Abstract
Introduction
Semaglutide, a peptidic GLP-1 receptor agonist, has been clinically approved for treatment of type 2 diabetes mellitus and is available in subcutaneous and oral dosage form. Diabetes, insulin resistance, and obesity are responsible for the pathological manifestations of non-alcoholic steatohepatitis (NASH). Similarly, insulin resistance in brain is also responsible for neurodegeneration and impaired cognitive functions.
Background
Observations from phase-3 clinical trials like SUSTAIN and PIONEER indicated anti-obesity potential of semaglutide, which was established in STEP trials. Various pre-clinical and phase-2 studies have indicated the therapeutic potential of semaglutide in non-alcoholic steatohepatitis and neurodegenerative disorders like Parkinson’s and Alzheimer’s disease.
Discussion
Significant weight reduction ability of semaglutide has been demonstrated in various phase-3 clinical trials, for which recently semaglutide became the first long-acting GLP-1 receptor agonist to be approved by the United States Food and Drug Administration for management of obesity. Various pre-clinical and clinical studies have revealed the hepatoprotective effect of semaglutide in NASH and neuroprotective effect in Parkinson’s and Alzheimer’s disease.
Conclusion
Many GLP-1 receptor agonists have shown hepatoprotective and neuroprotective activity in animal and human trials. As semaglutide is an already clinically approved drug, successful human trials would hasten its inclusion into therapeutic treatment of NASH and neurodegenerative diseases. Semaglutide improves insulin resistance, insulin signalling pathway, and reduce body weight which are responsible for prevention or progression of NASH and neurodegenerative diseases.
KEY WORDS: GLP-1 receptor agonist, metabolic diseases, neurodegenerative diseases, obesity, semaglutide
INTRODUCTION
Glucagon Like Peptide-1 (GLP-1), secreted from nutrient responsive enteroendocrine cells or L cells of intestine, is a prominent incretin hormone responsible for glucoregulatory effect in human body. GLP-1 augments glucose-dependent insulin secretion, improves β-cell functioning and glucose tolerance, inhibits glucagon secretion, and offers protection from glucolipotoxicity, apoptosis (1, 2). GLP-1 plays a vital role in ameliorating insulin resistance and production of VLDL (Very Low-Density Lipoprotein) via an intrinsic gut-liver signalling pathway (3). Although expression of GLP-1 receptors in hepatocytes remained a subject of debate among researchers, an insulin-independent direct action on liver has been suggested (4). GLP-1 plays an important role in digestion by delaying gastric emptying and suppress appetite centre in hypothalamus (5). GLP-1 is expressed in different parts of the brain and affects satiety centre, water intake, stress reaction, neurogenesis, neurodegeneration, thermogenesis, and energy homeostasis (6). Therefore, GLP-1 receptor agonists have been found to improve cognitive impairment and neurological deficit associated with diabetes or obesity (7). It suggests the potential of GLP-1 receptor agonists which can be deployed for treatment of diabetes, obesity, Non-alcoholic Steatohepatitis (NASH) and neurodegenerative diseases like Parkinson’s and Alzheimer’s.
Researchers focused on developing GLP-1 receptor agonists which needs less frequent dosing (weekly-once) to improve patient adherence and to reduce the treatment burden. Only three GLP-1 receptor agonists- exenatide extended release, semaglutide and dulaglutide are currently approved for weekly-once dosing (8). Semaglutide (C187H291N45O59) is a longer-acting GLP-1 receptor agonist with 94% structural similarity to the native GLP-1 (9). Semaglutide can be safely administered to adults and geriatric patients with renal, hepatic or cardiovascular (CV) disorders. It is available as Ozempic® (subcutaneous injection, weekly-once dosing; available in 0.5, 1.0 mg strength) and Rybelsus® (oral tablets, once-daily dosing; available in 3, 7, 14 mg strength), both approved by USFDA, Health Canada, European Medicines Agency, Japanese Health ministry for use in treatment of type 2 diabetes. Semaglutide has been found to reduce hyperglycaemia, body weight, steatosis, and improves cognitive ability in neurodegenerative diseases (Fig. 1). The status of semaglutide in treatment of type 2 diabetes has been well documented (10); hence this narrative review will discuss the therapeutic potential of semaglutide in three different sections- management of obesity, NASH and neurodegenerative diseases.
Fig. 1.
Mechanism of semaglutide for management of obesity, NASH, and neurodegenerative diseases.
OBESITY
Excess accumulation of body fat or a body mass index (BMI) of ≥ 30 indicates obesity, which is a life-style disorder caused by more calorie intake, reduced physical activity, endocrinological conditions, genetic factors and certain medications (11). Central obesity is often accompanied with metabolic syndrome characterized by hyperglycemia, hypertension and dyslipidemia. (12)
Pathogenesis
White adipose tissue is composed of mature fat cells or adipocytes which stores excess energy obtained from food as triglycerides and also hosts various immune cells like fibroblasts, macrophages, and lymphocytes. Adipose tissue dysfunction in obesity leads to imbalance in the distribution and activity of immune cells causing local as well as systemic inflammation (13). In obesity, radical changes occur in immune cell composition of adipose tissue which activates inflammatory signaling pathways with increased expression of inflammatory receptors (14). Visceral fat in obesity is linked to the display of inflammatory markers and development of insulin resistance (15). Research suggests that obesity with metabolic syndrome is linked to increased deaths among patients with type 2 diabetes and cardiovascular diseases. Homeostatic and hedonic pathway are two important systems for regulating food and energy intake in humans. Homeostatic system, controlled by hypothalamus and brain stem, stimulates food intake in case of low energy stores (16). Hedonic pathway represents reward or motivational aspects of food intake and thus stimulates intake of food during relative energy deficiency. In addition, it also stimulates intake of highly palatable foods during periods of low energy demand (17). Altered sympathetic nervous system activity contributes to obesity; overeating increases sympathetic nervous system activity (18), whereas fasting reduces this activity (19). Reduced serotonin signaling in hypothalamus causes overeating by impairing the negative feedback on food intake and thus contributes to obesity (20). Similarly, decrease in dopamine signaling causes overeating of palatable foods (21). In response to food intake, secretion of ghrelin or hunger hormone is suppressed, but this regulation is hampered in obesity. (22) In obesity, food consumption is associated with reduced activity of anorexigenic hormones like GLP-1, Peptide YY (PYY) and cholecystokinin (CCK). Leptin counterbalance the effect of ghrelin and also helps in synthesizing anorexigenic peptides. But in obese individuals, leptin signaling is impaired leading to overconsumption of food (23). Development of insulin resistance in brain impairs the insulin mediated response to food intake and metabolism in obesity. (24) Disturbance in circadian rhythm and altered timing of food intake causes gain in body weight (25); hence the night-shift workers are more prone to develop obesity (26). Apart from these factors, childhood-onset obesity is also caused by genetic predisposition (monogenic or polygenic mutations). (27)
GLP-1 is an intestinal incretin hormone which shows anorectic property centrally as well as peripherally by enhancing satiation and delayed gastric emptying (28). Various GLP-1 receptor agonists like exenatide, liraglutide, lixisenatide, dulaglutide and semaglutide have shown to reduce the body weight by reducing energy intake (29). Tirzepatide is a newer dual agonist of GLP-1 and GIP (gastric inhibitory peptide) which has shown promising anti-obesity effect. (30)
Anti-obesity Mechanism of Semaglutide
Semaglutide has exhibited significant weight loss in comparison to liraglutide and exenatide in various clinical studies (31). The weight loss has been attributed to reduced energy intake because semaglutide doesn’t affect energy expenditure and that too without raising the resting metabolic rate. It also improves the hedonic feature of food intake, i.e., less cravings for food due to enhanced satiation, less likeness for high fat foods (31, 32). Although a gut to brain communication is involved in mediating reduced appetite and energy intake of semaglutide, but the exact mechanism is still unclear (33, 34). It has been proposed that semaglutide’s effect on suppressed appetite might be facilitated through hypothalamus by a central mechanism similar to that of liraglutide (34). Another plausible explanation involves a peripheral mechanism similar to native GLP-1 which causes delay in gastric motility. It also activates gastro-mechano receptors which in turn suppresses the satiation centre situated in brainstem by relaying signals through vagus nerve (35, 36).
Clinical Trials for Anti-obesity Effect of Semaglutide
Semaglutide has exhibited significant weight reducing property in various phase-3 SUSTAIN and PIONEER trials (presented in Table I and Table II respectively) over dulaglutide and liraglutide (31). These results encouraged the researchers to carry out trials on weekly-once s.c. semaglutide for better management of obesity.
Table I.
