You are on page 1of 14

Postgraduate Medicine

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/ipgm20

Efficacy and safety of semaglutide for weight


management: evidence from the STEP program

Anastassia Amaro, Danny Sugimoto & Sean Wharton

To cite this article: Anastassia Amaro, Danny Sugimoto & Sean Wharton (2022) Efficacy and
safety of semaglutide for weight management: evidence from the STEP program, Postgraduate
Medicine, 134:sup1, 5-17, DOI: 10.1080/00325481.2022.2147326

To link to this article: https://doi.org/10.1080/00325481.2022.2147326

© 2023 The Author(s). Published by Informa


UK Limited, trading as Taylor & Francis
Group.

Published online: 23 Jan 2023.

Submit your article to this journal

Article views: 3790

View related articles

View Crossmark data

Citing articles: 1 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ipgm20
POSTGRADUATE MEDICINE
2022, VOL. 134, NO. S1, 5–17
https://doi.org/10.1080/00325481.2022.2147326

SUPPLEMENT: SEMAGLUTIDE FOR WEIGHT MANAGEMENT – AN INTRODUCTION


FOR PRIMARY CARE

Efficacy and safety of semaglutide for weight management: evidence from the STEP
program
a b c
Anastassia Amaro , Danny Sugimoto and Sean Wharton
a
Penn Metabolic Medicine, Division of Endocrinology, University of Pennsylvania, Philadelphia, PA, USA; bCedar Crosse Research Center, Chicago,
IL, USA; cYork University, McMaster University and Wharton Weight Management Clinic, Toronto, Ontario, Canada

ABSTRACT ARTICLE HISTORY


Obesity is a global health challenge. It is a multifactorial, complex, and progressive disease associated Received 15 September
with various health complications and increased mortality. Lifestyle modifications are central to weight 2022
management but may be insufficient to maintain clinically meaningful weight loss. Pharmacotherapies Accepted 10 November
are recommended as an adjunct to lifestyle interventions to induce and sustain clinically meaningful 2022
weight loss and reduce the risk of comorbidities in appropriate patients. Glucagon-like peptide-1 is an KEYWORDS
incretin metabolic hormone responsible for a range of physiological effects, including glucose and Obesity; glucagon-like
appetite regulation. Several glucagon-like peptide-1 receptor agonists (GLP-1RAs) have been approved peptide-1 receptor agonist;
for the treatment of type 2 diabetes since 2005 including exenatide (short- and extended-release), semaglutide; primary care
lixisenatide, liraglutide, dulaglutide, albiglutide, and semaglutide. Of these, semaglutide (subcuta­
neous) and liraglutide are currently US Food and Drug Administration (FDA)-approved for chronic
weight management in patients with or without diabetes. The phase 3 Semaglutide Treatment Effect in
People with obesity (STEP) program was designed to investigate the effect of semaglutide versus
placebo on weight loss, safety, and tolerability in adults with overweight or obesity. Following the
submission of the results of the STEP 1–4 trials, the FDA approved once-weekly subcutaneous
semaglutide 2.4 mg for chronic weight management in people with overweight or obesity in
April 2021. Data from the program demonstrated that semaglutide (2.4 mg once weekly) achieved
significant and sustained weight loss, together with improvements in cardiometabolic risk factors
compared with placebo, and was generally well tolerated, with a safety profile consistent with other
GLP-1RAs. The most common adverse events reported in STEP 1–5 were gastrointestinal events, which
were transient, mild-to-moderate in severity, and typically resolved without permanent treatment
discontinuation. This article reviews the data from STEP 1–5 and highlights clinically relevant findings
for primary care providers.

1. Introduction As an adjunct to lifestyle interventions, appropriate use


of pharmacotherapies for weight management can help
Obesity is a chronic, progressive disease with a complex
people with overweight or obesity achieve greater magni­
pathophysiology and a multifactorial origin, including
genetic, metabolic, behavioral, sociocultural, and environ­ tudes of weight loss and maintain weight loss [8,9].
mental factors [1,2]. Obesity is a global pandemic, with the Historically, pharmacotherapy options have been limited,
worldwide prevalence nearly tripling since 1975 [3]. In 2016, with a need for treatment options that can induce and
39% of adults aged 18 years and older were overweight sustain clinically meaningful weight loss and improve asso­
(defined as body mass index [BMI] ≥25 kg/m2) and 13% ciated complications such as T2D and CVD [2,10]. Several
were living with obesity (defined as BMI ≥30 kg/m2) [3]. agents for weight management have been approved or are
Furthermore, in the US, 42.4% of adults were living with currently being investigated. The glucagon-like peptide-1
obesity from 2017 to 2018 [4]. (GLP-1) receptor agonist (GLP-1RA) semaglutide, which was
Obesity is associated with a decrease in life expectancy and initially approved for the treatment of T2D, has been shown
the development of clinical complications, including cardio­ to improve cardiovascular outcomes in these patients
vascular diseases (CVDs), metabolic diseases (type 2 diabetes [11,12], and was approved in 2021 for chronic weight man­
[T2D]), mechanical dysfunction (musculoskeletal disorders agement in people with overweight or obesity. This manu­
such as osteoarthritis), sleep apnea, and some malignancies
script discusses recently published results from the
[2,5]. Although weight loss can improve complications arising
Semaglutide Treatment Effect in People with obesity
from obesity, the magnitude of weight loss achieved with the
(STEP) 1–5 trials and considers the clinical implications of
lifestyle changes of diet and physical activity can be limited
these findings for the treatment of patients with obesity.
and difficult to maintain [6,7].

CONTACT Anastassia Amaro Anastassia.Amaro@pennmedicine.upenn.edu Penn Metabolic Medicine, 3737 Market Street, Philadelphia, PA 19104, USA
© 2023 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License (http://creativecommons.org/licenses/by-nc-nd/4.0/),
which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited, and is not altered, transformed, or built upon in any way.
6 A. AMARO ET AL.

pathways [23,24]. These effects lower blood glucose by increasing


Article overview and relevance to clinical practice the storage of excess glucose in muscles and fat, and decrease
● Obesity is a chronic, complex disease associated with appetite and further food (energy) consumption [13,14]. Insulin is
cardiometabolic and other complications. released in response to stimulation by GLP-1 when glucose levels
● As an adjunct to lifestyle interventions, pharmacotherapy are elevated, therefore plasma glucose levels are lowered in
can aid weight management in patients with overweight a glucose-dependent manner and hypoglycemia is unlikely [25].
or obesity. Endogenous GLP-1 has a short half-life in plasma (approxi­
● This supplement explores the clinical profile of the mately 1.5 to 2 minutes) owing to its rapid degradation by the
once-weekly GLP-1RA semaglutide 2.4 mg for chronic enzyme dipeptidyl peptidase-4 (DPP-4) [26]; therefore, research
weight management, based on results from the phase efforts have focused on the development of longer-acting
3 STEP clinical trial program, and provides practical GLP-1RAs for the management of obesity and diabetes, including
guidance on integrating once-weekly semaglutide semaglutide.
2.4 mg into clinical practice in primary care. The first GLP-1RA approved in the US was twice-daily exena­
● In this first article in the supplement, we: tide for the treatment of T2D in 2005. Since then, several other
● Review the mechanism of action of GLP-1RAs in obesity; GLP-1RAs have been approved, also for the treatment of T2D,
● Introduce semaglutide and summarize the design including once-daily analogs, lixisenatide and liraglutide and
of the STEP 1–5 trials; once-weekly molecules exenatide extended-release, dulaglutide,
● Explore the efficacy and safety profile of semaglu­ albiglutide, and semaglutide [27–33]. All of these GLP-1RAs are
tide 2.4 mg in the STEP 1–5 trials; administered as a subcutaneous injection, although a daily oral
● Discuss other potential safety considerations for preparation of semaglutide has also been recently approved [34].
GLP-1RAs. Of these, only subcutaneous semaglutide and liraglutide are
currently US Food and Drug Administration (FDA)-approved for
chronic weight management (semaglutide at 2.4 mg once weekly
2. Mechanism of action of glucagon-like peptide-1 and liraglutide at 3.0 mg once daily) and for glucose control in
receptor agonists in patients with obesity T2D (semaglutide at 0.5 mg, 1.0 mg, and 2.0 mg once weekly and
liraglutide at 1.2 mg and 1.8 mg once daily) [30,33,35,36].
GLP-1 is an incretin (metabolic hormone) secreted from L-cells
in the small and large intestine, and cells in the central ner­
vous system, predominantly in the brainstem [13]. Native Key clinical take-home points: Mechanism of action
GLP-1 mediates many physiological effects via GLP-1 receptors of GLP-1RAs in patients with obesity
found in various tissues in the body, including the brain, ● GLP-1 is an incretin hormone with multiple physiological
cardiovascular system, kidneys, lung, and gastrointestinal (GI) effects, including glucose-dependent enhancement of
system [14–17] (Figure 1). insulin production and suppression of glucagon secre­
Eating food triggers the release of endogenous GLP-1 within tion, and effects on control of appetite.
minutes. This hormone has several effects on the body, including ● Through central effects, GLP-1 receptor agonism
inhibiting glucagon secretion while enhancing insulin production, decreases appetite, encourages feelings of satiety,
both in a glucose-dependent manner [13,14,23], and encouraging and decreases energy intake.
feelings of fullness (satiety) through central effects on neural

