leads to Significant Weight Loss in a Subset of Obese Women
with Newly Diagnosed Polycystic Ovary Syndrome. Hormones, 14, 81-90.
[29] Davies, M.J., Bergenstal, R., Bode, B., et al. (2015) Efficacy of Liraglutide for Weight Loss among Patients with Type
2 Diabetes: The SCALE Diabetes Randomized Clinical Trial. JAMA, 314, 687-699.
http://dx.doi.org/10.1001/jama.2015.9676
[30] Feng, P., Yu, D., Chang, B., et al. (2015) Liraglutide Reduces the Body Weight and Waist Circumference in Chinese
Overweight and Obese Type 2 Diabetic Patients. Acta Pharmacologica Sinica, 36, 200-208.
http://dx.doi.org/10.1038/aps.2014.136
[31] Bashier, A.M., Hussain, A., Abdelgabir, E., et al. (2015) Liraglutide Effect in Reducing HbA1c and Weight in Arab
Population with Type2 Diabetes, a Prospective Observational Trial. Journal of Diabetes & Metabolic Disorders, 14,
48-55. http://dx.doi.org/10.1186/s40200-015-0178-6
[32] Mathieu, C., Rodbard, H.W., Cariou, B., et al. (2014) A Comparison of Adding Liraglutide versus a Single Daily Dose
of Insulin Aspart to Insulin Degludec in Subjects with Type 2 Diabetes. Diabetes, Obesity and Metabolism, 16, 636-
644. http://dx.doi.org/10.1111/dom.12262
[33] DeVries, J.H., Bain, S.C., Rodbard, H.W., et al. (2012) Sequential Intensification of Metformin Treatment in Type 2
Diabetes with Liraglutide Followed by Randomized Addition of Basal Insulin Prompted by A1c Targets. Diabetes
Care, 35, 1446-1454. http://dx.doi.org/10.2337/dc11-1928
[34] US Food and Drug Administration. FDA Requires REMS Safety Information.
http://www.saxendarems.com/content/dam/novonordisk/saxendarems/Documents/LIOU14CDNY6583_A%20_REMS
%20Factsheet_r11.pdf
[35] Lee, S.L., Sheer, J.L., Lopez, J.D., et al. (2010) Coverage of Obesity Treatment. Public Health Reports, 125, 596-604.
E. St. Onge et al.
235
[36] NPR. Many Insurers Don’t Cover Drugs for Weight Loss.
http://www.npr.org/sections/health-shots/2015/01/06/375198217/many-insurers-dont-cover-drugs-for-weight-loss
[37] Tamblyn, R., Eguale, T., Huang, A., Winslade, N. and Doran, P. (2014) The Incidence and Determinants of Primary
Nonadherence with Prescribed Medication in Primary Care: A Cohort Study. Annals of Internal Medicine, 160, 441-
450. http://dx.doi.org/10.7326/M13-1705
... The FDA approved a new dose of liraglutide 3 mg in 2014 for the treatment of obesity in patients without diabetes under the brand name Saxenda ® [125]. This approval was made possible by the impressive results of weight loss brought on by 3 mg liraglutide in clinical trials in T2DM patients (an average of 5% weight loss) [125][126][127]. Daily subcutaneous injections of liraglutide are given [127]. ...
... This approval was made possible by the impressive results of weight loss brought on by 3 mg liraglutide in clinical trials in T2DM patients (an average of 5% weight loss) [125][126][127]. Daily subcutaneous injections of liraglutide are given [127]. The most frequent adverse effects are gastrointestinal, which often go away over time [127]. ...
... Daily subcutaneous injections of liraglutide are given [127]. The most frequent adverse effects are gastrointestinal, which often go away over time [127]. Liraglutide has been the subject of numerous clinical trials; the first of these was the Liraglutide Effect and Action in Diabetes (LEAD) program for diabetics, which enrolled more than 4000 T2DM patients from 40 different nations [128,129]. ...