Weight Reduction Property of Weekly-once s.c. Semaglutide in SUSTAIN Trials
SUSTAIN | Clinical trial identification number | Population | Duration (weeks) | Background medication | Semaglutide trial dose & Comparator | Weight loss (kg) | |
---|---|---|---|---|---|---|---|
1 | NCT02054897 | 388 | 30 | None |
0.5 mg 1.0 mg Placebo |
-3.73 -4.53 -0.98 |
|
2 | NCT01930188 | 1231 | 56 | Metformin and/or Pioglitazone/Rosiglitazone |
0.5 mg 1.0 mg Sitagliptin (100 mg) |
-4.3 -6.1 -1.9 |
|
3 | NCT01885208 | 813 | 56 | Metformin and/or Thiazolidinediones and/or Sulfonylurea |
1.0 mg Exenatide ER (2.0 mg) |
-5.6 -1.9 |
|
4 | NCT02128932 | 1089 | 30 | Metformin alone or with Sulfonylurea |
0.5 mg 1.0 mg Insulin glargine |
-3.47 -5.17 + 1.15 |
|
5 | NCT02305381 | 397 | 30 | Basal insulin alone or with Metformin |
0.5 mg 1.0 mg Placebo |
-3.7 -6.4 -1.4 |
|
6 | NCT01720446 | 3297 (83% with CV and/or CKD) | 104 | None |
0.5 mg 1.0 mg Placebo (0.5 mg) Placebo (1.0 mg) |
-3.6 -4.9 -0.7 -0.5 |
|
7 | NCT02648204 | 1201 | 40 | Metformin |
0.5 mg 1.0 mg Dulaglutide (0.75 mg) Dulaglutide (1.5 mg) |
-4.6 -6.5 -2.3 -3.0 |
|
8 | NCT03136484 | 788 | 52 | Metformin |
1.0 mg Canagliflozin (300 mg) |
-5.3 -4.2 |
|
9 | NCT03086330 | 302 | 30 | SGLT-2 inhibitor alone or with Sulfonylurea/Metformin |
1.0 mg Placebo |
-4.7 -0.9 |
|
10 | NCT03191396 | 577 | 30 | Metformin and Sulfonylurea/SGLT-2 inhibitor |
1.0 mg Liraglutide (1.2 mg) |
-5.8 -1.9 |
|
11 | NCT03689374 | 2275 | 52 | Metformin alone or with Sulfonylurea/Meglitinide/DPP-4 inhibitor/α-glucosidase inhibitor |
1.0 mg Insulin aspart (4 IU) |
- - |
|
Japan-sitagliptin | NCT02254291 | 308 | 30 | None |
0.5 mg 1.0 mg Sitagliptin (100 mg) |
-2.2 -3.9 0.0 |
|
Japan | NCT02207374 | 601 | 56 | None |
0.5 mg 1.0 mg OAD monotherapy |
-1.4 -3.2 + 0.4 |
|
China-MRCT | NCT03061214 | 868 | 30 | Metformin |
0.5 mg 1.0 mg Sitagliptin (100 mg) |
-2.9 -4.2 -0.4 |
|
FORTE | NCT03989232 | 961 | 40 | Metformin alone or with Sulfonylurea |
1.0 mg 2.0 mg Placebo |
Trial product estimand | Treatment policy estimand |
-6.0 kg -6.9 kg - |
-5.6 kg -6.4 kg - |
Weight reduction activity of weekly once s.c. semaglutide (0.5, 1.0, and 2.0 mg)
Table II.
Weight Reduction Property of Daily-Once Oral Semaglutide in PIONEER Trials
PIONEER | Clinical trial identification number | Population | Duration (weeks) | Background medication | Semaglutide trial dose & Comparator | Trial product estimand | Treatment policy estimand |
---|---|---|---|---|---|---|---|
Weight loss | Weight loss | ||||||
1 | NCT02906930 | 703 | 26 | None |
3 mg 7 mg 14 mg Placebo |
-0.2 -1.0 -2.6 -1.5 |
-0.1 -0.9 -2.3 -1.4 |
2 | NCT02863328 | 822 | 52 | Metformin |
14 mg Empagliflozin (25 mg) |
-4.7 -3.8 |
-3.8 -3.6 |
3 | NCT02607865 | 1864 | 78 | Metformin with/without Sulfonylurea |
3 mg 7 mg 14 mg Sitagliptin (100 mg) |
-1.8 -2.7 -3.5 -1.1 |
-1.8 -2.7 -3.2 -1.0 |
4 | NCT02863419 | 711 | 52 |
Metformin with/without SGLT-2 inhibitor |
14 mg Liraglutide (s.c. 1.8 mg) Placebo |
-5.0 -3.1 -1.2 |
-4.3 -3.0 -1.0 |
5 | NCT02827708 | 324 (with moderate renal impairment) | 26 |
Metformin/Sulfonylurea, or both, or Basal insulin with/without Metformin |
14 mg Placebo |
-3.7 -1.1 |
-3.4 -0.9 |
6 | NCT02692716 | 3183 (84.7% with CV or CKD) |
Event-driven; Median time = 69 weeks |
None |
14 mg Placebo |
-4.2 kg -0.8 kg |
|
7 | NCT02849080 | 504 | 52 |
One/two amongst Metformin, Sulfonylureas, SGLT-2 inhibitors, or Thiazolidinediones |
14 mg Sitagliptin (100 mg) |
-2.9 -0.8 |
-2.6 -0.7 |
8 | NCT03021187 | 731 | 52 |
Insulin with/without Metformin |
3 mg 7 mg 14 mg Placebo |
-1.0 -2.9 -4.3 + 0.6 |
-0.8 -2.0 -3.7 + 0.5 |
9 | NCT03018028 | 243 | 52 | None |
3 mg 7 mg 14 mg Placebo Liraglutide (s.c. 0.9 mg) |
0.0 -0.6 -2.8 -1.0 + 0.4 |
-0.3 -0.8 -2.6 -0.6 0.0 |
10 | NCT03015220 | 458 | 52 |
Sulfonylurea/glinide/thia zolidinedione/α-glucosidase inhibitor/SGLT-2 inhibitor monotherapy |
3 mg 7 mg 14 mg Dulaglutide (s.c. 0.75 mg) |
+ 0.1 -1.0 -1.9 + 1.1 |
0.0 -0.9 -1.6 + 1.0 |
11 | NCT04109547 | 664 | 26 | None |
3 mg 7 mg 14 mg Placebo |
- - - - |
- - - - |
12 | NCT04017832 | 1444 | 26 | Metformin |
3 mg 7 mg 14 mg Placebo Sitagliptin (100 mg) |
- - - - - |
- - - - - |
TEENS | NCT04596631 | 132 (aged 10–17 years) | 52 | Metformin and/or basal insulin |
Semaglutide (maximum tolerated dose) Placebo |
- | - |
Weight reduction activity of once-daily oral semaglutide (3, 7, and 14 mg)
Phase-1 Studies
NCT02079870: A cross over trial study on 30 obese subjects investigated the mechanism of semaglutide by studying energy intake, appetite sensation, gastric emptying, and postprandial glucose as well as triglyceride metabolism. Participants received weekly-once s.c. semaglutide (initially 0.25, then 0.5 and finally 1.0 mg with dose escalation) or placebo for 12-weeks, followed by a washout period of 5 to 7 weeks, and finally another 12-weeks treatment period. Semaglutide has shown about 24% drop in net calorie consumption with 5 kg weight reduction. Therefore, lower energy intake, enhanced satiation, less food craving, reduced body fat and weight loss are the plausible mechanism for the anorexic effect of semaglutide (32).
The cross-over design was the primary limitation of this study. Patients who received semaglutide likely recovered the weight during wash-out period but may not have reached the pre-treatment level. This could be a reason for slight weigh gain in placebo group (32).
NCT03842202: This trial assessed the effect of semaglutide on energy intake, appetite and gastric emptying in 72 subjects with obesity, who received weekly-once s.c. semaglutide (2.4 mg) or placebo for 20 weeks. Reduced energy intake, better appetite control, less food cravings were responsible for the weight reduction (9.9%) exhibited by semaglutide. Delayed gastric emptying (assessed indirectly through paracetamol absorption) was not found to be associated with semaglutide (37).
Indirect measurement of paracetamol absorption has several limitations for predicting gastric emptying. However, the limitations were mitigated by providing paracetamol with semi-solid food, measurement of post-dose paracetamol concentration and time to reach the peak concentration (37).
NCT03600480: This phase-1b trial assessed the safety, efficacy of cagrilintide (amylin analogue) in combination with semaglutide on 96 obese subjects (without lifestyle modification). They received a combined therapy of weekly-once s.c. cagrilintide (0·16/0·30/0·60/1·2/2·4/4·5 mg) and weekly-once s.c. 2.4 mg semaglutide or placebo for 20-weeks. Superior weight reduction was achieved in combination therapy with good safety profile over placebo (38).
The weight reduction data is not solely due to semaglutide treatment, because this study evaluated the safety, tolerability of cagrilintide in combination with semaglutide. Short study duration and only 4 weeks of treatment with final target dose limits the evaluation accuracy. Exclusion of persons with BMI more than 40 kg/m (2) and high cardiovascular risk is also a major limitation, because severe obese persons are often associated with cardiovascular diseases. Inclusion of persons of either sex with non-childbearing potential is also another limitation. However, it was decided to include persons with childbearing potential in future trials (38).
Phase-2 Study
NCT02453711: A dose-ranging study was conducted on 957 obese subjects to explore the effectiveness of semaglutide in obesity management. Participants received daily-once s.c. semaglutide (initially 0.05, or 0.1 or 0.2 or 0.3 or 0.4 mg with incremental escalation) or daily-once 3.0 mg s.c. liraglutide (initially 0.6 mg, then dose escalation of 0.6 mg every week) or placebo for 52-weeks. Superior weight reduction of 6.0–13.8% and 11.4–16.3% were exhibited by the semaglutide with slow escalation (4-week period) and fast escalation (2-week period) respectively over liraglutide and placebo (39).
Unmasking of participants and site staffs to the assigned dose might have incorporated biasness in reporting adverse effects or treatment discontinuation. As the participants were not evaluated for the source of body weight loss and estimation of energy balance, it was difficult to determine whether the participants strictly adhered to the diet and exercise activity recommendations (39).
Phase-3 Studies
STEP (Semaglutide Treatment Effect in People with obesity) trials: All the trials were randomised, parallel-group and placebo-controlled (presented in Table III). Except for STEP-3 trial, the net energy expenditure for all trials was determined by: estimated basal metabolic rate x physical activity level value (1.3) (40).
Table III.