GLP-1 is synthesized and secreted by: GLP-1R is expressed in:

Neurons in Body weight


hindbrain Brain Appetite

Lungs Satiety

Heart (AV node)


Glucose production
Gastric emptying
Pancreas Glucagon secretion
L-cells Apoptosis
of the gut
Kidney
Natriuresis
Diuresis
GI tract Insulin secretion
& bio-synthesis
Muscle

GLP-1R is not expressed in the liver

Figure 1. Location of GLP-1 synthesis and secretion, and GLP-1 receptor expression [13,14].
GLP-1 is secreted from two main sites in the body: L-cells in the gut and specialized cells in the nucleus tractus solitarius of the hindbrain. Native GLP-1 mediates its effects via GLP-1
receptors expressed in a broad range of tissues. GLP-1 facilitates a multitude of physiological actions to increase satiety, reduce appetite, and slow gastric emptying [14–22].
AV, atrioventricular; GI, gastrointestinal; GLP-1, glucagon-like peptide-1; GLP-1R, glucagon-like peptide-1 receptor.
POSTGRADUATE MEDICINE 7

Amino acid substitution at position 8


(Ala to Aib) protects against DPP-4 cleavage

8
His Aib Glu Gly Thr Phe Thr Ser Asp
Val

COOH
Spacer Ser
Lys Ala Ala Gln Gly Glu Leu Tyr Ser
Glu
Acetylation at position 26 with C18 fatty di-acid provides 26
strong binding to albumin Phe 34
Ile Ala Trp Leu Val Arg Gly Arg Gly

Amino acid substitution at position 34


(Lys to Arg) enables site-specific C18 fatty acid binding at position 26

Figure 2. Semaglutide key modifications from native GLP-1 [38–41].


Semaglutide is a close analog of native GLP-1, a 31-amino acid protein. Three important structural modifications have been made to the semaglutide molecule that extend its half-life to
approximately 1 week [38–41]. DPP-4, dipeptidyl peptidase-4; GLP-1, glucagon-like peptide-1.

3. Semaglutide The American Heart Association listed the STEP program as


one of the most pivotal scientific developments of 2021 [49].
Semaglutide is a human GLP-1 analog with 94% amino acid
homology to native GLP-1 and it has a half-life of approxi­
mately 1 week [37–39] (Figure 2).
Clinical studies in people with obesity have shown that 4.1. Eligibility
semaglutide affects body weight through multiple mechan­
Eligibility criteria for the STEP 1 to 5 trials included adults
isms that together lead to decreased energy intake and sub­
(≥18 years of age) with a stable body weight (≤5 kg weight
sequent weight loss [42,43].
change within 90 days before screening) and a history of at
Once-weekly subcutaneous semaglutide 2.4 mg is
least one self-reported unsuccessful dietary effort to lose
approved in the US as an adjunct to a reduced calorie diet
weight [2,44–48]. In addition, for the STEP 1, 3, 4, and 5 trials,
and increased physical activity for chronic weight manage­
ment in adults with overweight (BMI ≥27 kg/m2) with at participants were required to have a BMI of either ≥30 kg/m2,
least one weight-related comorbidity (e.g. hypertension, T2D, or ≥27 kg/m2 combined with at least one weight-related
or dyslipidemia), or obesity (BMI ≥30 kg/m2) [33]. comorbidity which could be dyslipidemia, obstructive sleep
apnea, hypertension, or CVD. In the STEP 2 trial, participants
were required to have a BMI of ≥27 kg/m2, a diagnosis of T2D
Key clinical take-home points: Semaglutide at least 180 days before screening, glycated hemoglobin levels
● Semaglutide is a GLP-1 analog that is approved as an of 7% to 10%, and to be managed with diet and exercise alone
adjunct to a reduced calorie diet and increased phy­ or treated with up to three oral glucose-lowering drugs for at
sical activity for chronic weight management as least 90 days before screening [44].
a once-weekly subcutaneous 2.4 mg dose in adults In the STEP 1, 3, 4, and 5 trials, patients were excluded if
with: they had renal impairment measured as an estimated glomer­
2
● Overweight (BMI ≥27 kg/m ) with at least one ular filtration rate value of <15 mL/min/1.73 m2 [45–48].
weight-related comorbidity; Additionally, participants with diabetes (type 1 or 2), glycated
2 hemoglobin levels of ≥6.5%, or who had received treatment
● Obesity (BMI ≥30 kg/m ).
with either glucose-lowering agents or anti-obesity medica­
tions within the prior 90 days were excluded [45–48]. In
STEP 2, patients were excluded if they had an estimated glo­
4. The Semaglutide Treatment Effect in People with merular filtration rate value of <30 mL/min/1.73 m2 (<60 mL/
obesity trials min/1.73 m2 in patients treated with sodium-glucose co-
As demonstrated in the STEP clinical development program, transporter-2 inhibitors) [44]. In the STEP 1 to 5 trials, additional
semaglutide produces clinically meaningful and durable exclusion criteria included participants with a history or pre­
weight loss [2,44–48]. Here, we discuss the STEP 1 to 5 trials: sence of chronic pancreatitis or with acute pancreatitis within
the phase 3, double-blinded, randomized, multinational trials the past 180 days [2], those who had a personal or first-degree
(STEP 3 was US only) that assessed once-weekly subcutaneous relative history of medullary thyroid cancer or multiple endo­
semaglutide 2.4 mg versus placebo for weight management in crine neoplasia type 2 [44–48], and participants classified as
adults with obesity or overweight and with and without T2D. being in New York Heart Association Class IV [2].
8 A. AMARO ET AL.

4.2. Trial design phase, where 327 participants were followed for an addi­
tional 45 weeks (a total of 52 weeks off-treatment) until
A total of 4,988 participants across the STEP 1 to 5 trials were
the end-of-trial visit at week 120 [50].
randomized to receive either semaglutide or placebo. To miti­
A primary endpoint for all trials was percentage change
gate side effects, the trials in the STEP program were designed
from baseline (at randomization) to end of treatment in body
to slowly escalate the dose of semaglutide over a 16-week
weight (Table 1), where baseline was week 0 in STEP 1–3, and
period. Therefore, once-weekly subcutaneous semaglutide
5, and week 20 in STEP 4. In addition, STEP 1–3 and 5 included
was initiated at a dose of 0.25 mg and escalated every
4 weeks to the subsequent dosing levels of 0.5 mg, 1.0 mg, the proportion of participants achieving weight loss of ≥5%
and 1.7 mg, until reaching the target dose of 2.4 mg. STEP 2 from baseline at end of treatment as a co-primary endpoint
included a semaglutide 1.0 mg treatment arm where patients (Table 1). For efficacy analyses, the results described in this
were escalated every 4 weeks, from 0.25 mg to 0.5 mg, and article are based on analyses that assessed treatment effects
then to the target dose of 1.0 mg [2,44–48]. In STEP 1, 2, 4, and regardless of adherence to treatment or the use of rescue
5, participants received treatment as an adjunct to lifestyle medications (anti-obesity medications or bariatric surgery)
intervention (monthly counseling on a −500-kcal/day diet [2,44–48]. This analysis approach is referred to as the ‘treat­
relative to estimated energy expenditure at week 0 and ment policy estimand’ in the original STEP trial results pub­
a recommended 150 minutes/week of physical activity). In lications (for further information on estimands in
STEP 3, patients received treatment as an adjunct to intensive contemporary reporting of clinical trial results in weight man­
behavioral therapy (IBT; defined as 30 individual dietitian vis­ agement, see Wharton et al. [51]) [2,44–48].
its) including an initial 8-week low-calorie diet with partial Statistical analysis for continuous endpoints were
meal replacement followed by a hypocaloric diet for achieved using an analysis of covariance model with ran­
60 weeks, together with physical activity (increasing gradually domized treatment, stratification groups, and the interac­
from 100 minutes/week to 200 minutes/week). tion between stratification groups as factors and baseline
The trials included a 68-week treatment period endpoint value as covariate. Missing data were imputed
(104-week treatment period for STEP 5 only) followed by and the results were combined using Rubin’s rules.
a 7-week off-treatment follow-up period [2]. STEP 4 Categorical endpoints were analyzed by logistical regres­
included a 20-week run-in period where all participants sion [44].
received once-weekly subcutaneous semaglutide 0.25 mg Participants were mostly white (62.1% to 93.1%), had
at week 0 and escalated dose every 4 weeks until reaching a mean age of 46.2 to 55.3 years, were mostly female (50.9%
the target of 2.4 mg. At week 20, after 4 weeks of treat­ to 81.0%), had a mean BMI of 35.7 to 38.5 kg/m2, and a mean
ment at the target dose of semaglutide, STEP 4 partici­ waist circumference of 113.0 to 115.7 cm [2,44–48].
pants were randomized to continue once-weekly A summary of the trial details and key outcomes are shown
subcutaneous semaglutide 2.4 mg or switch to placebo in Table 2. In the subsequent sections, we summarize the main
[2,45]. The STEP 1 trial had an off-treatment extension findings.