Article
Full-text available
Obesity is a complex metabolic condition that can have a negative impact on one’s health and even result in mortality. The management of obesity has been addressed in a number of ways, including lifestyle changes, medication using appetite suppressants and thermogenics, and bariatric surgery for individuals who are severely obese. Liraglutide and semaglutide are two of the five Food and Drug Administration (FDA)-approved anti-obesity drugs that are FDA-approved agents for the treatment of type 2 diabetes mellitus (T2DM) patients. In order to highlight the positive effects of these drugs as anti-obesity treatments, we analyzed the weight loss effects of T2DM agents that have demonstrated weight loss effects in this study by evaluating clinical studies that were published for each agent. Many clinical studies have revealed that some antihyperglycemic medications can help people lose weight, while others either cause weight gain or neutral results. Acarbose has mild weight loss effects and metformin and sodium-dependent glucose cotransporter proteins-2 (SGLT-2) inhibitors have modest weight loss effects; however, some glucagon-like peptide-1 (GLP-1) receptor agonists had the greatest impact on weight loss. Dipeptidyl peptidase 4 (DPP-4) inhibitors showed a neutral or mild weight loss effect. To sum up, some of the GLP-1 agonist drugs show promise as weight-loss treatments.
... Obesity is a global healthcare concern with an escalating trend associated with significant comorbidities like diabetes, hypertension, hyperlipidemia, heart diseases, stroke, and cancer (Mancini and de Melo 2017;Onge, Miller, and Motycka 2016). The prevalence of obese, especially morbid obese, individuals has increased continuously over the past few decades (Mancini and de Melo 2017;Onge, Miller, and Motycka 2016). ...
... Obesity is a global healthcare concern with an escalating trend associated with significant comorbidities like diabetes, hypertension, hyperlipidemia, heart diseases, stroke, and cancer (Mancini and de Melo 2017;Onge, Miller, and Motycka 2016). The prevalence of obese, especially morbid obese, individuals has increased continuously over the past few decades (Mancini and de Melo 2017;Onge, Miller, and Motycka 2016). The escalating trend was specifically monitored since 1980; 1980-2014 was reportedly the major transition period of lifestyle modification with reportedly double the prevalence of obesity (Mancini and de Melo 2017;Onge, Miller, and Motycka 2016). ...
... The prevalence of obese, especially morbid obese, individuals has increased continuously over the past few decades (Mancini and de Melo 2017;Onge, Miller, and Motycka 2016). The escalating trend was specifically monitored since 1980; 1980-2014 was reportedly the major transition period of lifestyle modification with reportedly double the prevalence of obesity (Mancini and de Melo 2017;Onge, Miller, and Motycka 2016). If this trend continues to grow similarly, more than three billion of the world's population will be affected by overweight or obesity by 2030 (Hassannejad et al. 2017). ...
Article
Objectives: This systematic review and meta-analysis is based on randomized controlled trials evaluating the effect of physical activity on weight loss in adults undergoing bariatric surgery. The study compared certain biomarkers for individuals with and without physical activity after bariatric surgery. Secondary, the study identified potential successful interventions for the target population. Method: PubMed, Embase, OVID, CINAHL, and Cochrane Library were searched from January 2000 to December 2020. Intervention studies on the effect of physical activity in adults after bariatric surgery were selected, included, and analyzed following the PRISMA guidelines. The primary outcome was weight loss followed by selected biomarkers. Results: Two independent reviewers extracted data and conducted quality assessments. Of the 11 studies included, six reported BMI, two reported fat-free mass, three reported fat mass, two reported waist-hip ratio, and two reported waist circumference. Six studies measuring change from baseline BMI reported a significant intervention effect: SMD = -0.93 (-1.65;-0.20) with high heterogeneity of included trials (I2 =72%). There was no significant difference between control and intervention groups for other outcomes. Conclusion: BMI as a measure of physical activity positively impacts the target population. Large-scale studies with better criteria and a longer evaluation follow-up may finalize pronounced outcomes. Keywords: intervention, bariatric surgery, physical activity, surgery patients
... A prominent example of GLP-1RA on the market, manufactured by Novo Nordisk, is the broadly used liraglutide, and was approved in 2010 (Saxenda TM ) [59] for weight management. This medication is to be used in addition to lifestyle changes regarding diet and exercise (not indicated for pediatric use) [60], and not indicated to treat T2DM, given in a higher dose than the first liraglutide [61][62][63]. The first liraglutide by Novo Nordisk was Victoza TM (original approval date 2010), which is indicated for pediatric and adult use (> 10 years) and is also used as an adjunct to lifestyle changes. ...