Weight Reduction Property of Weekly-once s.c. Semaglutide in STEP Trials
STEP | Objective | Exclusion criteria | Population | Duration (weeks) | Semaglutide trial dose | Weight loss (%) |
---|---|---|---|---|---|---|
1 (NCT03548935) | Efficacy of semaglutide in obese patients | Type 2 diabetic patients | 1961 | 68 |
2.4 mg Placebo |
-14.9 -2.4 |
2 (NCT03552757) | Efficacy of semaglutide in type 2 diabetic obese patients | Patients on insulin | 1210 | 68 |
2.4 mg Placebo |
-9.6 -3.4 |
3 (NCT03611582) | Efficacy of semaglutide along with intensive lifestyle program | Patients with HbA1c ≥ 6.5% | 611 | 68 |
2.4 mg Placebo |
-16.0 -5.7 |
4 (NCT03548987) | Efficacy of semaglutide in obese patients | Patients with HbA1c ≥ 6.5% | 902 | 68 |
2.4 mg Placebo (switch-on) |
-7.9 + 6.9 |
5 (NCT03693430) | Efficacy of semaglutide in obese patients | Patients with HbA1c ≥ 6.5% | 304 | 104 | 2.4 mg | - |
6 (NCT03811574) | Efficacy of semaglutide 1.7 mg and 2.4 mg in obese patients | Type 2 diabetic patients with renal impairment | 401 | 68 |
2.4 mg 1.7 mg Placebo |
-13.2 -9.6 -2.1 |
7 (NCT04251156) | Efficacy of semaglutide in obese patients | Type 2 diabetic patients with renal impairment | 375 | 44 | 2.4 mg | - |
8 (NCT04074161) | Efficacy of semaglutide as compared to liraglutide in obese patients | Type 2 diabetic patients | 338 | 68 |
2.4 mg Liraglutide 3.0 mg |
-15.8 -6.4 |
TEENS NCT04102189 | Efficacy of semaglutide in adolescents with obesity | Type 2 diabetic patients; Obesity of secondary origin (hypothalamic or endocrinal) | 163 | 68 | 1.0 mg | - |
Weight reduction activity of weekly-once semaglutide established by STEP (Semaglutide Treatment Effect in People with obesity) trials
STEP-1 (NCT03548935): This trial investigated the effectiveness of weekly-once s.c. 2.4 mg semaglutide on 1961 obese subjects, who received semaglutide (0.25 mg dose followed by 0.5 mg, 1.0 mg, 1.7 mg, 2.4 mg with dose escalation) or placebo for 68-weeks. A significant weight loss of 14.9% was achieved with semaglutide treated group over 2.4% with placebo. Apart from the gastrointestinal side effects, semaglutide was found to be safe and well-tolerated. A sub-population study demonstrated that semaglutide treatment causes reduction in fat mass as proved by whole body DXA scanning (40, 41).
Majority of the participants were women and belongs to White ethnicity. Persons with type 2 diabetes were excluded, but obesity and type 2 diabetes frequently co-occur in many patients. The DXA scanning was performed on a sub-population only which limits the accuracy of this data (41).
STEP-2 (NCT03552757): This trial explored the effectiveness of weekly-once s.c. 2.4 mg semaglutide on 1210 obese, type 2 diabetic subjects who received 1.0 mg semaglutide (initially 0.25, then 0.5, 1 mg) or 2.4 mg semaglutide (initially 0.25, then 0.5, 1.0, 1.7, 2.4 mg) or placebo for 68-weeks. A significant decrease of 9.6% in body weight was exhibited by the 2.4 mg semaglutide treated group over 3.4% with placebo (40, 42).
Patients on insulin therapy were excluded from this study. But weight loss effects due to add-on insulin therapy could have been studied as observed in SUSTAIN 5 (42).
STEP-3 (NCT03611582): Effectiveness of weekly-once 2.4 mg s.c. semaglutide was investigated on 611 obese subjects who received 2.4 mg semaglutide (initially 0.25, then 0.5, 1.0, 1.7, finally 2.4 mg) or placebo for 68-weeks. A significant decline in weight (16%) was exhibited by semaglutide over 5.7% with placebo (40, 43).
The relative benefit of behavioral therapy and low-calorie diet along with semaglutide therapy couldn’t be ascertained because it couldn’t identify the individual contribution towards weight reduction. This short duration study couldn’t make sure that persons treated with semaglutide would sustain this reduced weight after 68 weeks of treatment. Participants with HbA1c greater than 6.5% were excluded from this study (43).
STEP-4 (NCT03548987): Effectiveness of semaglutide was assessed on 902 obese subjects who received weekly-once s.c. semaglutide (initially 0.25 mg, 0.5, 1.0, 1.7, finally 2.4 mg) for initial 20-weeks. During maintenance period, 803 subjects (who achieved 2.4 mg dose during run-in period) randomly received either semaglutide (2.4 mg) or placebo for 48 weeks. Participants who reached the target dose may be switched to the placebo group to study the withdrawal effect of semaglutide therapy. A significant weight loss of 7.9% was observed during the maintenance period, whereas a mean weight loss of 10.6% was exhibited by the 2.4 mg semaglutide during the 68-week long trial. Participants who switched on to placebo treatment gained weight of 6.9% (40, 44).
Inflexibility in run-in period might have resulted in participants with more tolerance and adherence towards semaglutide therapy. It may cause biased results as the trial setting clearly differs from normal clinical settings. Contribution of lifestyle intervention couldn’t be assessed. The withdrawal design of the study may cause overestimation of weight loss in semaglutide treated population as compared to a normal participant. Participants with HbA1c greater than 6.5% were excluded from this study (44).
STEP-5 (NCT03693430): Long-term efficacy of weekly-once s.c. semaglutide is being investigated on 304 obese subjects (without type 2 diabetes but having at least one weight related co-morbidities like CV disorder/hypertension/dyslipidaemia/obstructive sleep apnoea). Participants will receive 2.4 mg s.c. semaglutide (initially 0.25, then 0.5, 1.0, 1.7, finally 2.4 mg) or placebo for 104-weeks. The results have not been published (40).
Participants with HbA1c greater than equal to 6.5% were excluded. Contribution of low-calorie diet and physical activity would be difficult to determine (45).
STEP-6 (NCT03811574): Effectiveness of two different doses of weekly-once s.c. semaglutide was explored on 401 East Asian obese type 2 diabetic subjects, who received 2.4 mg s.c. semaglutide (initially 0.25, then 0.5, 1.0, 1.7 and finally 2.4 mg) or placebo or 1.7 mg semaglutide (initially 0.25 mg, then 0.5, 1.0, then 1.7 mg) or placebo. Various parameters like weight loss, change in BMI, FPG, HbA1c, number of adverse events have been studied. A significant decline in weight was exhibited by 2.4 mg semaglutide (13.2%) and 1.7 mg semaglutide (9.6%) over 2.1% with placebo (46).
Type 2 diabetic patients who were also diagnosed with renal impairment or with uncontrolled diabetic retinopathy were excluded from this study. The long-term effects of semaglutide for management of chronic obesity couldn’t be studied in this study; whereas STEP-5 covers this aspect with a global population. Adherence to lifestyle intervention couldn’t be assessed in this study (46).
STEP-7 (NCT04251156): This on-going trial is aimed at exploring the effectiveness of semaglutide on 375 Chinese obese subjects (with weight related co-morbidity or type 2 diabetes), who receive 2.4 mg s.c. semaglutide or placebo for 44-weeks (47).
Type 2 diabetic patients who were also diagnosed with renal impairment or with uncontrolled diabetic retinopathy were excluded from this study (47).
STEP-8 (NCT04074161): This trial has compared the effectiveness of weekly-once 2.4 mg s.c. semaglutide with daily-once 3.0 mg s.c. liraglutide and placebo on 338 obese subjects (48). Semaglutide was found to be more potent and has achieved a weight loss of 15.8% over 6.4% with liraglutide (49).
Participants with a history of type 1 or type 2 diabetes were excluded from this study. Difference in initial doses of semaglutide and liraglutide may be the reason for high discontinuation rate and relatively lower weight loss achieved by liraglutide. Further biasness was incorporated as the missing data were handled by imputation; however, number of such cases were low (49).
STEP-TEENS (NCT04102189): This on-going trial would explore the safety, efficacy of semaglutide (weekly-once 2.4 mg s.c.) on 163 obese adolescent subjects with or without type 2 diabetes (12–18 years). The results have not been published (50).
Participants with obesity due to secondary reasons (hypothalamic, endocrinal) and those with Type 2 diabetic patients along with unstable diabetic retinopathy were excluded from this study. Apart from this, small sample population is also a limitation of this study (50).
SELECT (NCT03574597): This event-driven, on-going trial is aimed at investigating the efficacy of semaglutide in reducing CV risks (CV death, non-fatal stroke or myocardial infarction) on 17,500 obese subjects (age ≥ 45 years) with CV history, who receive weekly-once 2.4 mg s.c. semaglutide (initially 0.24 mg, then 0.5, 1.0, 1.7, finally 2.4 mg) or placebo (51, 52).
Participants with myocardial infarction, stroke within 60 days prior to the screening, and those with type 1 or 2 diabetes were excluded from this study. Irrespective of the positive outcomes, it couldn’t provide insight for persons suffering from obesity, CVD, and type 2 diabetes (52).
Semalgutide as An Anti-Obesity Drug
Remarkable weight loss with good safety profile in clinical studies has paved the way for developing semaglutide into a promising anti-obesity drug. Encouraging results from phase-3 STEP trials led to the submission of new drug application (NDA) with priority review voucher for weekly-once s.c. 2.4 mg semaglutide as anti-obesity drug to the USFDA by Novo Nordisk on 4th December, 2020 (53). A similar Marketing Authorization Application (MAA) has been filed with European Medicines Agency (EMA) on 18th December 2020 for introducing weekly-once s.c. 2.4 mg semaglutide for management of obesity (54). Although oral semaglutide didn’t show superior efficacy over placebo in the CV outcome trial (PIONEER-6), Novo Nordisk has announced to initiate phase-3a studies on oral semaglutide for treatment of obesity (55). On 4th June, 2021 USFDA has approved weekly-once s.c. 2.4 mg semaglutide (under brand name Wegovy™) for management of obesity (56). EMA has also approved the use of s.c. semaglutide on 11th November, 2021 for management of chronic obesity in patients with at least one weight-related comorbidity like type 2 diabetes, hypertension or CV diseases (57). Thus semaglutide has become the first weekly-once GLP-1 receptor agonist to be used for chronic weight management.
NON-ALCOHOLIC STEATOHEPATITIS (NASH)
Accumulation of excess fat in hepatocytes in absence or little consumption of alcohol is commonly known as non-alcoholic fatty liver disease (NAFLD), which may progress to liver cirrhosis or even to hepatocellular carcinoma. Based on pathological findings, it can be differentiated into two types- simple steatosis or non-alcoholic fatty liver (NAFL) with ballooned hepatocytes or non-alcoholic steatohepatitis (NASH) without ballooned hepatocytes (58, 59). Augmentation of de novo lipogenesis in hyperglycaemia provides a direct link between NAFLD and insulin resistance (60). Insulin resistance accompanied with obesity causes increased lipolysis to release free fatty acids from adipose tissue to the liver. Excess food intake causes increased free fatty acid (FFA) load to the liver which increase their oxidation and esterification to triglycerides (61). Impaired free fatty acid metabolism along with increased secretion of VLDL causes accumulation of triglycerides in the form of lipid drops inside hepatocytes, known as steatosis. Disrupted insulin signalling, lipid peroxidation, activation of inflammatory pathways predisposes to liver injury and inflammation that leads to NAFLD or NASH (62). Various metabolic syndromes like diabetes, obesity, hypertension, atherosclerosis, and dyslipidaemia are commonly associated with NAFLD (63). Even, a direct link has been found to exist between type 2 diabetes and NAFLD (64). Unfortunately, no treatment approach apart from the diet and life style management has been approved till date for the management of NASH/NAFLD.