Table 1. Key endpoints of the STEP trials [2,44–48].


STEP 2 STEP 3 STEP 4 STEP 5
STEP 1 Weight management Weight management Sustained weight Long-term weight
Weight management in T2D with IBT (US only) managementc management
Primary/co-primary endpoints
For once-weekly subcutaneous semaglutide 2.4 mg vs placebo
% change in body weighta ✓ ✓ ✓ ✓ ✓
Achievement of ≥5% body weight ✓ ✓ ✓ ✓
lossb
Confirmatory secondary endpoints
For once-weekly subcutaneous semaglutide 2.4 mg vs placebo
Achievement of ≥10% body weight ✓ ✓ ✓ ✓
lossb
Achievement of ≥15% body weight ✓ ✓ ✓ ✓
lossb
a
Change in waist circumference ✓ ✓ ✓ ✓ ✓
Change in systolic blood pressurea ✓ ✓ ✓ ✓ ✓
Change in SF-36 Physical Functioning ✓ ✓ ✓ ✓
a
score
Change in IWQOL-Lite-CT Physical ✓ ✓
Function scorea
Change in HbA1ca ✓
For once-weekly subcutaneous semaglutide 2.4 mg vs once-weekly subcutaneous semaglutide 1.0 mg
a
% change in body weight ✓
a
Assessed as change from baseline (randomization) to end of treatment at week 68 (STEP 1–4) or week 104 (STEP 5). bAssessed from baseline (randomization) at
week 68 (STEP 1–4) or at week 104 (STEP 5). cSTEP 4 primary and confirmatory secondary endpoints were assessed from randomization (week 20) to week 68.
HbA1c, glycated hemoglobin; IBT, intensive behavioral therapy; IWQOL-Lite-CT, Impact of Weight on Quality of Life-Lite Clinical Trials; SF-36, 36-Item Short Form
Health Survey; T2D, type 2 diabetes.
Table 2. Trial details and key outcomes of the STEP 1 to 5 trials [2,44–48].
STEP 3 STEP 4 STEP 5
STEP 1 STEP 2 Weight management Sustained weight Long-term weight
Weight management, Weight management with IBTc (US only), management management
without T2Da in T2Db without T2Da without T2Da without T2Da
Trial length (weeks) 68 68 68 68 104
Participants 1,961 1,210 611 Run-in period: 902 304
Randomized: 803
Trial objectives To show superiority of To show superiority of To maximize the effect To maintain the effect of To show superiority of
semaglutide 2.4 mg OW semaglutide 2.4 mg vs placebo and of semaglutide 2.4 semaglutide 2.4 mg vs semaglutide 2.4 mg vs
vs placebo on WL semaglutide 1.0 mg OW on WL mg vs placebo on placebo on WL from placebo on WL and to
To assess safety and To assess safety and tolerability WL randomization to EOT compare safety and
tolerability and baseline (week tolerability in adults
20) to EOT with obesity or
To compare safety in overweight after 2
adults with obesity or years of treatment
overweight who
reached the target
dose of semaglutide
during 20-week run-in
Interventions Lifestyle interventiond Lifestyle interventiond plus: IBTc plus: Lifestyle interventiond Lifestyle interventiond
plus: Semaglutide 2.4 mg OW Semaglutide 2.4 mg plus: plus:
Semaglutide 2.4 mg OW or OW Run-in period (weeks 0– Semaglutide 2.4 mg OW
or Semaglutide 1.0 mg OW or 20) or
Placebo or Placebo Semaglutide OW (target Placebo
(randomized 2:1) Placebo (randomized 2:1) dose of 2.4 mg) (randomized 1:1)
(randomized 1:1:1) From week 20
Semaglutide 2.4 mg OW
or
Placebo (randomized 2:1)
Key results
Semaglutide Placebo Semaglutide Semaglutide Placebo Semaglutide Placebo Semaglutide Placebo Semaglutide Placebo
2.4 mg 2.4 mg 1.0 mg 2.4 mg 2.4 mg 2.4 mg
Mean body weight change from baselinee,f, % –14.9 –2.4 –9.6 –7.0 –3.4 –16.0 –5.7 –7.9 6.9 –15.2 –2.6
Participants with ≥5% body weight lossg,h, % 86.4 31.5 68.8 57.1 28.5 86.6 47.6 88.7 47.6 77.1 34.4
g,h
Participants with ≥10% body weight loss , % 69.1 12.0 45.6 28.7 8.2 75.3 27.0 79.0 20.4 61.8 13.3
Improvements in trial endpoints with semaglutide 2.4 mg vs placebo ● Waist circumference ● Waist circumference ● Waist circumference ● Waist circumference ● Waist circumference
● BMI ● BMI ● BMI ● BMI ● BMI
● Blood pressure ● Systolic blood pressure ● Blood pressure ● Systolic blood ● Blood pressure
● HbA1c level ● HbA1c level ● HbA1c level pressure ● HbA1c
● Fasting plasma glucose ● Fasting plasma glucose levels ● Fasting plasma glu­ ● HbA1c level ● Lipid profile
i
levels ● Lipid profile cose levels ● Lipid profile ● Fasting plasma glucose
● Lipid profile ● C-reactive protein levels ● Lipid profile ● Fasting plasma glu­ levels
● C-reactive protein levels ● SF-36 Physical Functioning score ● C-reactive protein cose levels ● C-reactive protein level
● SF-36 Physical ● IWQOL-Lite-CT Physical Function level ● SF-36 Physical ● A greater proportion of
Functioning score score ● SF-36 Mental Functioning score, and participants with predia­
● IWQOL-Lite-CT Physical ● Urine albumin-to-creatinine ratio Component Physical and Mental betes at baseline had
Function score ● Liver parameters Summary score Component Summary normoglycemia at
● A greater proportion of scores week 104
participants with pre­
diabetes at baseline had
normoglycemia at
week 68
POSTGRADUATE MEDICINE
9
10
A. AMARO ET AL.

a
Adults with overweight or obesity (BMI ≥30.0 kg/m2 or ≥27.0 kg/m2) with at least one comorbidity, and without diabetes. bAdults with overweight or obesity and T2D, with HbA1c levels of 7.0% to 10.0%. cIBT, including initial 8-week low-
calorie diet followed by a hypocaloric diet for 60 weeks, 30 counseling sessions, and 100 minutes/week of physical activity increased by 25 minutes/week every 4 weeks until 200 minutes/week. dParticipants received treatment as an adjunct
to lifestyle intervention (monthly counseling on a −500-kcal/day diet relative to estimated energy expenditure at week 0, and a recommended 150 minutes/week of physical activity). eAssessed as change from baseline (randomization) to
end of treatment at week 68 (STEP 1–4) or week 104 (STEP 5). fAnalyses assessed treatment effects regardless of adherence to treatment or the use of rescue medications (anti-obesity medications or bariatric surgery) (treatment policy
estimand data). gAssessed from baseline (randomization) at week 68 (STEP 1–4) or at week 104 (STEP 5). hIn-trial data, defined as the time from randomization to last contact with trial site, irrespective of treatment discontinuation or rescue
intervention. iFor patients who maintained semaglutide versus those who switched to placebo over weeks 20 to 68, a non-significant difference was observed for the high-density lipoprotein cholesterol and free-fatty acids lipid values.
BMI, body mass index; EOT, end of trial; HbA1c, glycated hemoglobin; IBT, intensive behavioral therapy; IWQOL-Lite-CT, Impact of Weight on Quality of Life-Lite Clinical Trials Version; OW, once weekly; T2D, type 2 diabetes;
SF-36, 36-Item Short-Form Health Survey; WL, weight loss.
POSTGRADUATE MEDICINE 11