Article
Full-text available
Pharmaceutical interventions play a vital role in managing various conditions, including weight-related issues such as obesity. In this context, lifestyle changes are often challenging to maintain, especially for individuals struggling with this condition. Obesity is strongly linked to serious health conditions like cardiovascular disease and insulin resistance, leading to a cascade of health risks. Importantly, the development of effective and safe weight loss medications has been challenging. Diabetes mellitus (DM), the incidence of which is also rising, is closely related to obesity. The annual rate of DM cases has increased significantly, mirroring trends in obesity. Pharmaceutical companies have made significant progress in developing drugs that address both diabetes and obesity. Glucagon-like peptide-1 receptor agonists (GLP-1RAs) have emerged as a promising class of medications with dual benefits in managing diabetes and aiding weight loss such as semaglutide, liraglutide, dulaglutide, exenatide, among others. However, despite their effectiveness, they can be expensive. The availability of various GLP-1RAs offers flexibility in diabetes management, but the surge in their prescription has led to a global shortage. Health authorities are working to address this issue, while pharmaceutical companies are exploring new paths to improve the quality of these drugs. In this context, tirzepatide stands out as a medication targeting key hormones involved in obesity and DM. Another potential breakthrough, retatrutide, is also being developed for these two conditions, but it requires further research. In this paper, the authors address all the GLP-1RA options developed to date, covering their mechanisms of action, efficacy, and chemical structures, among other aspects.
... Side Effects Liraglutide (Saxenda ® ) [32] Abdominal pain, constipation, vomiting, dyspepsia, fatigue, dizziness Orlistat [33] Steatorrhoea, bloating, oily spot, fecal urgency, fecal incontinence Naltrexone/bupropion (Mysimba ® ) [34] Increased risk of epileptic fits, nausea Lorcaserin (Belviq ® ) [35] Mitral regurgitation, depression, pulmonary hypertension Phentermine/topiramate (Qsymia™) [36] Paresthesia, dry mouth, dysgeusia, insomnia, dizziness, constipation ...
Article
Full-text available
Obesity is known as a transgenerational vicious cycle and has become a global burden due to its unavoidable complications. Modern approaches to obesity management often involve the use of pharmaceutical drugs and surgeries that have been associated with negative side effects. In contrast, natural antioxidants, such as flavonoids, have emerged as a promising alternative due to their potential health benefits and minimal side effects. Thus, this narrative review explores the potential protective role of flavonoids as a natural antioxidant in managing obesity. To identify recent in vivo studies on the efficiency of flavonoids in managing obesity, a comprehensive search was conducted on Wiley Online Library, Scopus, Nature, and ScienceDirect. The search was limited to the past 10 years; from the search, we identified 31 articles to be further reviewed. Based on the reviewed articles, we concluded that flavonoids offer novel therapeutic strategies for preventing obesity and its associated co-morbidities. This is because the appropriate dosage of flavonoid compounds is able to reduce adipose tissue mass, the formation of intracellular free radicals, enhance endogenous antioxidant defences, modulate the redox balance, and reduce inflammatory signalling pathways. Thus, this review provides an insight into the domain of a natural product therapeutic approach for managing obesity and recapitulates the transgenerational inheritance of obesity, the current available treatments to manage obesity and its side effects, flavonoids and their sources, the molecular mechanism involved, the modulation of gut microbiota in obesity, redox balance, and the bioavailability of flavonoids. In toto, although flavonoids show promising positive outcome in managing obesity, a more comprehensive understanding of the molecular mechanisms responsible for the advantageous impacts of flavonoids—achieved through translation to clinical trials—would provide a novel approach to inculcating flavonoids in managing obesity in the future as this review is limited to animal studies.
... GLP-1 targets the central nervous system (CNS) and pancreas to induce satiety and insulin secretion and several GLP-1 receptor agonists have been approved for the treatment of type 2 diabetes (Lu et al., 2020;Müller et al., 2019). In addition, GLP-1 receptor agonists exhibited promising utility for the management of obesity, as exemplified by the approval of the high-dose version of liraglutide (3 mgÁday À1 , Saxenda) (Onge et al., 2016). Notably, the recently published results from STEP (Semaglutide Treatment Effect in People) programmes of semaglutide showed that semaglutide treatment resulted in $16-18% body weight loss, thus demonstrating superior weight loss efficacy . ...