Although presence of GLP-1 receptor in hepatocytes is still controversial (65, 66), GLP-1 has shown indirect protective action on liver through the gut-pancreas-liver axis. It stimulates hepatic lipogenesis, glucose uptake, reduces hepatic gluconeogenesis and improves insulin resistance (4). GLP-1 receptor agonists have shown hepato-protection by improving hepatic mitochondrial function, insulin sensitivity and by inhibiting the stress response of injured endoplasmic reticulum. They also promotes autophagy to reduce accumulation of free fatty acids and reduces lipotoxicity (67, 68). Reports suggest that patients with NASH have impaired GLP-1 secretion (69); it strengthens the proposition of GLP-1 receptor agonists as a therapeutic option for management of NASH. Various GLP-1 receptor agonists like liraglutide, dulaglutide, semaglutide have shown to improve the pathogenesis of NASH (70). Tirzepatide, a dual agonist of GLP-1 and GIP has been found to improve various NASH-biomarkers in patients with type 2 diabetes (71). Semaglutide, apart from glycaemic control, has also been reported to decrease hepatic steatosis via stimulation of GLP-1 receptors and by reducing inflammation (65, 72, 73). Therefore, many trials have been investigating the effectiveness of semaglutide in the management of NASH/NAFLD (9).
Clinical Trials to Study the Efficacy of Semaglutide in Treatment of NASH
A phase-2 trial (NCT02970942) has been conducted on 320 patients with NASH and fibrosis to explore the efficacy and safety of daily-once s.c. semaglutide. The subjects were randomly assigned (3:3:3:1:1:1) to be given 0.1 mg (n = 80) or 0.2 mg (n = 78) or 0.4 mg (n = 82) semaglutide or daily-once s.c. placebo (n = 80) for 72 weeks. All the three doses (0.1, 02, 0.4 mg) of semaglutide improved NASH without worsening fibrosis in 40%, 36%, and 59% patients respectively as compared to 17% in placebo. However, no significant difference between various treatment groups was observed in improving fibrosis. Semaglutide was well tolerated in all the three doses but without any significant improvement in fibrosis (74).
A proof of concept, phase-2 trial (NCT03987074) explored the effectiveness of semaglutide, when taken alone or with firsocostat and cilofexor for treatment of NASH. This study involved 108 patients with NASH to take either weekly-once s.c. semaglutide (0.24 mg initial dose escalated to 2.4 mg), or semaglutide + daily-once oral 20 mg firsocostat, or semaglutide + daily-once oral 30 mg cilofexor, or semaglutide + daily-once oral 100 mg cilofexor, or semaglutide + daily-once oral 20 mg firsocostat + daily-once oral 30 mg cilofexor for 24 weeks. Significant improvement in NASH and fibrosis was observed with semaglutide monotherapy, but the combination regimen improved further reduction in hepatic steatosis. Semaglutide along with the combination regimen was well tolerated, but highest impact was observed in NAFLD activity score due to the combination regimen (75, 76).
Another placebo-controlled, phase-2 trial (NCT03987451) is underway investigating the effectiveness of weekly-once s.c. semaglutide in NAFLD and cirrhosis patients. This study involves 65 subjects, who received semaglutide (0.25 mg escalated to 2.4 mg) or weekly-once s.c. placebo for 48 weeks. Magnetic resonance imaging (MRI) scans of liver would measure improvement in liver cirrhosis, fibrosis, changes in liver stiffness, liver fat content, NAFLD activity score (77).
A phase-3 trial (NCT04822181) is underway which aims to study the effect of weekly-once s.c. semaglutide monotherapy in NASH patients with no cirrhosis. Progression in cirrhosis, resolution of steatohepatitis, inflammation, histology of ballooned hepatocytes are some of the important end points which would be assessed during this 5 year long study (78).
As there is no clinically approved treatment available, except for dietary management along with exercise; semaglutide monotherapy or in a combination regimen is a ray of hope for successful treatment of NASH. Successful phase-2 trial results encouraged researchers for further clinical evaluation of semaglutide to develop it into a first drug candidate against NAFLD/NASH.
NEURODEGENERATIVE DISEASES
Neurodegenerative diseases like Parkinson’s and Alzheimer’s diseases are the common form of progressive neurodegeneration which mentally incapacitates a person. Recently, the role of insulin resistance in brain has been deciphered which elucidates a direct link with the pathology of Alzheimer’s and Parkinson’s disease (79, 80). GLP-1 plays important role in augmenting insulin signalling inside the brain. GLP-1 receptors, present inside the brain, are also involved in cognition, synaptic transmission in hippocampal neurons, and cell apoptosis. Overexpression of this receptor is responsible for cognition enhancement and neuroprotection, while deficiency increases the chances of seizure and neurodegeneration (81, 82). Therefore, GLP-1 receptors are considered as validated target for exploring candidates with better neuroprotection and cognition enhancing abilities (83). After discovering neuroprotective effects of exenatide, gradually other GLP-1 receptor agonists got the attention for a possible new therapeutic application in the arena of neurodegenerative disorders. Based on drug repurposing strategy, researchers focused on FDA approved GLP-1 receptor agonists with known safety profiles for further exploring their neuroprotective efficacy (84). GLP-1 receptor agonists have been reported to regulate dopamine levels in Parkinson’s disease and also aggregation of amyloid β peptides in Alzheimer’s disease (7).
Although GLP-1 receptors are expressed in discrete regions of brain like- hypothalamus, brain stem, septal nucleus, and many other neurons (9), till date no data confirms permeation of semaglutide across the Blood Brain Barrier (BBB) in humans. However, certain report suggests that the non-protien form of semaglutide may get access to the GLP-1 receptors in brain through leaks in BBB to carry out it’s centrally mediated action (35). Another possibility is that the acylated form of liraglutide and semalgutide helps it in facilitating their entry into some additional regions of the brain (85). Gabery et al. have reported that semaglutide doesn’t interact with endothelial cells of BBB (86). Salameh et al. have also reported that semaglutide do not cross BBB while studying pharmacokinetics of 125I-labeled exendin-4, liraglutide, lixisenatide, semaglutide in adult mice (82).
Alzheimer’s Disease (AD)
AD is characterized by impaired memory and cognition affecting speech, behaviour, visuo-spatial orientation and motor system. AD, the most common reason of dementia, is caused by progressive neurodegeneration, often associated with cerebrovascular lesions in brain. The pathogenesis includes cortical atrophy, enlarged frontal and temporal horns in the lateral ventricles of brain, and decreased brain weight. However, presence of extracellular amyloid plaques, intracellular neurofibrillary tangles, cerebral amyloid angiopathy, Hirano bodies, tau-positive neurophil threads, activated microglia and reactive astrocytes are commonly found in AD (87).
Clinical Trials for Alzheimer’s Disease
Animal Studies
Zhang et al. have demonstrated that daily-once semaglutide (25 nmol/kg) improved the motor impairment, reduced the oxidative injury, inflammation and apoptosis in methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mice model (88). A similar study carried out by the same research group has shown that semaglutide protects dopaminergic neurons from injury by reducing the aggregation of α-synuclein and by enhanced expression of “glial cell line-derived neurotrophic factor” (GDNP), a growth factor. It also augmented the dopamine levels by supplementing tyrosine hydroxylase levels in brain (89). Yang et al. have demonstrated the neuroprotective efficacy of semaglutide in the rat model of stroke through decreased inflammation, apoptosis and stabilizing the neurogenesis process (90).
Human Studies
Chang et al. have demonstrated the neuroprotective property of semaglutide in human neuroblastoma (SH-SY5Y) cell line against amyloid-β plaques (hallmark of Alzheimer’s disease), possibly mediated by enhanced autophagy and inhibition of apoptosis (91). Various pre-clinical and phase-2 study results supported 14 mg oral semaglutide to proceed for phase-3 clinical trials on Alzheimer’s patients (92).
Two placebo-controlled phase-3 trials (EVOKE NCT04777396 and EVOKE Plus NCT04777409) have been initiated to study the efficacy of oral semaglutide (14 mg) in patients with early Alzheimer’s disease. Change in clinical dementia rating, time taken to reach dementia, change in the Alzheimer's Disease Composite Score are some of the important outcomes that would be assessed from these trials (93, 94).
Parkinson’s Disease (PD)
PD is caused by degradation of dopaminergic neurons in substantia nigra followed by loss of axons of these neurons projecting towards nigrostriatal pathway. It is characterized by various motor and non-motor symptoms which includes bradykinesia, tremors, rigidity, ataxia, shuffling gait, and sleep disorder, psychosis. Accumulation of α -synuclein protein in the form of Lewy bodies and activation of microglia, astrocytes are some of the important pathological features in PD. Mutations in several genes like PARK7, PINK1, SNCA, and LRRK2 are responsible for development of various familial and idiopathic forms of PD (95).
Clinical Trials for Parkinson’s Disease
A phase-2 clinical study for exploring the effectiveness of semaglutide in Parkinson’s disease has been designed and registered by the Oslo University hospital since 2018, but it has not yet started (NCT03659682) (96). This placebo-controlled trial aims to study the neuroprotective and anti-inflammatory effect of semaglutide on both motor and non-motor symptoms, nigrostriatal degeneration, cognitive function, and on quality of life. In addition, this study will confirm the penetration of semaglutide into BBB.
Safety & Tolerability
Adverse Effects
Semaglutide shows mild to moderate gastrointestinal side effects like nausea, vomiting, diarrhoea which is generally dose-dependent and subsides within 2 weeks (97). A few other adverse events like headache, nasopharyngitis, urinary tract and upper respiratory tract infections, increased level of pancreatic enzymes, increased heart and pulse rate, cholecystitis, cholelithiasis have also been reported but were infrequent and not of much concern (31, 39, 98–101). Semaglutide needs dose reduction when it is co-administered with insulin analogues or other hypoglycaemic drugs. SUSTAIN-6 trial has reported 76% higher risk of retinopathy related complications in semaglutide treatment population (102).