the importance of recognizing obesity as a chronic disease


Key clinical take-home points: The Semaglutide requiring long-term treatment to maintain beneficial thera­
Treatment Effect in People with obesity trials peutic effects.
● The STEP 1–5 trials were phase 3, double-blind, ran­
domized trials that assessed once-weekly subcuta­ 4.3.2. STEP 2
neous semaglutide 2.4 mg versus placebo for weight STEP 2 demonstrated that in patients with T2D and over­
management in adults with obesity or overweight. weight or obesity, once-weekly subcutaneous semaglutide
● Trials included patients with T2D (STEP 2) and without 2.4 mg (n = 404) plus lifestyle intervention led to greater
T2D (STEP 1 and 3–5). reductions in body weight than placebo (n = 403) or semaglu­
● STEP 1–3 comprised 68-weeks’ treatment. Patients in tide 1.0 mg (n = 403). The mean weight loss in the semaglu­
STEP 1 were also followed for an additional 45 weeks tide 2.4 mg group was 9.6% versus 7.0% for the semaglutide
(a total of 52 weeks off-treatment) until the end-of- 1.0 mg group and 3.4% for the placebo group. The ETD for
trial visit at week 120. semaglutide 2.4 mg versus placebo was −6.2 percentage
● STEP 4 included an initial 20-week semaglutide run-in points (95% CI: −7.3, −5.2%; P < 0.0001), whereas it was
period, followed by randomization to continued sema­ −2.7 percentage points (95% CI: −3.7%, −1.6; P < 0.0001) for
glutide 2.4 mg or switch to placebo for the remaining semaglutide 2.4 mg versus semaglutide 1.0 mg. A greater
48 weeks. proportion of participants achieved ≥5% weight loss with
● STEP 5 comprised 104-weeks’ treatment. semaglutide 2.4 mg (68.8%) versus placebo (28.5%;
● All trials used a gradual dose escalation over the initial P < 0.0001 for odds) and versus semaglutide 1.0 mg (57.1%
16-week period to mitigate potential side effects. [statistical significance not tested]). Semaglutide 2.4 mg also
● Trial participants also received lifestyle intervention resulted in improvement in cardiometabolic risk factors com­
(counseling on diet and physical activity) or IBT pared with placebo [44].
(STEP 3 only; including 30 therapy sessions, physical
activity, and an initial 8-week low-calorie diet with 4.3.3. STEP 3
partial meal replacement followed by a hypocaloric When used as an adjunct to IBT and an initial low-calorie diet,
diet). once-weekly subcutaneous semaglutide 2.4 mg (n = 407) pro­
● STEP 1–5 trial endpoints assessed changes in body duced significantly greater weight loss than placebo (n = 204)
weight and cardiometabolic parameters. Changes in (16.0% vs 5.7%; treatment difference of −10.3 percentage
self-reported physical functioning were also assessed points [95% CI: −12.0, −8.6]; P < 0.001) at 68 weeks in adults
in STEP 1–4. with overweight or obesity without T2D [46].
Interpreting the findings of STEP 3 (placebo-subtracted
weight loss of 10.3%) in the context of STEP 1 (placebo-
4.3. Efficacy of semaglutide in the STEP 1 to 5 trials subtracted weight loss of 12.4%), the inclusion of an intensive
4.3.1. STEP 1 lifestyle intervention (including a partial meal replacement
In STEP 1, in adults with overweight or obesity without T2D program and 30 treatment sessions) provided only a modest
receiving semaglutide (n = 1,306) or placebo (n = 655), once- contribution to additional weight loss beyond that achieved
weekly subcutaneous semaglutide plus lifestyle intervention with semaglutide and less intensive lifestyle intervention. It
from baseline to week 68 was associated with substantial, sus­ should be noted, however, that definitive conclusions about
tained, clinically relevant mean weight loss of 14.9% versus 2.4% the contributions of IBT and less intensive lifestyle interven­
for placebo, with an estimated treatment difference (ETD) of tion cannot be made [46].
−12.4 percentage points (95% confidence interval [CI]: −13.4,
−11.5; P < 0.001). Consistent with this finding, a greater propor­ 4.3.4. STEP 4
tion of participants achieved the co-primary endpoint of ≥5% Among adults with overweight or obesity completing a 20-week
weight loss with semaglutide (86.4%) versus placebo (31.5%; run-in period on subcutaneous semaglutide 2.4 mg (n = 535;
P < 0.001 for odds). Semaglutide was also associated with mean change in body weight –10.6%), maintaining treatment
greater improvements compared with placebo in cardiometa­ with semaglutide compared with switching to placebo (n = 268)
bolic risk factors, including reductions in waist circumference, resulted in continued weight loss; the mean weight loss from
blood pressure, glycated hemoglobin levels, and lipid levels [47]. week 20 to 68 in the semaglutide group was 7.9%, compared
These endpoints are discussed in Article 2 in the supplement by with a weight gain of 6.9% in the placebo group, with a treatment
Amaro et al. difference of −14.8 percentage points (95% CI: −16.0, −13.5;
In the 52-week off-treatment extension phase of STEP 1 P < 0.001) [45].
(n = 327), there was weight regain in both treatment arms
resulting in net weight loss of 5.6% with semaglutide 2.4 mg 4.3.5. STEP 5
and 0.1% with placebo over the full 120-week period [50]. In In STEP 5, once-weekly subcutaneous semaglutide 2.4 mg
addition, the improvements in cardiometabolic risk factors (n = 152) resulted in substantial initial body weight reductions
observed from baseline to week 68 reverted toward baseline that were then maintained over 104 weeks compared to pla­
at week 120 for most endpoints [50]. These results highlight cebo (n = 152) [48]. There was no additional weight loss
12 A. AMARO ET AL.