Article
Full-text available
Background and Purpose Glucagon‐like peptide‐1 (GLP‐1) and glucagon (GCG) receptor dual agonist have promising therapeutic effects in the treatment of obesity and diabetes. Moreover, GLP‐1 and cholecystokinin 2 (CCK2) dual agonists have been shown to restore pancreas function and improve glycaemic control in preclinical studies. We describe, for the first time, the beneficial effects of GLP‐1/glucagon receptor and GLP‐1/CCK2 dual agonists, which can be integrated into one peptide, resulting in significant anti‐diabetes and anti‐obesity effectiveness. Experimental Approach The in vitro potency of this novel peptide Xenopus (x) GLP‐1/GCG/CCK2 tri‐agonist (xGLP/GCG/gastrin) against GLP‐1, GCG, CCK1 and CCK2 receptors was determined on cells expressing the corresponding receptors by cAMP accumulation or ERK1/2 phosphorylation assays. The in vivo anti‐diabetes and anti‐obesity effects of this tri‐agonist xGLP/GCG/gastrin were studied in both db/db and diet induced obesity (DIO) mice. Key Results xGLP/GCG/gastrin was a potent and selective GLP‐1, GCG and CCK2 tri‐agonist. In DIO mice, the metabolic benefits of xGLP‐1/GCG/gastrin, such as reduction of body weight and hepatic lipid contents were significantly better than those of the peptide ZP3022 (GLP‐1/CCK‐2 dual agonist) and liraglutide. In a short‐term study in db/db mice, xGLP/GCG/gastrin treatment had considerable effects, increasing islet numbers, islet areas and insulin content. In a long‐term treatment study using db/db mice, xGLP‐1/GCG/gastrin showed a significantly and sustained improvement in glucose tolerance and glucose control compared with that of liraglutide, ZP3022, cotadutide (GLP‐1/GCG dual agonist) and xGLP/GCG‐15. Conclusions and Implications These results demonstrate the therapeutic potential of xGLP‐1/GCG/gastrin for the treatment of obesity and diabetes.
Chapter
Diabetes Mellitus (DM) receives significant attention from most scientists due to being a major global public health threat. Diabetes is considered one of the leading causes of death due to its micro and macrovascular complications. This insidious killer is estimated to reach a staggering number of 578 million (700 million by 2045) cases by 2030. In this book, utilizing the disciplines of clinical sciences, various contemporary topics related to diabetes are extensively discussed. These include the classification of diabetes, underlying causes, epidemiology, pathogenesis, impact on sexual functions, association with cancer, clinical manifestations, diagnostic methods, complications, and treatment options. Furthermore, intriguing and current subjects such as ""antidiabetic phytotherapy"" and ""the relationship between oral health and diabetes"" are also covered. By doing so, readers will acquire comprehensive and detailed knowledge about the clinical management of diabetes. Thus, this book not only serves those who seek to understand the scientific aspects of diabetes but also proves to be a valuable resource for healthcare professionals, researchers, and students in a clinical setting. We believe that this book will contribute to understanding the complexity of diabetes and provide beneficial solutions while shedding light on future studies.
Article
Pharmacotherapies for obesity and type 2 diabetes (T2D) are thought to bridge the gap between lifestyle modification and the weight loss obtained with bariatric surgery. Although the effect of monotherapies, namely amylin and glucagon-like peptide-1 receptor (GLP-1R) agonists, has shown great potential, combination therapy is now becoming a strategy to optimize efficacy for weight management while minimizing adverse effects. This study investigated a dual amylin and calcitonin receptor agonist (DACRA); KBP-066A in combination with the GLP-1R agonist semaglutide or the sodium-glucose co transporter-2 inhibitor (SGLT2i) empagliflozin for anti-obesity and anti-diabetic treatment. The effect of KBP-066A, semaglutide, and empagliflozin alone and in combination was studied with respect to their impact on body weight, food intake, and glucose metabolism in high-fat diet (HFD) and Zucker diabetic fatty (fa/fa) (ZDF) rats. Treatment with KBP-066A and semaglutide lowered body weight by 13% and 9.7%. In contrast, a combination of both KBP-066A + semaglutide reduced body weight by 21% in HFD rats demonstrating superiority compared to monotherapies alone. A combination of KBP-066A with semaglutide or empagliflozin significantly lowered fasting blood glucose, and HbA1C (%) levels in ZDF rats. The complementary action by KBP-066A to GLP-1R agonist and SGLT2i on BW, food intake and glucose control endorsed the potential of DACRAs as an add-on therapy to therapeutic options for T2D and obesity.