Precautions
Semaglutide can’t be used in children, adolescents and for treatment of type 1 diabetes and ketoacidosis (103, 104). Although no evidence of pancreatitis has been found with semaglutide treatment (105), still it is advised to discontinue semaglutide in patients with acute pancreatitis. It should be avoided in patients with medullary thyroid carcinoma and MEN 2 (multiple endocrine neoplasia syndrome type 2) (103, 104). European Medicines Agency advises to avoid semaglutide in patients with congestive heart failure (106, 107).
Cardiovascular Safety
Both SUSTAIN-6 and PIONEER-6 study have reported that semaglutide therapy is associated with 24% reduction in various cardiovascular adverse effects (108).
Patient Adherence
Semaglutide can be safely used without any dose reduction in adults, geriatric patients, or in those suffering from renal and hepatic impairment. Semaglutide has not been found to interfere with the bioavailability of digoxin, metformin, lisinopril, oral contraceptives, warfarin, furosemide, atorvastatin, rosuvastatin, and omeprazole (109–113). Therefore, semaglutide is considered as well tolerated among the patients.
DISCUSSION
Drug repurposing strategy is an effective approach for searching different therapeutic opportunities of already established drug molecules. It helps in reducing the cost as well as time needed for successfully launching a drug with known safety and pharmacokinetics profile into the market. GLP-1 receptor agonists improve insulin resistance in liver as well as brain, regulate energy intake in obesity, provide hepato-protective and neuro-protective effect. Reports suggest that various metabolic disorders, NASH are linked to carbohydrate and fat metabolism, insulin resistance, obesity and CV factors. Even neurodegeneration and its progress are linked with insulin resistance and defective insulin signalling in brain. As semaglutide is a clinically approved drug for treatment of type 2 diabetes, obesity and has been proved to reduce CV risk factors, scientists took keen interest in repurposing it for therapeutic use in patients with NASH and neurodegenerative disorders. Semaglutide can be truly considered as a quintessential of GLP-1 receptor agonist which has the potential to act as a drug with versatile therapeutic utilities.
During phase-3 SUSTAIN trials s.c. semaglutide was found to decrease the body weight by reducing energy and calorie intake. The clinical efficacy of semaglutide in obese individuals without type 2 diabetes was studied in STEP-1, STEP-3, STEP-4, STEP-5, and STEP-8 trials; whereas those with type 2 diabetes or with HbA1c greater than 6.5% were included in the STEP-2, STEP-6, STEP-7 and STEP-TEENS. In addition, two of the STEP trials have included individuals from a specific geographical region such as East Asian in STEP-6 and Chinese subjects in STEP-7 trials. STEP-TEENS restricted the study to obese adolescent participants with or without type 2 diabetes. Most of the STEP trials were carried out for a duration of 68 weeks, whereas STEP-5 trial was conducted for 108 weeks investigating the long-term efficacy of semaglutide. Although many of these trials were conducted on obese participants with or without type 2 diabetes, none of them included obese patients with renal impairment or diabetic retinopathy. The primary outcome of all the STEP trials is to determine the mean % reduction in body weight. In addition, various secondary outcomes like change in waist circumference, body mass index, HbA1c, Blood pressure, cholesterol, lipids, triglycerides, free fatty acids, pulse rate, amylase and lipase levels are also studied in these trials. Semaglutide (s.c.) has shown greater efficacy in reducing body weight as compared to placebo and liraglutide in STEP trials. The CV risk attenuating ability and anti-obesity efficacy of semaglutide is being studied in SELECT and STEP-TEENS trial respectively. Encouraging results obtained from the phase-3 STEP trials established the use of weekly-once 2.4 mg semaglutide (s.c.) for management of chronic obesity, as approved by USFDA as well as EMA. Oral semaglutide have also shown weight reducing property in PIONEER trials, although the efficacy was not superior to that of the placebo, still it is expected that very soon trials would be started regarding anti-obesity effect of oral semaglutide to increase the patient convenience and adherence. Hence, semaglutide can provide dual benefit to patients with type 2 diabetes and obesity (diabesity).
Semaglutide has also shown hepato-protective property and is currently being accessed in phase-3 trial for exploring it’s efficacy both in mono- as well as in combination therapy with other drugs in management and prevention of NASH. GLP-1 receptors have been expressed in various regions of the brain and has been linked with brain insulin sensitivity, neuroprotection and cognition abilities. After the neuroprotective effects of exenatide was discovered, gradually other GLP-1 receptor agonists got the attention for a possible new therapeutic application in the arena of neurodegenerative disorders. Based on Drug repurposing strategy, researchers focused on FDA approved GLP-1 receptor agonists with known safety profiles for further exploring their neuroprotective efficacy (73).
An ongoing phase-2 SEMPATICO trial will further explore the efficacy of semaglutide in alleviating myocardial injury in COVID-19 patients with diabetes, obesity, hypertension and CV diseases. This study will also reveal the ability of semaglutide to reduce morbidity in COVID-19 patients (114).
CONCLUSION
Semaglutide is a clinically approved drug for type 2 diabetes, obesity, and a clinical trial candidate for management of NASH, Parkinson’s and Alzheimer’s disease. Semaglutide is well tolerated and can be safely used in patients with renal, hepatic and CV diseases. Hepatoprotective and neuroprotective role of semaglutide in abating NASH and neurodegenerative diseases respectively has been proved in phase-2 trials and still under study. It improves insulin resistance in periphery as well as in brain, reduce body weight, reduce fat content in liver by acting as agonist on GLP-1 receptor. The efficacy of semaglutide in alleviating cardiac injury in COVID-19 patients with diabetes, obesity, hypertension and CV diseases is being studied. Based on drug repurposing strategy, researchers are trying their best to explore various therapeutic potential of semaglutide. Clinical efficacy in different therapeutic fields proved semaglutide to be an indispensable treatment option for better management of “diabesity” and an emerging cynosure for management of NASH and neurodegenerative diseases.
Author Contributions
Conceptualization and Literature search: Manoj K. Mahapatra; Data analysis: Manoj K. Mahapatra, Muthukumar Karuppasamy, Biswa M. Sahoo; Manuscript draft: Manoj K. Mahapatra; Critical revision: Muthukumar Karuppasamy, Biswa M. Sahoo. All authors have read and approved the final version of this manuscript.
Conflicts of Interest
The authors certify that there is no conflict of interest with any financial organization regarding the material discussed in the manuscript.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- 1.Zummo FP, Cullen KS, Honkanen-Scott M, Shaw JA, Lovat PE, Arden C. Glucagon-like peptide 1 protects pancreatic b-cells from death by increasing autophagic flux and restoring lysosomal function. Diabetes. 2017;66:1272–1285. doi: 10.2337/db16-1009. [DOI] [PubMed] [Google Scholar]
- 2.Cornu M, Yang J-Y, Jaccard E, Poussin C, Widmann C, Thorens B. Glucagon like peptide-1 protects b-cells against apoptosis by increasing the activity of an IGF-2/IGF-1 receptor autocrine loop. Diabetes. 2009;58:1816–1825. doi: 10.2337/db09-0063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Khound R, Taher J, Baker C, Adeli K, Su Q. GLP-1 Elicits an Intrinsic Gut-Liver Metabolic Signal to Ameliorate Diet-Induced VLDL Overproduction and Insulin Resistance. Arterioscler Thromb Vasc Biol. 2017;37(12):2252–2259. doi: 10.1161/ATVBAHA.117.310251. [DOI] [PubMed] [Google Scholar]
- 4.Jin T, Weng J. Hepatic functions of GLP-1 and its based drugs: current disputes and perspectives. Am J Physiol Endocrinol Metab. 2016;311(3):E620–E627. doi: 10.1152/ajpendo.00069.2016. [DOI] [PubMed] [Google Scholar]
- 5.Turton M, Shea D, Gunn I, Beak S. A role for glucagon-like peptide-1 in the central regulation of feeding. Nature. 1996;379:69–72. doi: 10.1038/379069a0. [DOI] [PubMed] [Google Scholar]
- 6.Katsurada K, Yada T. Neural effects of gut- and brain-derived glucagon-like peptide-1 and its receptor agonist. J Diabetes Investig. 2016;7(Suppl. 1):64–69. doi: 10.1111/jdi.12464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Grieco M, Giorgi A, Gentile MC, d’Erme M, Morano S, Maras B, et al. Glucagon-Like Peptide-1: A Focus on Neurodegenerative Diseases Front Neurosci 2019;13:1112. 10.3389/fnins.2019.01112 [DOI] [PMC free article] [PubMed]
- 8.Rosenstock J, Nino A, Soffer J, Erskine L, Acusta A, Dole J, et al. Impact of a Weekly Glucagon-Like Peptide 1 Receptor Agonist, Albiglutide, on Glycemic Control and on Reducing Prandial Insulin Use in Type 2 Diabetes Inadequately Controlled on Multiple Insulin Therapy: A Randomized Trial. Diabetes Care. 2020;43(10):2509–2518. doi: 10.2337/dc19-2316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Knudsen LB, Lau J. The Discovery and Development of Liraglutide and Semaglutide. Front Endocrinol (Lausanne) 2019;10:155. doi: 10.3389/fendo.2019.00155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Mahapatra MK, Karuppasamy M, Sahoo BM. Semaglutide, a glucagon like peptide-1 receptor agonist with cardiovascular benefits for management of type 2 diabetes. Rev Endocr Metab Disord 2022:1–19. 10.1007/s11154-021-09699-1 [DOI] [PMC free article] [PubMed]