between weeks 52–104; weight loss was maintained during this More participants discontinued treatment with semaglutide
period [48]. The mean weight loss in the semaglutide group 2.4 mg than placebo due to GI AEs in STEP 1–3 and 5, but
was –15.2%, compared to –2.6% in the placebo group (ETD: overall few participants discontinued treatment due to such
−12.6 percentage points [95% CI: –15.3, −9.8] P < 0.0001). AEs [44–48]. In STEP 4, most GI AEs occurred during the run-in
Similar to the results of the STEP 1–3 trials, a greater proportion period, during which semaglutide was escalated to the target
of participants achieved ≥5% weight loss with semaglutide maintenance dose of 2.4 mg once weekly in all participants
2.4 mg versus placebo (77.1% vs 34.4%; P < 0.0001 for odds). who entered the randomized period. After the 20-week run-in
Treatment with semaglutide also improved cardiometabolic risk period, the proportion of participants discontinuing treatment
factors compared with placebo, indicating a favorable benefit- due to any AE was low among those randomized to continued
risk profile of semaglutide 2.4 mg for the long-term manage­ semaglutide 2.4 mg treatment and was similar to the rate in
ment of weight. The effects of semaglutide 2.4 mg on cardio­ those who switched to placebo [45].
metabolic parameters and other outcomes in STEP 1–5 are GI side effects, which are common to all GLP-1RAs, may
discussed in subsequent articles in this supplement. affect patient adherence [52]. However, such GI effects are
typically mild-to-moderate in severity and decline in preva­
Key clinical take-home points: Efficacy of semaglutide lence after initial dose-escalation, particularly for nausea, and
in the STEP 1 to 5 trials a gradual dose escalation schedule may help alleviate or pre­
● The STEP 1–3 68-week trials show that significant vent GI side effects [52,53]. Recommendations for the manage­
weight loss of 15% to 16% in participants without ment of GI side effects with GLP-1RAs in clinical practice
T2D, and 9.6% in those with T2D, can be achieved suggest an approach described as ‘the three Es: Education
with once-weekly subcutaneous semaglutide 2.4 mg and explanation, Escalation to an appropriate dose, and
in adults with overweight or obesity. Effective management of GI side effects’ [53]. As part of the
● In STEP 4, switching to placebo after a 20-week run-in Education and explanation approach, patients should be
period on semaglutide 2.4 mg resulted in weight counseled on the potential for GI side effects and their usual
regain, while continuing treatment with semaglutide mild-to-moderate nature, alongside recommendations for
2.4 mg resulted in further substantial weight loss after dietary modifications and management of current GI symp­
48 weeks. toms. Gradual escalation to an appropriate dose is detailed in
● Improvements in weight loss were maintained over the prescribing information for many GLP-1RAs and should
a longer term of 104 weeks in STEP 5. therefore be the standard approach when initiating treatment;
● Results from the STEP 4 and 5 trials highlight the however, for patients reporting challenges with GI symptoms
chronicity of obesity and the importance of longer- during the first weeks of treatment, a slower dose escalation is
term treatment to achieve and maintain clinically recommended as part of ‘the three Es.’ Finally, effective man­
meaningful weight loss. agement of GI side effects is essential, and a stepwise, sever­
● Substantially more patients achieved clinically mean­ ity-based approach is recommended, including dietary
ingful weight losses of ≥5% with semaglutide 2.4 mg modifications for mild side effects, identifying/ruling out any
in the STEP 1–5 trials than with placebo. underlying GI disorders, GLP-1RA dose adjustment, considera­
tion of pharmacological treatment, and switching to an alter­
native to GLP-1RAs [53]. Further information on such practical
considerations for GLP-1RA use is provided by Kyrillos et al. in
the final article in this supplement.
5. Overall safety profile of semaglutide 2.4 mg in
GLP-1RAs have a glucose-dependent mechanism of action,
the STEP 1 to 5 trials
therefore there is no expectation of symptomatic hypoglyce­
Semaglutide 2.4 mg once weekly was generally well tolerated mia and there were no concerns identified in the STEP trials
in adults with obesity, or overweight with comorbidities, with­ [23]. In STEP 1, hypoglycemia was reported as an AE in 0.6% of
out T2D (STEP 1, 3, 4, and 5), and in adults with T2D and participants in the semaglutide group versus 0.8% of partici­
overweight or obesity (STEP 2) [44–48]. Table 3 gives pants in the placebo group [47]. In STEP 2, which included
a summary of the safety profile of semaglutide 2.4 mg in the participants with T2D, severe or blood-glucose symptomatic
STEP 1 to 5 trials. hypoglycemia was reported in 5.7% and 5.5% of participants
The most common adverse events (AEs) among people in the 2.4 mg and 1.0 mg semaglutide groups, respectively,
treated with semaglutide 2.4 mg were GI events (most com­ versus 3.0% in the placebo group [44]. It is important to note
monly nausea, diarrhea, vomiting, and constipation) [44–47]. that during STEP 2, participants were permitted to receive
Most GI AEs in STEP 1 to 5 were mild-to-moderate in severity a stable dose of up to three oral glucose-lowering agents
and resolved without permanent treatment discontinuation [44]. In the total STEP 2 population, the most commonly
[42–46]. These GI AEs were transient, with median durations received agents were biguanides (92% of participants), sulfo­
with semaglutide 2.4 mg lasting up to 8 days for nausea, nylureas (25% of participants), and sodium-glucose co-
5 days for diarrhea, 2 days for vomiting, and 55 days for transporter-2 inhibitors (25% of participants) [44]. In STEP 3,
constipation [44–48]. Nausea was typically most prevalent 0.5% of participants in the semaglutide group and none in the
during the initial dose-escalation periods, and the proportion placebo group reported hypoglycemia as an AE [46].
of participants with nausea declined thereafter [44,46–48]. Hypoglycemia incidence was similarly low in STEP 4, with
Table 3. Summary of AEs from the STEP 1 to 5 trials [2,44–48].
STEP 3 STEP 5
STEP 1 STEP 2a Weight management STEP 4 Long-term
Weight management Weight management in T2D with IBT (US only) Sustained weight management weight management
Semaglutide
Run-in 2.4 mg
Semaglutide Semaglutide Semaglutide Semaglutide (semaglutide (randomized Placebo (randomized Semaglutide
2.4 mg Placebo 2.4 mg 1.0 mg Placebo 2.4 mg Placebo 2.4 mg) period) period) 2.4 mg Placebo
AEs, % 89.7 86.4 87.6 81.8 76.9 95.8 96.1 84.3 81.3 75 96.1 89.5
GI AEs, % 74.2 47.9 63.5 57.5 34.3 82.8 63.2 71.4 41.9 26.1 82.2 53.9
Nausea 4.2 17.4 33.7 32.1 9.2 58.2 22.1 46.8 14.0 4.9 53.3 21.7
Diarrhea 31.5 15.9 21.3 22.1 11.9 36.1 22.1 23.5 14.4 7.1 34.9 23.7
Constipation 23.4 9.5 17.4 12.7 5.5 36.9 24.5 22.2 11.6 6.3 30.9 11.2
Vomiting 24.8 6.6 21.8 13.4 2.7 27.3 10.8 15.5 10.3 3.0 30.3 4.6
Median duration of GI AEs, days
Nausea 8 6 8 10 6 5 5 NR NR NR 4 2
Diarrhea 3 3 5 4 4 3 3 NR NR NR 5 3
Constipation 35 25 55 51 21 27 16 NR NR NR 58 39
Vomiting 2 1 1 2 1 2 2 NR NR NR 2 2
SAEs, % 9.8 6.4 9.9 7.7 9.2 9.1 2.9 2.3 7.7 5.6 7.9 11.8
AEs leading to trial product 7.0 3.1 6.2 5.0 3.5 5.9 2.9 5.3 2.4 2.2 5.9 4.6
discontinuation, %
GI AEs leading to trial product 4.5 0.8 4.2 3.5 1.0 3.4 0 NR NR NR 3.9 0.7
discontinuation, %
a
STEP 2 was the only trial that enrolled patients with T2D.
AEs, adverse events; GI, gastrointestinal; IBT, intensive behavioral therapy; NR, not reported; SAEs, serious adverse events; T2D, type 2 diabetes.
POSTGRADUATE MEDICINE
13
14 A. AMARO ET AL.