Article
Full-text available
Obesity is a significant health concern, as it causes a massive cascade of chronic inflammations and multiple morbidities. Rheumatoid arthritis and osteoarthritis are chronic inflammatory conditions and often manifest as comorbidities of obesity. Adipose tissues serve as a reservoir of energy as well as releasing several inflammatory cytokines (including IL-6, IFN-γ, and TNF-α) that stimulate low-grade chronic inflammatory conditions such as rheumatoid arthritis, osteoarthritis, diabetes, hypertension, cardiovascular disorders, fatty liver disease, oxidative stress, and chronic kidney diseases. Dietary intake, low physical activity, unhealthy lifestyle, smoking, alcohol consumption, and genetic and environmental factors can influence obesity and arthritis. Current arthritis management using modern medicines produces various adverse reactions. Medicinal plants have been a significant part of traditional medicine, and various plants and phytochemicals have shown effectiveness against arthritis and obesity; however, scientifically, this traditional plant-based treatment option needs validation through proper clinical trials and toxicity tests. In addition, essential oils obtained from aromatic plants are being widely used as for complementary therapy (e.g., aromatherapy, smelling, spicing, and consumption with food) against arthritis and obesity; scientific evidence is necessary to support their effectiveness. This review is an attempt to understand the pathophysiological connections between obesity and arthritis, and describes treatment options derived from medicinal, spice, and aromatic plants. (Full-text link: https://www.mdpi.com/2072-6643/14/5/985)
Chapter
Lifestyle modification continues to be the basis of obesity treatment despite the lack of substantial long-term efficacy. Pharmacotherapy is an important viable treatment option for many patients who do not respond to lifestyle therapy and who are not candidates for bariatric surgery. When associated with the intragastric balloon treatment, pharmacotherapy can be an excellent tool for optimizing results. In this chapter, we will address the current antiobesity drugs that can be used concomitantly with the intragastric balloon, as well as the symptomatic drugs used to reduce the common adverse effects of this procedure, and pharmacologic strategies for risk reduction and complications of the method.
Article
Full-text available
The long-term effects of sibutramine treatment on the rates of cardiovascular events and cardiovascular death among subjects at high cardiovascular risk have not been established. METHODS: We enrolled in our study 10,744 overweight or obese subjects, 55 years of age or older, with preexisting cardiovascular disease, type 2 diabetes mellitus, or both to assess the cardiovascular consequences of weight management with and without sibutramine in subjects at high risk for cardiovascular events. All the subjects received sibutramine in addition to participating in a weight-management program during a 6-week, single-blind, lead-in period, after which 9804 subjects underwent random assignment in a double-blind fashion to sibutramine (4906 subjects) or placebo (4898 subjects). The primary end point was the time from randomization to the first occurrence of a primary outcome event (nonfatal myocardial infarction, nonfatal stroke, resuscitation after cardiac arrest, or cardiovascular death). RESULTS: The mean duration of treatment was 3.4 years. The mean weight loss during the lead-in period was 2.6 kg; after randomization, the subjects in the sibutramine group achieved and maintained further weight reduction (mean, 1.7 kg). The mean blood pressure decreased in both groups, with greater reductions in the placebo group than in the sibutramine group (mean difference, 1.2/1.4 mm Hg). The risk of a primary outcome event was 11.4% in the sibutramine group as compared with 10.0% in the placebo group (hazard ratio, 1.16; 95% confidence interval [CI], 1.03 to 1.31; P=0.02). The rates of nonfatal myocardial infarction and nonfatal stroke were 4.1% and 2.6% in the sibutramine group and 3.2% and 1.9% in the placebo group, respectively (hazard ratio for nonfatal myocardial infarction, 1.28; 95% CI, 1.04 to 1.57; P=0.02; hazard ratio for nonfatal stroke, 1.36; 95% CI, 1.04 to 1.77; P=0.03). The rates of cardiovascular death and death from any cause were not increased. CONCLUSIONS: Subjects with preexisting cardiovascular conditions who were receiving long-term sibutramine treatment had an increased risk of nonfatal myocardial infarction and nonfatal stroke but not of cardiovascular death or death from any cause. (Funded by Abbott; ClinicalTrials.gov number, NCT00234832.)