- 11.Omer T. The causes of obesity: an in-depth review. Adv Obes Weight Manag Control. 2020;10(4):90–94. doi: 10.15406/aowmc.2020.10.00312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Grundy SM. Metabolic complications of obesity. Endocrine. 2000;13(2):155–165. doi: 10.1385/ENDO:13:2:155. [DOI] [PubMed] [Google Scholar]
- 13.Maurizi G, Della Guardia L, Maurizi A, Poloni A. Adipocytes properties and crosstalk with immune system in obesity-related inflammation. J Cell Physiol. 2018;233(1):88–97. doi: 10.1002/jcp.25855. [DOI] [PubMed] [Google Scholar]
- 14.Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444(7121):860–867. doi: 10.1038/nature05485. [DOI] [PubMed] [Google Scholar]
- 15.Herman MA, Peroni OD, Villoria J, Schön MR, Abumrad NA, Blüher M, Klein S, Kahn BB. A novel ChREBP isoform in adipose tissue regulates systemic glucose metabolism. Nature. 2012;484(7394):333–338. doi: 10.1038/nature10986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lutter M, Nestler EJ. Homeostatic and hedonic signals interact in the regulation of food intake. J Nutr. 2009;139(3):629–632. doi: 10.3945/jn.108.097618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ferrario CR, Labouèbe G, Liu S, Nieh EH, Routh VH, Xu S, O'Connor EC. Homeostasis Meets Motivation in the Battle to Control Food Intake. J Neurosci. 2016;36(45):11469–11481. doi: 10.1523/JNEUROSCI.2338-16.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Welle S, Campbell RG. Stimulation of thermogenesis by carbohydrate overfeeding. Evidence against sympathetic nervous system mediation. J Clin Invest 1983;71(4):916–25. 10.1172/jci110846. [DOI] [PMC free article] [PubMed]
- 19.Young JB, Landsberg L. Suppression of sympathetic nervous system during fasting. Obes Res. 1997;5(6):646–649. doi: 10.1002/j.1550-8528.1997.tb00590.x. [DOI] [PubMed] [Google Scholar]
- 20.Hesse S, van de Giessen E, Zientek F, Petroff D, Winter K, Dickson JC, et al. Association of central serotonin transporter availability and body mass index in healthy Europeans. Eur Neuropsychopharmacol. 2014;24(8):1240–1247. doi: 10.1016/j.euroneuro.2014.05.005. [DOI] [PubMed] [Google Scholar]
- 21.Blum K, Thanos PK, Gold MS. Dopamine and glucose, obesity, and reward deficiency syndrome. Front Psychol. 2014;5:919. doi: 10.3389/fpsyg.2014.00919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.English PJ, Ghatei MA, Malik IA, Bloom SR, Wilding JP. Food fails to suppress ghrelin levels in obese humans. J Clin Endocrinol Metab. 2002;87(6):2984. doi: 10.1210/jcem.87.6.8738. [DOI] [PubMed] [Google Scholar]
- 23.El-Haschimi K, Pierroz DD, Hileman SM, Bjørbaek C, Flier JS. Two defects contribute to hypothalamic leptin resistance in mice with diet-induced obesity. J Clin Invest. 2000;105(12):1827–1832. doi: 10.1172/JCI9842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kim B, Feldman EL. Insulin resistance in the nervous system. Trends Endocrinol Metab. 2012;23(3):133–141. doi: 10.1016/j.tem.2011.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Arble DM, Bass J, Laposky AD, Vitaterna MH, Turek FW. Circadian timing of food intake contributes to weight gain. Obesity (Silver Spring) 2009;17(11):2100–2102. doi: 10.1038/oby.2009.264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Karlsson B, Knutsson A, Lindahl B. Is there an association between shift work and having a metabolic syndrome? Results from a population based study of 27,485 people. Occup Environ Med. 2001;58(11):747–752. doi: 10.1136/oem.58.11.747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Oussaada SM, van Galen KA, Cooiman MI, Kleinendorst L, Hazebroek EJ, van Haelst MM, et al. The pathogenesis of obesity. Metabolism. 2019;92:26–36. doi: 10.1016/j.metabol.2018.12.012. [DOI] [PubMed] [Google Scholar]
- 28.Perez-Montes DE, Oca A, Pellitero S, Puig-Domingo M. Obesity and GLP-1. Minerva Endocrinol (Torino) 2021;46(2):168–176. doi: 10.23736/S2724-6507.20.03369-6. [DOI] [PubMed] [Google Scholar]
- 29.Barrea L, Pugliese G, Muscogiuri G, Laudisio D, Colao A, Savastano S. New-generation anti-obesity drugs: naltrexone/bupropion and liraglutide An update for endocrinologists and nutritionists. Minerva Endocrinol 2020;45(2):127–137. 10.23736/S0391-1977.20.03179-X. [DOI] [PubMed]
- 30.Ryan DH. Next Generation Antiobesity Medications: Setmelanotide, Semaglutide, Tirzepatide and Bimagrumab: What do They Mean for Clinical Practice? J Obes Metab Syndr. 2021;30(3):196–208. doi: 10.7570/jomes21033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Christou GA, Katsiki N, Blundell J, Fruhbeck G, Kiortsis DN. Semaglutide as a promising antiobesity drug. Obesity Rev. 2019;20(6):805–815. doi: 10.1111/obr.12839. [DOI] [PubMed] [Google Scholar]
- 32.Blundell J, Finlayson G, Axelsen M, Flint A, Gibbons C, Kvist T, et al. Effects of once-weekly semaglutide on appetite, energy intake, control of eating, food preference and body weight in subjects with obesity. Diabetes Obes Metab. 2017;19(9):1242–1251. doi: 10.1111/dom.12932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.van Bloemendaal L, Ten Kulve JS, la Fleur SE, Ijzerman RG, Diamant M. Effects of glucagon-like peptide 1 on appetite and body weight: focus on the CNS. J Endocrinol. 2014;221(1):T1–16. doi: 10.1530/JOE-13-0414. [DOI] [PubMed] [Google Scholar]
- 34.Gaykema RP, Newmyer BA, Ottolini M, Raje V, Warthen DM, Lambeth PS, et al. Activation of murine pre-proglucagon-producing neurons reduces food intake and body weight. J Clin Invest. 2017;127(3):1031–1045. doi: 10.1172/JCI81335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Secher A, Jelsing J, Baquero AF, Hecksher-Sørensen J, Cowley MA, Dalbøge LS, et al. The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss. J Clin Invest. 2014;124(10):4473–4488. doi: 10.1172/JCI75276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Krieger JP, Arnold M, Pettersen KG, Lossel P, Langhans W, Lee SJ. Knockdown of GLP-1 receptors in vagal afferents affects normal food intake and glycemia. Diabetes. 2016;65(1):34–43. doi: 10.2337/db15-0973. [DOI] [PubMed] [Google Scholar]
- 37.Friedrichsen M, Breitschaft A, Tadayon S, Wizert A, Skovgaard D. The effect of semaglutide 2.4 mg once weekly on energy intake, appetite, control of eating, and gastric emptying in adults with obesity. Diabetes Obes Metab. 2021;23(3):754–62. doi: 10.1111/dom.14280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Enebo LB, Berthelsen KK, Kankam M, Lund MT, Rubino DM, Satylganova A, et al. Safety, tolerability, pharmacokinetics, and pharmacodynamics of concomitant administration of multiple doses of cagrilintide with semaglutide 2·4 mg for weight management: a randomised, controlled, phase 1b trial. Lancet. 2021;397(10286):1736–1748. doi: 10.1016/S0140-6736(21)00845-X. [DOI] [PubMed] [Google Scholar]
- 39.O'Neil PM, Birkenfeld AL, McGowan B, Mosenzon O, Pedersen SD, Wharton S, et al. Efficacy and safety of semaglutide compared with liraglutide and placebo for weight loss in patients with obesity: a randomised, double-blind, placebo and active controlled, dose-ranging, phase 2 trial. Lancet. 2018;392(10148):637–649. doi: 10.1016/S0140-6736(18)31773-2. [DOI] [PubMed] [Google Scholar]
- 40.Kushner RF, Calanna S, Davies M, Dicker D, Garvey WT, Goldman B, et al. Semaglutide 2.4 mg for the Treatment of Obesity: Key Elements of the STEP Trials 1 to 5. Obesity (Silver Spring) 2020;28(6):1050–61. 10.1002/oby.22794 [DOI] [PMC free article] [PubMed]
- 41.Wilding JPH, Batterham RL, Calanna S, Davies M, Van Gaal LF, Lingvay I, et al. STEP 1 Study Group. Once-Weekly Semaglutide in Adults with Overweight or Obesity. N Engl J Med 2021;384(11):989. 10.1056/NEJMoa2032183. [DOI] [PubMed]
- 42.Davies M, Færch L, Jeppesen OK, Pakseresht A, Pedersen SD, Perreault L, et al. STEP 2 Study Group. Semaglutide 2·4 mg once a week in adults with overweight or obesity, and type 2 diabetes (STEP 2): a randomised, double-blind, double-dummy, placebo-controlled, phase 3 trial. Lancet 2021;397(10278):971–84. 10.1016/S0140-6736(21)00213-0. [DOI] [PubMed]
- 43.Wadden TA, Bailey TS, Billings LK, Davies M, Frias JP, Koroleva A, et al. STEP 3 Investigators. Effect of Subcutaneous Semaglutide vs Placebo as an Adjunct to Intensive Behavioral Therapy on Body Weight in Adults With Overweight or Obesity: The STEP 3 Randomized Clinical Trial. JAMA 2021;325(14):1403–13. 10.1001/jama.2021.1831. [DOI] [PMC free article] [PubMed]
- 44.Rubino D, Abrahamsson N, Davies M, Hesse D, Greenway FL, Jensen C, et al. STEP 4 Investigators. Effect of Continued Weekly Subcutaneous Semaglutide vs Placebo on Weight Loss Maintenance in Adults With Overweight or Obesity: The STEP 4 Randomized Clinical Trial. JAMA 2021;325(14):1414–25. 10.1001/jama.2021.3224. [DOI] [PMC free article] [PubMed]
- 45.Two-year Research Study Investigating How Well Semaglutide Works in People Suffering From Overweight or Obesity (STEP 5). [cited 2021 Feb 05]. Available from: https://clinicaltrials.gov/ct2/show/NCT03693430
- 46.Kadowaki T, Isendahl J, Khalid U, Lee SY, Nishida T, Ogawa W, et al. Semaglutide once a week in adults with overweight or obesity, with or without type 2 diabetes in an east Asian population (STEP 6): a randomised, double-blind, double-dummy, placebo-controlled, phase 3a trial. Lancet Diabetes Endocrinol. 2022;10(3):193–206. doi: 10.1016/S2213-8587(22)00008-0. [DOI] [PubMed] [Google Scholar]
- 47.Research Study of How Well Semaglutide Works in People Living With Overweight or Obesity (STEP 7). [cited 2021 Feb 07]. Available from: https://clinicaltrials.gov/ct2/show/NCT04251156.