hypoglycemia reported as an AE in 0.1% of participants during A consensus statement by the American Association of
the initial 20-week semaglutide run-in period, and thereafter Clinical Endocrinologists and the American College of
in 0.6% of participants continuing semaglutide 2.4 mg and in Endocrinology on the management of T2D recommends that
1.1% of participants switching to placebo [45]. In STEP 5, GLP-1RAs should be used cautiously (if at all) in patients with
hypoglycemia was reported as an AE in 2.6% of participants a history of pancreatitis (due to a lack of clinical trial data), and
in the semaglutide group and none in the placebo group [48]. that treatment should be discontinued if acute pancreatitis
develops [62].
Key clinical take-home points: Overall safety profile
of semaglutide 2.4 mg in the STEP 1 to 5 trials
● Semaglutide 2.4 mg once weekly was generally well 6.2. Gallbladder disorders
tolerated in adults with overweight or obesity in the
STEP 1–5 trials. Weight loss is known to increase the risk of cholelithiasis,
● The safety profile of semaglutide was consistent with with new gallstone prevalence reaching 10% to 12% after 8
the known profile of GLP-1RAs, with GI events the to 16 weeks of a low-calorie diet and reaching greater than
most commonly reported AEs. 30% within 12 to 18 months after gastric bypass sur­
gery [63].
● Most GI events were transient, mild-to-moderate in
severity, and resolved without permanent treat­ GLP-1RA treatment has been linked with an increase in
ment discontinuation. gallbladder AEs, including cholelithiasis and cholecystitis
● Gradual dose-escalation may help alleviate or prevent [64–66]. In STEP 1, 3, and 5, gallbladder-related disorders
GI side effects – practical guidance on this and other were reported in a higher proportion of participants in the
topics is discussed in more detail in the final article in semaglutide 2.4 mg groups (2.6%, 4.9%, and 2.6%, respec­
this supplement by Kyrillos et al. tively) compared with the placebo groups (1.2%, 1.5%, and
1.3%, respectively). In STEP 2 and during maintenance dos­
ing in STEP 4 (week 20 onwards), slightly fewer participants
experienced gallbladder-related disorders in the semaglu­
6. Addressing other potential safety concerns tide 2.4 mg groups (0.2% and 2.8%, respectively) compared
with the placebo groups (0.7% and 3.7%, respectively)
6.1. Pancreatitis
[44–48].
Cases of pancreatitis have been described in connection with It is important to note that the presence of gallstones
the use of GLP-1RAs, including exenatide and liraglutide, and was not an exclusion criterion in any of the STEP trials [67].
also with DPP-4 inhibitors such as sitagliptin [54,55]. However,
trials and a meta-analysis have shown that GLP-1RA treatment
does not appear to substantially increase pancreatic events 6.3. Thyroid C-cell tumors
[52,56].
In the STEP 1 to 5 trials, participants with a history or The prescribing information for semaglutide, as well as other
presence of chronic pancreatitis, or with acute pancreatitis FDA-approved long-acting GLP-1RAs (albiglutide, dulaglutide,
within the past 180 days, were excluded. In STEP 1–5, there extended-release exenatide, and liraglutide), state that they
were very few reports of acute pancreatitis and no notable have been reported to cause thyroid C-cell tumors in rodents,
difference in the incidences in semaglutide 2.4 mg groups including medullary thyroid carcinoma (MTC), while also not­
(0–0.2% of participants across trials) and placebo groups ing that the human relevance of this has not been determined
(0–0.2% of participants across trials) [44–48]. [29–33].
Amylase and lipase are biomarkers of pancreatic inflam­ MTC is distinct from other types of thyroid cancers (derived
mation and are routinely measured in clinical trials of incre­ from thyroid follicular cells) as it originates from the parafolli­
tin-based therapies as a regulatory requirement [57]. Both cular C-cells of the thyroid gland [68]. Furthermore, the pre­
GLP-1RAs and DPP-4 inhibitors have been linked to valence of MTC is very low, comprising 2% to 4% of all thyroid
increased levels of serum lipase and serum amylase [58]. cancers [68,69].
Elevations in amylase and lipase have been described fol­ A meta-analysis of 11 cardiovascular outcomes studies of
lowing treatment with GLP-1RAs and DPP-4 inhibitors, how­ GLP-1RAs including over 55,000 patients identified no
ever little is known about amylase or lipase activity in increased risk of MTC with GLP-1RAs [70].
individuals with overweight or obesity without T2D, or No cases of MTC were reported in STEP 1 to 5.
whether routinely monitoring these enzymes can predict Furthermore, there were no imbalances in calcitonin levels
subsequent acute pancreatitis onset [57]. between semaglutide 2.4 mg and the placebo group in
In STEP 3, increases in amylase were seen in one patient these studies (calcitonin is a marker for potential MTC)
treated with semaglutide, and one patient treated with sema­ [44–48].
glutide had increased lipase [46]. Owing to contraindications of FDA-approved long-
As with other GLP-1RAs and DDP-4 inhibitors, the prescribing acting GLP-1RAs, as noted earlier, participants with
information for subcutaneous semaglutide in both T2D and a personal or first-degree relative history of MTC or multi­
obesity includes warnings and precautions relating to pancrea­ ple endocrine neoplasia type 2 were excluded from STEP 1
titis [29–33,59–61]. to 5 [44–48].
POSTGRADUATE MEDICINE 15

obesity in the SURMOUNT-1 clinical trial [72]. Currently, there


Key clinical take-home points: Addressing other are no published head-to-head trials of weight management
potential safety concerns agents, except for the STEP 8 trial which evaluated semaglutide
● The prescribing information for semaglutide should be versus liraglutide in adults with overweight or obesity. STEP 8
referred to for information on contraindications, warn­ showed that semaglutide resulted in significantly greater weight
ings, and precautions relating to pancreatitis, gallbladder loss at 68 weeks compared with liraglutide [73].
disorders, and MTC. Obesity management continues to evolve. Ten years ago
● Participants with a history or presence of chronic there were very limited treatments for people living with obesity.
pancreatitis, or with acute pancreatitis within the Today, we are at the stage where many individuals can receive
past 180 days, were excluded from the STEP trials. effective treatment for this chronic condition. The use of sema­
● In STEP 1–5, there were very few reports of acute glutide in conjunction with lifestyle intervention provides sus­
pancreatitis and no notable difference in the inci­ tained, clinically relevant reductions in body weight and offers an
dences between semaglutide 2.4 mg groups and effective, pharmacotherapeutic option with a magnitude of
placebo groups. weight loss increasingly closing the gap to bariatric surgery
● GLP-1RA treatment has been linked with an increase [74–77]. The future is much brighter for those living with obesity.
in gallbladder AEs, including cholelithiasis and chole­
cystitis, and a slightly higher incidence of gallbladder-
related disorders was reported with semaglutide ver­ Abbreviations
sus placebo in some STEP studies. AE, adverse event
● No cases of MTC were reported in STEP 1 to 5. AV, atrioventricular
BMI, body mass index
CI, confidence interval
CVD, cardiovascular disease
7. Conclusions DPP-4, dipeptidyl peptidase-4
EOT, end of trial
The road to ideal pharmacological weight management has ETD, estimated treatment difference
been complex, and GLP-1RAs offer opportunities for effective FDA, US Food and Drug Administration
weight control alongside lifestyle interventions. GI, gastrointestinal
GLP-1, glucagon-like peptide-1
The primary goal of pharmacotherapies like semaglutide
GLP-1RA, glucagon-like peptide-1 receptor agonist
when combined with lifestyle intervention for chronic weight HbA1c, glycated hemoglobin
management is to achieve a clinically meaningful weight loss. IBT, intensive behavioral therapy
A secondary goal is to provide long-term weight management IWQOL-Lite-CT, Impact of Weight on Quality of Life-Lite Clinical Trials
and to minimize weight regain [71]. The phase 3 STEP 1 to 5 MTC, medullary thyroid carcinoma
NR, not reported
trials have shown that significant and sustained weight loss of
OW, once weekly
14% to 16% in participants without T2D, and 9.6% in those SAEs, serious adverse events
with T2D, can be achieved with a once-weekly dose of sema­ SF-36, 36-Item Short Form Health Survey
glutide, offering clinicians a new tool to manage obesity and STEP, Semaglutide Treatment Effect in People with obesity
associated complications. T2D, type 2 diabetes
WL, weight loss
In the STEP 1 to 5 trials, semaglutide 2.4 mg once weekly
was generally well tolerated with a safety profile consistent
with the GLP-1RA class. Mild-to-moderate and transient GI
Acknowledgments
events were the most common AEs and resolved without
permanent treatment discontinuation in most cases [44–48]. Medical writing support was provided by Laura Moore, PhD, of Axis,
Obesity is being increasingly recognized as a chronic dis­ a division of Spirit Medical Communications Group Limited, and Gemma
Hall, a contract writer working on behalf of Axis, and funded by Novo
ease which requires long-term treatment. Results from the
Nordisk Inc., in accordance with Good Publication Practice 3 (GPP3) guide­
STEP 4 and 5 trials support the need for continued therapy lines (www.ismpp.org/gpp3).
with semaglutide to maintain weight loss [45,48], further
demonstrated by the STEP 1 extension phase in which parti­
cipants regained weight upon withdrawal of treatment [50]. Funding
Obesity is also a crucial contributor to CVD. The ongoing
Semaglutide Effects on Heart Disease and Stroke in Patients This peer-reviewed article was supported by Novo Nordisk Inc.; the com­
pany was provided with the opportunity to perform a medical accuracy
with Overweight or Obesity (SELECT) study (NCT03574597) will
review.
assess the effect of once-weekly subcutaneous semaglutide
2.4 mg treatment on CVD risk reduction in patients with over­
weight or obesity and established CVD without T2D. Declaration of financial/other relationships
Multiple agents for weight management, with varying
Anastassia Amaro: advisory boards and consultant – Medality Medical,
mechanisms of action, have been approved or are currently in
Novo Nordisk, and Pfizer, and research support – Altimmune, Eli Lilly,
clinical development. For example, tirzepatide (a novel glucose- Fractyl Health, and Novo Nordisk.
dependent insulinotropic polypeptide and GLP-1RA) resulted in Danny Sugimoto: grants – AstraZeneca, Boehringer Ingelheim, Bristol
substantial and sustained weight reduction in patients with Myers Squibb, Lilly, Merck, and Novo Nordisk.
16 A. AMARO ET AL.