Article
Full-text available
Liraglutide is an acylated glucagon-like peptide-1 analogue with 97 % amino acid homology with native glucagon-like peptide-1 and greatly protracted action. It is widely used for the treatment of type 2 diabetes mellitus, and administered by subcutaneous injection once daily. The pharmacokinetic properties of liraglutide enable 24-h exposure coverage, a requirement for 24-h glycaemic control with once-daily dosing. The mechanism of protraction relates to slowed release from the injection site, and a reduced elimination rate owing to metabolic stabilisation and reduced renal filtration. Drug exposure is largely independent of injection site, as well as age, race and ethnicity. Increasing body weight and male sex are associated with reduced concentrations, but there is substantial overlap between subgroups; therefore, dose escalation should be based on individual treatment outcome. Exposure is reduced with mild, moderate or severe renal or hepatic impairment. There are no clinically relevant changes in overall concentrations of various drugs (e.g. paracetamol, atorvastatin, griseofulvin, digoxin, lisinopril and oral combination contraceptives) when co-administered with liraglutide. Pharmacodynamic studies show multiple beneficial actions with liraglutide, including improved fasting and postprandial glycaemic control (mediated by increased insulin and reduced glucagon levels and minor delays in gastric emptying), reduced appetite and energy intake, and effects on postprandial lipid profiles. The counter-regulatory hormone response to hypoglycaemia is largely unaltered. The effects of liraglutide on insulin and glucagon secretion are glucose dependent, and hence the risk of hypoglycaemia is low. The pharmacokinetic and pharmacodynamic properties of liraglutide make it an important treatment option for many patients with type 2 diabetes.
Article
Full-text available
Weight loss of 5% to 10% can improve type 2 diabetes and related comorbidities. Few safe, effective weight-management drugs are currently available. To investigate efficacy and safety of liraglutide vs placebo for weight management in adults with overweight or obesity and type 2 diabetes. Fifty-six-week randomized (2:1:1), double-blind, placebo-controlled, parallel-group trial with 12-week observational off-drug follow-up period. The study was conducted at 126 sites in 9 countries between June 2011 and January 2013. Of 1361 participants assessed for eligibility, 846 were randomized. Inclusion criteria were body mass index of 27.0 or greater, age 18 years or older, taking 0 to 3 oral hypoglycemic agents (metformin, thiazolidinedione, sulfonylurea) with stable body weight, and glycated hemoglobin level 7.0% to 10.0%. Once-daily, subcutaneous liraglutide (3.0 mg) (n = 423), liraglutide (1.8 mg) (n = 211), or placebo (n = 212), all as adjunct to 500 kcal/d dietary deficit and increased physical activity (≥150 min/wk). Three coprimary end points: relative change in weight, proportion of participants losing 5% or more, or more than 10%, of baseline weight at week 56. Baseline weight was 105.7 kg with liraglutide (3.0-mg dose), 105.8 kg with liraglutide (1.8-mg dose), and 106.5 kg with placebo. Weight loss was 6.0% (6.4 kg) with liraglutide (3.0-mg dose), 4.7% (5.0 kg) with liraglutide (1.8-mg dose), and 2.0% (2.2 kg) with placebo (estimated difference for liraglutide [3.0 mg] vs placebo, -4.00% [95% CI, -5.10% to -2.90%]; liraglutide [1.8 mg] vs placebo, -2.71% [95% CI, -4.00% to -1.42%]; P < .001 for both). Weight loss of 5% or greater occurred in 54.3% with liraglutide (3.0 mg) and 40.4% with liraglutide (1.8 mg) vs 21.4% with placebo (estimated difference for liraglutide [3.0 mg] vs placebo, 32.9% [95% CI, 24.6% to 41.2%]; for liraglutide [1.8 mg] vs placebo, 19.0% [95% CI, 9.1% to 28.8%]; P < .001 for both). Weight loss greater than 10% occurred in 25.2% with liraglutide (3.0 mg) and 15.9% with liraglutide (1.8 mg) vs 6.7% with placebo (estimated difference for liraglutide [3.0 mg] vs placebo, 18.5% [95% CI, 12.7% to 24.4%], P < .001; for liraglutide [1.8 mg] vs placebo, 9.3% [95% CI, 2.7% to 15.8%], P = .006). More gastrointestinal disorders were reported with liraglutide (3.0 mg) vs liraglutide (1.8 mg) and placebo. No pancreatitis was reported. Among overweight and obese participants with type 2 diabetes, use of subcutaneous liraglutide (3.0 mg) daily, compared with placebo, resulted in weight loss over 56 weeks. Further studies are needed to evaluate longer-term efficacy and safety. clinicaltrials.gov Identifier:NCT01272232.