- 48.Research Study to Investigate How Well Semaglutide Works Compared to Liraglutide in People Living With Overweight or Obesity (STEP 8). [cited 2021 Feb 07]. Available from: https://clinicaltrials.gov/ct2/show/NCT04074161.
- 49.Rubino DM, Greenway FL, Khalid U, O'Neil PM, Rosenstock J, Sørrig R, et al. Effect of Weekly Subcutaneous Semaglutide vs Daily Liraglutide on Body Weight in Adults With Overweight or Obesity Without Diabetes: The STEP 8 Randomized Clinical Trial. JAMA. 2022;327(2):138–150. doi: 10.1001/jama.2021.23619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.STEP TEENS: A Research Study on How Well Semaglutide Works in Adolescents With Overweight or Obesity. [cited 2021 Feb 10]. Available from: https://clinicaltrials.gov/ct2/show/NCT04102189.
- 51.Semaglutide Effects on Heart Disease and Stroke in Patients With Overweight or Obesity (SELECT). [cited 2021 Feb 26]. Available from: https://clinicaltrials.gov/ct2/show/NCT03574597.
- 52.Ryan DH, Lingvay I, Colhoun HM, Deanfield J, Emerson SS, Kahn SE, et al. Semaglutide Effects on Cardiovascular Outcomes in People With Overweight or Obesity (SELECT) rationale and design. Am Heart J. 2020;229:61–69. doi: 10.1016/j.ahj.2020.07.008. [DOI] [PubMed] [Google Scholar]
- 53.Novo Nordisk: Company announcements. Novo Nordisk files for US FDA regulatory approval of once-weekly semaglutide 2.4 mg for weight management. [cited 2021 Jan 15]. Available from: https://www.novonordisk.com/news-and-media/news-and-ir-materials/news-details.html?id=37551
- 54.Novo Nordisk: Company announcements. Novo Nordisk files for EU regulatory approval of once-weekly semaglutide 2.4 mg for weight management. [cited 2021 Jan 28]. Available from: https://ml-eu.globenewswire.com/Resource/Download/736c741c-98e9-4518-a98b-15975f68d5e1
- 55.Novo Nordisk Company Announcements. Novo Nordisk to initiate phase 3a development in obesity with oral semaglutide. [cited 2021 Apr 21]. Available from: https://ml-eu.globenewswire.com/Resource/Download/3e7d1e68-06dd-4417-afc8-8f5ac1ee8367
- 56.Novo Nordisk: Company announcements. Wegovy™ (semaglutide 2.4 mg), the first and only once-weekly GLP-1 therapy for weight management, approved in the US. [cited 2021 Jul 08]. Available from: https://ml-eu.globenewswire.com/Resource/Download/77d24f13-7de0-4032-8239-efed71e6e0bf
- 57.Novo Nordisk: Company announcements. WegovyTM recommended for approval for the treatment of obesity by the European regulatory authorities. [cited 2022 January 21]. Available from: https://www.novonordisk.com/news-and-media/news-and-ir-materials/news-details.html?id=86334
- 58.Hashimoto E, Tokushige K, Ludwig J. Diagnosis and classification of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis: Current concepts and remaining challenges. Hepatol Res. 2015;45(1):20–28. doi: 10.1111/hepr.12333. [DOI] [PubMed] [Google Scholar]
- 59.Sakamoto M, Tsujikawa H, Effendi K, Ojima H, Harada K, Zen Y, et al. Pathological findings of nonalcoholic steatohepatitis and nonalcoholic fatty liver disease. Pathol Int. 2017;67(1):1–7. doi: 10.1111/pin.12485. [DOI] [PubMed] [Google Scholar]
- 60.Tanaka N, Aoyama T, Kimura S, Gonzalez FJ. Targeting nuclear receptors for the treatment of fatty liver disease. Pharmacol Ther. 2017;179:142–157. doi: 10.1016/j.pharmthera.2017.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Yki-Järvinen H. Non-alcoholic fatty liver disease as a cause and a consequence of metabolic syndrome. Lancet Diabetes Endocrinol. 2014;2(11):901–910. doi: 10.1016/S2213-8587(14)70032-4. [DOI] [PubMed] [Google Scholar]
- 62.Seghieri M, Christensen AS, Andersen A, Solini A, Knop FK, Vilsbøll T. Future Perspectives on GLP-1 Receptor Agonists and GLP-1/glucagon Receptor Co-agonists in the Treatment of NAFLD. Front Endocrinol (Lausanne) 2018;9:649. doi: 10.3389/fendo.2018.00649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Tanaka N, Kimura T, Fujimori N, Nagaya T, Komatsu M, Tanaka E. Current status, problems, and perspectives of non-alcoholic fatty liver disease research. World J Gastroenterol. 2019;25(2):163–177. doi: 10.3748/wjg.v25.i2.163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Bril F, Cusi K. Nonalcoholic Fatty Liver Disease: The New Complication of Type 2 Diabetes Mellitus. Endocrinol Metab Clin North Am. 2016;45(4):765–781. doi: 10.1016/j.ecl.2016.06.005. [DOI] [PubMed] [Google Scholar]
- 65.Gupta NA, Mells J, Dunham RM, Grakoui A, Handy J, Saxena NK, et al. Glucagon-like peptide-1 receptor is present on human hepatocytes and has a direct role in decreasing hepatic steatosis in vitro by modulating elements of the insulin signaling pathway. Hepatology. 2010;51(5):1584–1592. doi: 10.1002/hep.23569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Panjwani N, Mulvihill EE, Longuet C, Yusta B, Campbell JE, Brown TJ, et al. GLP-1 receptor activation indirectly reduces hepatic lipid accumulation but does not attenuate development of atherosclerosis in diabetic male ApoE(-/-) mice. Endocrinology. 2013;154(1):127–139. doi: 10.1210/en.2012-1937. [DOI] [PubMed] [Google Scholar]
- 67.Kalogirou M, Sinakos E. Treating nonalcoholic steatohepatitis with antidiabetic drugs: Will GLP-1 agonists end the struggle? World J Hepatol. 2018;10(11):790–794. doi: 10.4254/wjh.v10.i11.790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Mantovani A, Targher G. Glucagon-like peptide-1 receptor agonists for treatment of nonalcoholic steatohepatitis: new insights from subcutaneous semaglutide. Hepatobiliary Surg Nutr. 2021;10(4):518–521. doi: 10.21037/hbsn-2021-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Bernsmeier C, Meyer-Gerspach AC, Blaser LS, Jeker L, Steinert RE, Heim MH, et al. Glucose-induced glucagon-like Peptide 1 secretion is deficient in patients with non-alcoholic fatty liver disease. PLoS ONE. 2014;9(1):e87488. doi: 10.1371/journal.pone.0087488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Patel Chavez C, Cusi K, Kadiyala S. The Emerging Role of Glucagon-like Peptide-1 Receptor Agonists for the Management of NAFLD. J Clin Endocrinol Metab. 2022;107(1):29–38. doi: 10.1210/clinem/dgab578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Hartman ML, Sanyal AJ, Loomba R, Wilson JM, Nikooienejad A, Bray R, et al. Effects of Novel Dual GIP and GLP-1 Receptor Agonist Tirzepatide on Biomarkers of Nonalcoholic Steatohepatitis in Patients With Type 2 Diabetes. Diabetes Care. 2020;43(6):1352–1355. doi: 10.2337/dc19-1892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Katsiki N, Athyros VG, Karagiannis A, Mikhailidis DP. Semaglutide, lipid-lowering drugs, and NAFLD. Lancet Diabetes Endocrinol. 2017;5(5):329–330. doi: 10.1016/S2213-8587(17)30109-2. [DOI] [PubMed] [Google Scholar]
- 73.Rakipovski G, Rolin B, Nøhr J, Klewe I, Frederiksen KS, Augustin R, et al. The GLP-1 Analogs Liraglutide and Semaglutide Reduce Atherosclerosis in ApoE-/- and LDLr-/- Mice by a Mechanism That Includes Inflammatory Pathways. JACC Basic Transl Sci. 2018;3(6):844–857. doi: 10.1016/j.jacbts.2018.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Newsome PN, Buchholtz K, Cusi K, Linder M, Okanoue T, Ratziu V, et al. NN9931–4296 Investigators. A Placebo-Controlled Trial of Subcutaneous Semaglutide in Nonalcoholic Steatohepatitis. N Engl J Med 2021;384(12):1113–24. 10.1056/NEJMoa2028395 [DOI] [PubMed]
- 75.Safety, Tolerability, and Efficacy of Monotherapy and Combination Regimens in Adults With Nonalcoholic Steatohepatitis (NASH). [cited 2021 May 25]. Available from: https://clinicaltrials.gov/ct2/show/NCT03987074
- 76.Novo Nordisk Press Release: Gilead and Novo Nordisk present new data from proof-of concept trial in NASH. [cited 2021 May 25]. Available from: https://ml-eu.globenewswire.com/Resource/Download/1fb7fd74-0c03-46ef-9c84-4ae5e5855766
- 77.A Research Study on How Semaglutide Works in People With Fatty Liver Disease and Liver Damage. [cited 2021 Feb 24]. Available from: https://clinicaltrials.gov/ct2/show/NCT03987451
- 78.Research Study on Whether Semaglutide Works in People With Non-alcoholic Steatohepatitis (NASH). [cited 2022 January 15]. Available from: https://clinicaltrials.gov/ct2/show/NCT04822181
- 79.Diehl T, Mullins R, Kapogiannis D. Insulin resistance in Alzheimer's disease. Transl Res. 2017;183:26–40. doi: 10.1016/j.trsl.2016.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Athauda D, Foltynie T. Insulin resistance and Parkinson's disease: A new target for disease modification? Prog Neurobiol. 2016;145–146:98–120. doi: 10.1016/j.pneurobio.2016.10.001. [DOI] [PubMed] [Google Scholar]
- 81.Hölscher C. Novel dual GLP-1/GIP receptor agonists show neuroprotective effects in Alzheimer's and Parkinson's disease models. Neuropharmacology. 2018;136(Pt B):251–259. doi: 10.1016/j.neuropharm.2018.01.040. [DOI] [PubMed] [Google Scholar]