Sean Wharton: research funding, advisory/consulting fees, and/or response and promoting macroautophagy. PLoS One. 2011;6(9):
other support – AstraZeneca, Bausch Health Inc., Boehringer Ingelheim, e25269.
CIHR, Janssen, Lilly, and Novo Nordisk. 19. Vrang N, Larsen PJ. Preproglucagon derived peptides GLP-1, GLP-2
Peer reviewers on this manuscript have no relevant financial or other and oxyntomodulin in the CNS: role of peripherally secreted and
relationships to disclose. centrally produced peptides. Prog Neurobiol. 2010;92(3):442–462.
20. Wang XC, Gusdon AM, Liu H, et al. Effects of glucagon-like peptide-1
receptor agonists on non-alcoholic fatty liver disease and
ORCID inflammation. World J Gastroenterol. 2014;20(40):14821–14830.
Anastassia Amaro http://orcid.org/0000-0002-3132-5778 21. Lee J, Hong S-W, Rhee E-J, et al. GLP-1 receptor agonist and
Danny Sugimoto http://orcid.org/0000-0002-7801-1231 non-alcoholic fatty liver disease. Diabetes Metab J. 2012;36
Sean Wharton http://orcid.org/0000-0003-0111-1530 (4):262–267.
22. Kanoski SE, Hayes MR, Skibicka KP. GLP-1 and weight loss: unravel­
ing the diverse neural circuitry. Am J Physiol Regul Integr Comp
Physiol. 2016;310(10):R885–R895.
References 23. Müller TD, Finan B, Bloom SR, et al. Glucagon-like peptide 1 (GLP-1).
Mol Metab. 2019;30:72–130.
1. Kyle TK, Dhurandhar EJ, Allison DB. Regarding obesity as a disease:
24. Drucker DJ. GLP-1 physiology informs the pharmacotherapy of
evolving policies and their implications. Endocrinol Metab Clin
obesity. Mol Metab. 2022;57:101351.
North Am. 2016;45(3):511–520.
25. Garber AJ. Long-acting glucagon-like peptide 1 receptor agonists:
2. Kushner RF, Calanna S, Davies M, et al. Semaglutide 2.4 mg for the
a review of their efficacy and tolerability. Diabetes Care. 2011;34
treatment of obesity: key elements of the STEP trials 1 to 5. Obesity
(Suppl 2):S279–S284.
(Silver Spring). 2020;28(6):1050–1061.
26. Pratley RE, Gilbert M. Targeting incretins in type 2 diabetes: role of
3. World Health Organization. Obesity and overweight. 2021 [cited
GLP-1 receptor agonists and DPP-4 inhibitors. Rev Diabet Stud.
2022 Jul 21]. Available from: https://www.who.int/news-room/fact-
2008;5(2):73–94.
sheets/detail/obesity-and-overweight
27. US Food and Drug Administration. Adlyxin highlights of prescribing
4. Hales CM, Carroll MD, Fryar CD, et al. Prevalence of obesity and
information. 2016 [cited 2022 Jul 21]. Available from: https://www.
severe obesity among adults: United States, 2017-2018. NCHS Data
accessdata.fda.gov/drugsatfda_docs/label/2016/
Brief. 2020;(360):1–8.
208471orig1s000lbl.pdf
5. Prospective Studies Collaboration, Whitlock G, Lewington S, et al.
Body-mass index and cause-specific mortality in 900 000 adults:
28. ®
US Food and Drug Administration. Byetta highlights of prescrib­
ing information. 2009 [cited 2022 Jul 21]. Available from: https://
collaborative analyses of 57 prospective studies. Lancet. 2009;373
www.accessdata.fda.gov/drugsatfda_docs/label/2009/
(9669):1083–1096.
021773s9s11s18s22s25lbl.pdf
6. Magkos F, Fraterrigo G, Yoshino J, et al. Effects of moderate and
subsequent progressive weight loss on metabolic function and adipose
29. ®
US Food and Drug Administration. Bydureon highlights of pre­
scribing information. 2018 [cited 2022 Jul 21]. Available from:
tissue biology in humans with obesity. Cell Metab. 2016;23(4):591–601.
https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/
7. Saunders KH, Umashanker D, Igel LI, et al. Obesity
022200s026lbl.pdf
pharmacotherapy. Med Clin North Am. 2018;102(1):135–148.
8. Garvey WT, Mechanick JI, Brett EM, et al. American Association of
30. ®
US Food and Drug Administration. Saxenda highlights of pre­
scribing information. 2014 [cited 2022 Jul 21]. Available from:
Clinical Endocrinologists and American College of Endocrinology
https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/
comprehensive clinical practice guidelines for medical care of
206321Orig1s000lbl.pdf
patients with obesity. Endocr Pract. 2016;22(Suppl 3):1–203.
31. US Food and Drug Administration. Tanzeum highlights of prescribing
9. Wharton S, Lau DCW, Vallis M, et al. Obesity in adults: a clinical
information. 2017 [cited 2022 Jul 21]. Available from: https://www.
practice guideline. CMAJ. 2020;192(31):E875–E891.
accessdata.fda.gov/drugsatfda_docs/label/2017/125431s019lbl.pdf
10. Rose F, Bloom S, Tan T. Novel approaches to anti-obesity drug
32. US Food and Drug Administration. Trulicity highlights of prescrib­
discovery with gut hormones over the past 10 years. Expert Opin
ing information. 2017 [cited 2022 Jul 21]. Available from: https://
Drug Discov. 2019;14(11):1151–1159.
www.accessdata.fda.gov/drugsatfda_docs/label/2017/
11. Husain M, Birkenfeld AL, Donsmark M, et al. Oral semaglutide and
125469s007s008lbl.pdf
cardiovascular outcomes in patients with type 2 diabetes. N Engl
33. US Food and Drug Administration. Wegovy highlights of prescribing
J Med. 2019;381(9):841–851.
information. 2021 [cited 2022 Jul 21]. Available from: https://www.
12. Marso SP, Bain SC, Consoli A, et al. Semaglutide and cardiovascular
outcomes in patients with type 2 diabetes. N Engl J Med. 2016;375 accessdata.fda.gov/drugsatfda_docs/label/2021/215256s000lbl.pdf
(19):1834–1844. ®
34. US Food and Drug Administration. Rybelsus highlights of pre­
13. Campbell JE, Drucker DJ. Pharmacology, physiology, and mechan­ scribing information. 2019 [cited 2022 Jul 21]. Available from:
isms of incretin hormone action. Cell Metab. 2013;17(6):819–837. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/
14. Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. 213051s000lbl.pdf
Gastroenterology. 2007;132(6):2131–2157. ®
35. US Food and Drug Administration. Ozempic highlights of pre­
scribing information. 2017 [cited 2022 Jul 21]. Available from:
15. Ban K, Noyan-Ashraf MH, Hoefer J, et al. Cardioprotective and vasodi­
latory actions of glucagon-like peptide 1 receptor are mediated https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/
through both glucagon-like peptide 1 receptor-dependent and - 209637lbl.pdf
independent pathways. Circulation. 2008;117(18):2340–2350. ®
36. US Food and Drug Administration. Victoza highlights of prescrib­
16. Merchenthaler I, Lane M, Shughrue P. Distribution of pre-pro- ing information. 2017 [cited 2022 Jul 21]. Available from: https://
glucagon and glucagon-like peptide-1 receptor messenger RNAs www.accessdata.fda.gov/drugsatfda_docs/label/2017/
in the rat central nervous system. J Comp Neurol. 1999;403 022341s027lbl.pdf
(2):261–280. 37. Kapitza C, Nosek L, Jensen L, et al. Semaglutide, a once-weekly
17. Pyke C, Heller RS, Kirk RK, et al. GLP-1 receptor localization in human GLP-1 analog, does not reduce the bioavailability of the
monkey and human tissue: novel distribution revealed with exten­ combined oral contraceptive, ethinylestradiol/levonorgestrel. J Clin
sively validated monoclonal antibody. Endocrinology. 2014;155 Pharmacol. 2015;55(5):497–504.
(4):1280–1290. 38. Lau J, Bloch P, Schäffer L, et al. Discovery of the once-weekly
18. Sharma S, Mells JE, Fu PP, et al. GLP-1 analogs reduce hepatocyte glucagon-like peptide-1 (GLP-1) analogue semaglutide. J Med
steatosis and improve survival by enhancing the unfolded protein Chem. 2015;58(18):7370–7380.
POSTGRADUATE MEDICINE 17