Article
Full-text available
Background Obesity is a chronic disease with serious health consequences, but weight loss is difficult to maintain through lifestyle intervention alone. Liraglutide, a glucagon-like peptide-1 analogue, has been shown to have potential benefit for weight management at a once-daily dose of 3.0 mg, injected subcutaneously. Methods We conducted a 56-week, double-blind trial involving 3731 patients who did not have type 2 diabetes and who had a body-mass index (BMI; the weight in kilograms divided by the square of the height in meters) of at least 30 or a BMI of at least 27 if they had treated or untreated dyslipidemia or hypertension. We randomly assigned patients in a 2:1 ratio to receive once-daily subcutaneous injections of liraglutide at a dose of 3.0 mg (2487 patients) or placebo (1244 patients); both groups received counseling on lifestyle modification. The coprimary end points were the change in body weight and the proportions of patients losing at least 5% and more than 10% of their initial body weight. Results At baseline, the mean (±SD) age of the patients was 45.1±12.0 years, the mean weight was 106.2±21.4 kg, and the mean BMI was 38.3±6.4; a total of 78.5% of the patients were women and 61.2% had prediabetes. At week 56, patients in the liraglutide group had lost a mean of 8.4±7.3 kg of body weight, and those in the placebo group had lost a mean of 2.8±6.5 kg (a difference of −5.6 kg; 95% confidence interval, −6.0 to −5.1; P<0.001, with last-observation-carried-forward imputation). A total of 63.2% of the patients in the liraglutide group as compared with 27.1% in the placebo group lost at least 5% of their body weight (P<0.001), and 33.1% and 10.6%, respectively, lost more than 10% of their body weight (P<0.001). The most frequently reported adverse events with liraglutide were mild or moderate nausea and diarrhea. Serious events occurred in 6.2% of the patients in the liraglutide group and in 5.0% of the patients in the placebo group. Conclusions In this study, 3.0 mg of liraglutide, as an adjunct to diet and exercise, was associated with reduced body weight and improved metabolic control. (Funded by Novo Nordisk; SCALE Obesity and Prediabetes NN8022-1839 ClinicalTrials.gov number, NCT01272219.)
Article
Full-text available
The pathophysiology of type2 diabetes differs between different ethnic groups. Asians develop type2 diabetes at younger age, lower body mass index, and in relatively short time. Not only that, some ethnicities have different responses and dosing regimens to different classes of anti-diabetic agents. Data from Japanese population showed that the optimal doses of liraglutide used are smaller than other population and that weight loss is not as effective as seen in Caucasians. We aimed to assess liraglutide efficacy in reducing weight and HbA1c in Arab population when used as add on to other anti-diabetic agents. We prospectively followed patients who were recruited to treatment with liraglutide for a 6 months period; at the start of the study we checked patients' demographics, weight, blood pressure, fasting blood glucose, HbA1c, lipid panel, LFTs and creatinine. Patients were checked at 3 months and at the end of the study at 6 months. There was a significant reduction in weight at 3 and 6 months from a mean weight of 96.01 ± 19.2 kg to (94.8 ± 20 kg with (P < 0.001)) and 94.5 ± 19 kg with (p < 0.001) respectively. Mean HbA1c at baseline was 8.3 ± 1.7 % dropped to 7.7 ± 1.4 % (p < 0.001) at 3 months, and 7.6 + 1.6 % (p < 0.001) at 6 months. Liraglutide is effective in reducing weight, HbA1c as well as other metabolic parameters in Arab population with type2 diabetes. The trial is approved and registered with the Institutional Ethical Committee Board (Dubai Health Authority Medical Research Committee) under registration Number (MRC-08/2013_03).