- 82.Salameh TS, Rhea EM, Talbot K, Banks WA. Brain uptake pharmacokinetics of incretin receptor agonists showing promise as Alzheimer's and Parkinson's disease therapeutics. Biochem Pharmacol. 2020;180:114187. doi: 10.1016/j.bcp.2020.114187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Hölscher C. Brain insulin resistance: role in neurodegenerative disease and potential for targeting. Expert Opin Investig Drugs. 2020;29(4):333–348. doi: 10.1080/13543784.2020.1738383. [DOI] [PubMed] [Google Scholar]
- 84.McFarthing K, Larson D, Simuni T. Clinical Trial Highlights - GLP-1 agonists. J Parkinsons Dis. 2020;10(2):355–368. doi: 10.3233/JPD-200002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Holst JJ, Madsbad S. Semaglutide seems to be more effective the other GLP-1Ras. Ann Transl Med. 2017;5(24):505. doi: 10.21037/atm.2017.11.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Gabery S, Salinas CG, Paulsen SJ, Ahnfelt-Rønne J, Alanentalo T, Baquero AF, et al. Semaglutide lowers body weight in rodents via distributed neural pathways. JCI Insight. 2020;5(6):e133429. doi: 10.1172/jci.insight.133429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.DeTure MA, Dickson DW. The neuropathological diagnosis of Alzheimer's disease. Mol Neurodegener. 2019;14(1):32. doi: 10.1186/s13024-019-0333-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Zhang L, Zhang L, Li L, Hölscher C. Neuroprotective effects of the novel GLP-1 long acting analogue semaglutide in the MPTP Parkinson's disease mouse model. Neuropeptides. 2018;71:70–80. doi: 10.1016/j.npep.2018.07.003. [DOI] [PubMed] [Google Scholar]
- 89.Zhang L, Zhang L, Li L, Hölscher C. Semaglutide is Neuroprotective and Reduces α-Synuclein Levels in the Chronic MPTP Mouse Model of Parkinson's Disease. J Parkinsons Dis. 2019;9(1):157–171. doi: 10.3233/JPD-181503. [DOI] [PubMed] [Google Scholar]
- 90.Yang X, Feng P, Zhang X, Li D, Wang R, Ji C, et al. The diabetes drug semaglutide reduces infarct size, inflammation, and apoptosis, and normalizes neurogenesis in a rat model of stroke. Neuropharmacology. 2019;158:107748. doi: 10.1016/j.neuropharm.2019.107748. [DOI] [PubMed] [Google Scholar]
- 91.Chang YF, Zhang D, Hu WM, Liu DX, Li L. Semaglutide-mediated protection against Aβ correlated with enhancement of autophagy and inhibition of apoptosis. J Clin Neurosci. 2020;81:234–239. doi: 10.1016/j.jocn.2020.09.054. [DOI] [PubMed] [Google Scholar]
- 92.Novo Nordisk Company Announcement: Novo Nordisk to enter phase 3 development in Alzheimer’s disease with oral semaglutide. [cited 2021 Feb 15]. Available from: https://ml-eu.globenewswire.com/Resource/Download/e379383d-436d-462d-907b-87fb87afe234
- 93.A Research Study Investigating Semaglutide in People With Early Alzheimer's Disease (EVOKE). [cited 2022 January 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT04777396
- 94.A Research Study Investigating Semaglutide in People With Early Alzheimer's Disease (EVOKE Plus). [cited 20221 January 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT04777409
- 95.MacMahon Copas AN, McComish SF, Fletcher JM, Caldwell MA. The Pathogenesis of Parkinson's Disease: A Complex Interplay Between Astrocytes, Microglia, and T Lymphocytes? Front Neurol. 2021;12:666737. doi: 10.3389/fneur.2021.666737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.GLP1R in Parkinson's Disease (GIPD). [cited 2021 Feb 25]. Available from: https://clinicaltrials.gov/ct2/show/NCT03659682.
- 97.Tan X, Cao X, Zhou M, Zou P, Hu J. Efficacy and safety of once-weekly semaglutide for the treatment of type 2 diabetes. Expert Opin Investig Drugs. 2017;26(9):1083–1089. doi: 10.1080/13543784.2017.1360274. [DOI] [PubMed] [Google Scholar]
- 98.Sorli C, Harashima SI, Tsoukas GM, Unger J, Karsbøl JD, Hansen T, Bain SC. Efficacy and safety of once-weekly semaglutide monotherapy versus placebo in patients with type 2 diabetes (SUSTAIN 1): a double-blind, randomised, placebo-controlled, parallel-group, multinational, multicentre phase 3a trial. Lancet Diabetes Endocrinol. 2017;5(4):251–260. doi: 10.1016/S2213-8587(17)30013-X. [DOI] [PubMed] [Google Scholar]
- 99.Ahrén B, Masmiquel L, Kumar H, Sargin M, Karsbøl JD, Jacobsen SH, Chow F. Efficacy and safety of once-weekly semaglutide versus once-daily sitagliptin as an add-on to metformin, thiazolidinediones, or both, in patients with type 2 diabetes (SUSTAIN 2): a 56-week, double-blind, phase 3a, randomised trial. Lancet Diabetes Endocrinol. 2017;5(5):341–354. doi: 10.1016/S2213-8587(17)30092-X. [DOI] [PubMed] [Google Scholar]
- 100.Aroda VR, Bain SC, Cariou B, Piletič M, Rose L, Axelsen M, Rowe E, DeVries JH. Efficacy and safety of once-weekly semaglutide versus once-daily insulin glargine as add-on to metformin (with or without sulfonylureas) in insulin-naive patients with type 2 diabetes (SUSTAIN 4): a randomised, open-label, parallel-group, multicentre, multinational, phase 3a trial. Lancet Diabetes Endocrinol. 2017;5(5):355–366. doi: 10.1016/S2213-8587(17)30085-2. [DOI] [PubMed] [Google Scholar]
- 101.Seino Y, Terauchi Y, Osonoi T, Yabe D, Abe N, Nishida T, Zacho J, Kaneko S. Safety and efficacy of semaglutide once weekly vs sitagliptin once daily, both as monotherapy in Japanese people with type 2 diabetes. Diabetes Obes Metab. 2018;20(2):378–388. doi: 10.1111/dom.13082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jódar E, Leiter LA, Lingvay I, Rosenstock J, Seufert J, Warren ML, Woo V, Hansen O, Holst AG, Pettersson J, Vilsbøll T; SUSTAIN-6 Investigators. Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med. 2016;375(19):1834–1844. 10.1056/NEJMoa1607141. [DOI] [PubMed]
- 103.Ozempic Product Monograph. Available at: https://www.novonordisk.ca/content/dam/nncorp/ca/en/products/ozempic-product-monograph.pdf Assessed on Oct 5, 2021.
- 104.Rybelsus Product Monograph. Available at: https://www.novonordisk.ca/content/dam/nncorp/ca/en/products/Rybelsus-PM-EN-monograph.pdf Assessed on October 5, 2021.
- 105.Nreu B, Dicembrini I, Tinti F, Mannucci E, Monami M. Pancreatitis and pancreatic cancer in patients with type 2 diabetes treated with glucagon-like peptide-1 receptor agonists: an updated meta-analysis of randomized controlled trials. Minerva Endocrinol. 2020 doi: 10.23736/S0391-1977.20.03219-8. [DOI] [PubMed] [Google Scholar]
- 106.European Medicines Agency. Ozempic; summary of product characteristics. 2020. Available at: http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Product_Information/human/004174/WC500244163.pdf. Assessed on June 18, 2021.
- 107.European Medicines Agency. Rybelsus; summary of product characteristics. 2020. Available at: https://www.ema.europa.eu/en/documents/product-information/rybelsus-epar-product-information_en.pdf. Assessed on June 18, 2021.
- 108.Husain M, Bain SC, Jeppesen OK, Lingvay I, Sørrig R, Treppendahl MB, Vilsbøll T. Semaglutide (SUSTAIN and PIONEER) reduces cardiovascular events in type 2 diabetes across varying cardiovascular risk. Diabetes Obes Metab. 2020;22(3):442–451. doi: 10.1111/dom.13955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Kapitza C, Nosek L, Jensen L, Hartvig H, Jensen CB, Flint A. Semaglutide, a once-weekly human GLP-1 analog, does not reduce the bioavailability of the combined oral contraceptive, ethinylestradiol/levonorgestrel. J Clin Pharmacol. 2015;55(5):497–504. doi: 10.1002/jcph.443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Hausner H, Derving Karsbøl J, Holst AG, Jacobsen JB, Wagner FD, Golor G, Anderson TW. Effect of Semaglutide on the Pharmacokinetics of Metformin, Warfarin, Atorvastatin and Digoxin in Healthy Subjects. Clin Pharmacokinet. 2017;56(11):1391–1401. doi: 10.1007/s40262-017-0532-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Bækdal TA, Borregaard J, Hansen CW, Thomsen M, Anderson TW. Effect of Oral Semaglutide on the Pharmacokinetics of Lisinopril, Warfarin, Digoxin, and Metformin in Healthy Subjects. Clin Pharmacokinet. 2019;58(9):1193–1203. doi: 10.1007/s40262-019-00756-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Jordy AB, Albayaty M, Breitschaft A, Anderson TW, Christiansen E, Houshmand-Øregaard A, Manigandan E, Bækdal TA. Effect of Oral Semaglutide on the Pharmacokinetics of Levonorgestrel and Ethinylestradiol in Healthy Postmenopausal Women and Furosemide and Rosuvastatin in Healthy Subjects. Clin Pharmacokinet. 2021. 10.1007/s40262-020-00976-x [DOI] [PMC free article] [PubMed]
- 113.Bækdal TA, Breitschaft A, Navarria A, Hansen CW. A randomized study investigating the effect of omeprazole on the pharmacokinetics of oral semaglutide. Expert Opin Drug Metab Toxicol. 2018;14(8):869–877. doi: 10.1080/17425255.2018.1488965. [DOI] [PubMed] [Google Scholar]
- 114.A Clinical Trial Comparing Semaglutide in Healthy People Who Eat and Take the Medicine at Different Times. [Cited 2022 January 21]. Available from: https://www.clinicaltrials.gov/ct2/show/NCT04615871