39. Marbury TC, Flint A, Jacobsen JB, et al. Pharmacokinetics and 58. Lando HM, Alattar M, Dua AP. Elevated amylase and lipase levels in
tolerability of a single dose of semaglutide, a human patients using glucagonlike peptide-1 receptor agonists or
glucagon-like peptide-1 analog, in subjects with and without dipeptidyl-peptidase-4 inhibitors in the outpatient setting. Endocr
renal impairment. Clin Pharmacokinet. 2017;56(11):1381–1390. Pract. 2012;18(4):472–477.
40. Kalra S, Sahay R. A review on semaglutide: an oral glucagon-like 59. US Food and Drug Administration. Nesina highlights of prescribing
peptide 1 receptor agonist in management of type 2 diabetes information. 2015 [cited 2022 Jul 21]. Available from: https://www.
mellitus. Diabetes Ther. 2020;11(9):1965–1982. accessdata.fda.gov/drugsatfda_docs/label/2015/022271s007lbl.pdf
41. National Center for Biotechnology Information. PubChem com­
pound summary for CID 56843331, semaglutide. 2022 [cited 2022
®
60. US Food and Drug Administration. Onglyza highlights of prescrib­
ing information. 2015 [cited 2022 Jul 21]. Available from: https://
Jul 21]. Available from: https://pubchem.ncbi.nlm.nih.gov/com www.accessdata.fda.gov/drugsatfda_docs/label/2017/
pound/Semaglutide 022350s018lbl.pdf
42. Blundell J, Finlayson G, Axelsen M, et al. Effects of once-weekly
semaglutide on appetite, energy intake, control of eating, food
®
61. US Food and Drug Administration. Januvia highlights of prescrib­
ing information. 2011 [cited 2022 Jul 21]. Available from: https://
preference and body weight in subjects with obesity. Diabetes www.accessdata.fda.gov/drugsatfda_docs/label/2012/
Obes Metab. 2017;19(9):1242–1251. 021995s019lbl.pdf
43. Friedrichsen M, Breitschaft A, Tadayon S, et al. The effect of sema­ 62. Garber AJ, Handelsman Y, Grunberger G, et al. Consensus state­
glutide 2.4 mg once weekly on energy intake, appetite, control of ment by the American Association of Clinical Endocrinologists and
eating, and gastric emptying in adults with obesity. Diabetes Obes American College of Endocrinology on the comprehensive type 2
Metab. 2021;23(3):754–762. diabetes management algorithm - 2020 executive summary.
44. Davies M, Færch L, Jeppesen OK, et al. Semaglutide 2·4 mg Endocr Pract. 2020;26(1):107–139.
once a week in adults with overweight or obesity, and type 2 63. Erlinger S. Gallstones in obesity and weight loss. Eur
diabetes (STEP 2): a randomised, double-blind, double-dummy, J Gastroenterol Hepatol. 2000;12(12):1347–1352.
placebo-controlled, phase 3 trial. Lancet. 2021;397 64. Nauck MA, Muus Ghorbani ML, Kreiner E, et al. Effects of liraglutide
(10278):971–984. compared with placebo on events of acute gallbladder or biliary
45. Rubino D, Abrahamsson N, Davies M, et al. Effect of continued disease in patients with type 2 diabetes at high risk for cardiovas­
weekly subcutaneous semaglutide vs placebo on weight loss main­ cular events in the LEADER randomized trial. Diabetes Care.
tenance in adults with overweight or obesity: the STEP 4 rando­ 2019;42(10):1912–1920.
mized clinical trial. JAMA. 2021;325(14):1414–1425. 65. He L, Wang J, Ping F, et al. Association of glucagon-like peptide-1
46. Wadden TA, Bailey TS, Billings LK, et al. Effect of subcutaneous receptor agonist use with risk of gallbladder and biliary diseases:
semaglutide vs placebo as an adjunct to intensive behavioral a systematic review and meta-analysis of randomized clinical trials.
therapy on body weight in adults with overweight or obesity: the JAMA Intern Med. 2022;182(5):513–519.
STEP 3 randomized clinical trial. JAMA. 2021;325(14):1403–1413. 66. Woronow D, Chamberlain C, Niak A, et al. Acute cholecystitis
47. Wilding JPH, Batterham RL, Calanna S, et al. Once-weekly semaglutide in associated with the use of glucagon-like peptide-1 receptor ago­
adults with overweight or obesity. N Engl J Med. 2021;384(11):989– nists reported to the US Food and Drug Administration. JAMA
1002. Intern Med. 2022;182(10):1104–1106.
48. Garvey WT, Batterham RL, Bhatt M, et al. Two-year effects of 67. Faillie JL, Yu OH, Yin H, et al. Association of bile duct and gallbladder
semaglutide in adults with overweight or obesity: the STEP 5 trial. diseases with the use of incretin-based drugs in patients with type 2
Nat Med. 2022;28(10):2083–2091. diabetes mellitus. JAMA Intern Med. 2016;176(10):1474–1481.
49. American Heart Association. AHA names top heart disease and 68. Cote GJ, Grubbs EG, Hofmann MC. Thyroid C-cell biology and
stroke research advances of 2021. 2021 [cited 2022 July 21]. oncogenic transformation. Recent Results Cancer Res.
Available from: https://www.heart.org/en/around-the-aha/aha- 2015;204:1–39.
names-top-heart-disease-and-stroke-research-advances-of-2021 69. American Thyroid Association. Medullary thyroid cancer. 2020
50. Wilding JPH, Batterham RL, Davies M, et al. Weight regain and [cited 2022 Jul 21]. Available from: https://www.thyroid.org/wp-
cardiometabolic effects after withdrawal of semaglutide: the STEP content/uploads/patients/brochures/medullary-thyroid-cancer-
1 trial extension. Diabetes Obes Metab. 2022;24(8):1553–1564. brochure.pdf
51. Wharton S, Astrup A, Endahl L, et al. Estimating and reporting 70. Abd El Aziz M, Cahyadi O, Meier JJ, et al. Incretin-based
treatment effects in clinical trials for weight management: using glucose-lowering medications and the risk of acute pancreatitis
estimands to interpret effects of intercurrent events and missing and malignancies: a meta-analysis based on cardiovascular out­
data. Int J Obes (Lond). 2021;45(5):923–933. comes trials. Diabetes Obes Metab. 2020;22(4):699–704.
52. Trujillo J. Safety and tolerability of once-weekly GLP-1 receptor agonists 71. Bray GA, Frühbeck G, Ryan DH, et al. Management of obesity.
in type 2 diabetes. J Clin Pharm Ther. 2020;45(Suppl 1):43–60. Lancet. 2016;387(10031):1947–1956.
53. Wharton S, Davies M, Dicker D, et al. Managing the gastrointestinal 72. Jastreboff AM, Aronne LJ, Ahmad NN, et al. Tirzepatide once weekly
side effects of GLP-1 receptor agonists in obesity: recommenda­ for the treatment of obesity. N Engl J Med. 2022;387(3):205–216.
tions for clinical practice. Postgrad Med. 2022;134(1):14–19. 73. Rubino DM, Greenway FL, Khalid U, et al. Effect of weekly subcu­
54. Nauck MA, Friedrich N. Do GLP-1-based therapies increase cancer taneous semaglutide vs daily liraglutide on body weight in adults
risk? Diabetes Care. 2013;36(Suppl 2):S245–S252.
with overweight or obesity without diabetes: the STEP 8 rando­
55. Roshanov PS, Dennis BB. Incretin-based therapies are associated
mized clinical trial. JAMA. 2022;327(2):138–150.
with acute pancreatitis: meta-analysis of large randomized con­
74. Arterburn DE, Telem DA, Kushner RF, et al. Benefits and risks of
trolled trials. Diabetes Res Clin Pract. 2015;110(3):e13–e17.
bariatric surgery in adults: a review. JAMA. 2020;324(9):879–887.
56. Nreu B, Dicembrini I, Tinti F, et al. Pancreatitis and pancreatic
cancer in patients with type 2 diabetes treated with glucagon-like 75. Schauer PR, Bhatt DL, Kirwan JP, et al. Bariatric surgery versus
peptide-1 receptor agonists: an updated meta-analysis of rando­ intensive medical therapy for diabetes–3-year outcomes. N Engl
mized controlled trials. Minerva Endocrinol. 2020;Online ahead of J Med. 2014;370(21):2002–2013.
print. DOI:10.23736/S0391-1977.20.03219-8. 76. Maciejewski ML, Arterburn DE, Van Scoyoc L, et al. Bariatric surgery
57. Steinberg WM, Rosenstock J, Wadden TA, et al. Impact of liraglutide and long-term durability of weight loss. JAMA Surg. 2016;151
on amylase, lipase, and acute pancreatitis in participants with over­ (11):1046–1055.
weight/obesity and normoglycemia, prediabetes, or type 2 dia­ 77. Schauer PR, Kashyap SR, Wolski K, et al. Bariatric surgery versus
betes: secondary analyses of pooled data from the SCALE clinical intensive medical therapy in obese patients with diabetes. N Engl
development program. Diabetes Care. 2017;40(7):839–848. J Med. 2012;366(17):1567–1576.

You might also like