Article
Subcutaneous liraglutide (Victoza((R))), a glucagon-like peptide 1 receptor agonist, is approved for the treatment of adult patients with type 2 diabetes mellitus. Once-daily liraglutide, as monotherapy or add-on therapy to other antidiabetic agents (including basal insulin), was an effective and generally well tolerated treatment in adult patients with type 2 diabetes in several well-designed phase III trials and in the real world clinical practice setting. In addition to improving glycaemic control, liraglutide had beneficial effects on bodyweight, systolic blood pressure and surrogate measures of beta-cell function in clinical trials, with these benefits maintained during long-term treatment (<= 2 years). Liraglutide has a convenient once-daily administration regimen, a low potential for drug-drug interactions and low propensity to cause hypoglycaemia. Thus, liraglutide continues to be a useful option for the management of type 2 diabetes. This article reviews the therapeutic use of liraglutide in adult patients with type 2 diabetes and summarizes its pharmacological properties.
Article
Obesity continues to pose a major public health risk to the United States and across the world, with an estimated one-third of adult Americans being defined as obese. Obesity treatment guidelines recommend the use of pharmacologic therapy in adults who have a body mass index (BMI) of 30 kg/m(2) or higher or in patients with a BMI of 27 kg/m(2) or higher who have at least one weight-related comorbid condition (e.g., hypertension, dyslipidemia, insulin resistance, type 2 diabetes mellitus). Liraglutide is a glucagon-like peptide-1 receptor agonist that has been successfully used in the treatment of type 2 diabetes for several years. Weight loss has been well described as an additional benefit with liraglutide therapy, which prompted the manufacturer to evaluate and develop a higher dose formulation specifically for the treatment of obesity. Liraglutide 3 mg/day was approved by the U.S. Food and Drug Administration for this indication in December 2014. We performed a search of the Medline database to identify relevant literature focused on liraglutide's role specifically in treating obesity. Five clinical trials with this primary end point were identified. Data demonstrated that liraglutide can successfully achieve weight-loss benchmarks of 5% or more and 10% or more loss from baseline. The most common adverse effects were gastrointestinal and mild to moderate in intensity. The cost of therapy is high, averaging over $1000/month for out-of-pocket expenses if insurance coverage is not available. Liraglutide is also available for delivery only by subcutaneous injection, which may represent a barrier for patients. Liraglutide 3 mg/day represents another pharmacologic option for the treatment of obesity.
Article
Globally, obesity has reached epidemic proportions and poses an ever increasing burden from a societal and healthpayer perspective. Although lifestyle interventions are fundamental in its management, in the real world setting most obese or overweight adults require adjunctive pharmacotherapy to achieve clinically relevant reductions in bodyweight (i.e. a ≥5 % reduction). Subcutaneous liraglutide (Saxenda(®)) 3 mg once daily is indicated as an adjunct to a reduced-calorie diet and increased physical activity for chronic bodyweight management in adults with an initial body mass index (BMI) of ≥30 kg/m(2) (obese) or a BMI of ≥27 kg/m(2) (overweight) and at least one bodyweight-related comorbidity [e.g. hypertension, dyslipidaemia, type 2 diabetes mellitus or obstructive sleep apnoea (OSA)]. In phase III trials (32 or 56 weeks' duration) in these populations, subcutaneous liraglutide was associated with clinically relevant reductions in fasting bodyweight and was generally well tolerated. Liraglutide was significantly more effective than placebo in terms of reductions in fasting bodyweight and waist circumference, and improvements in some biomarkers of cardiovascular risk. Improvements in bodyweight were maintained after up 2 years of liraglutide therapy. In nondiabetic adults with moderate to severe OSA, liraglutide improved apnoea-hypopnoea index scores at 32 weeks, which was largely driven by significant reductions in bodyweight. In the absence of head-to-head trials, the relative position of individual anti-obesity drugs remains to be fully determined. In the meantime, liraglutide is an emerging option, as an adjunct to a reduced-calorie diet and increased physical activity, for chronic bodyweight management in obese adults and overweight adults with at least one bodyweight-related comorbidity.