ArticlePDF AvailableLiterature Review

Use of GLP-1 Receptor Agonists and Occurrence of Thyroid Disorders: a Meta-Analysis of Randomized Controlled Trials

Frontiers
Frontiers in Endocrinology
Authors:

Abstract and Figures

The association between glucagon-like peptide-1 (GLP-1) receptor agonists and the risk of various kinds of thyroid disorders remains uncertain. We aimed to evaluate the relationship between the use of GLP-1 receptor agonists and the occurrence of 6 kinds of thyroid disorders. We searched PubMed (MEDLINE), EMBASE, the Cochrane Central Register of Controlled Trials (CENTRAL) and Web of Science from database inception to 31 October 2021 to identify eligible randomized controlled trials (RCTs). We performed meta-analysis using a random-effects model to calculate risk ratios (RRs) and 95% confidence intervals (CIs). A total of 45 trials were included in the meta-analysis. Compared with placebo or other interventions, GLP-1 receptor agonists’ use showed an association with an increased risk of overall thyroid disorders (RR 1.28, 95% CI 1.03-1.60). However, GLP-1 receptor agonists had no significant effects on the occurrence of thyroid cancer (RR 1.30, 95% CI 0.86-1.97), hyperthyroidism (RR 1.19, 95% CI 0.61-2.35), hypothyroidism (RR 1.22, 95% CI 0.80-1.87), thyroiditis (RR 1.83, 95% CI 0.51-6.57), thyroid mass (RR 1.17, 95% CI 0.43-3.20), and goiter (RR 1.17, 95% CI 0.74-1.86). Subgroup analyses and meta-regression analyses showed that underlying diseases, type of control, and trial durations were not related to the effect of GLP-1 receptor agonists on overall thyroid disorders (all P subgroup > 0.05). In conclusion, GLP-1 receptor agonists did not increase or decrease the risk of thyroid cancer, hyperthyroidism, hypothyroidism, thyroiditis, thyroid mass and goiter. However, due to the low incidence of these diseases, these findings need to be examined further. Systematic Review Registration PROSPERO https://www.crd.york.ac.uk/prospero/, identifier: CRD42021289121.
This content is subject to copyright.
Use of GLP-1 Receptor Agonists and
Occurrence of Thyroid Disorders:
a Meta-Analysis of Randomized
Controlled Trials
Weiting Hu
1
, Rui Song
1
, Rui Cheng
2
, Caihong Liu
3
, Rui Guo
3
, Wei Tang
2
, Jie Zhang
2
,
Qian Zhao
2
, Xing Li
2
*and Jing Liu
2
*
1
The Second Clinical Medical College, Shanxi Medical University, Taiyuan, China,
2
Department of Endocrinology, Second
Hospital of Shanxi Medical University, Taiyuan, China,
3
Department of Physiology, Shanxi Medical University, Taiyuan, China
The association between glucagon-like peptide-1 (GLP-1) receptor agonists and the risk
of various kinds of thyroid disorders remains uncertain. We aimed to evaluate the
relationship between the use of GLP-1 receptor agonists and the occurrence of 6 kinds
of thyroid disorders. We searched PubMed (MEDLINE), EMBASE, the Cochrane Central
Register of Controlled Trials (CENTRAL) and Web of Science from database inception to
31 October 2021 to identify eligible randomized controlled trials (RCTs). We performed
meta-analysis using a random-effects model to calculate risk ratios (RRs) and 95%
condence intervals (CIs). A total of 45 trials were included in the meta-analysis.
Compared with placebo or other interventions, GLP-1 receptor agonistsuse showed
an association with an increased risk of overall thyroid disorders (RR 1.28, 95% CI 1.03-
1.60). However, GLP-1 receptor agonists had no signicant effects on the occurrence of
thyroid cancer (RR 1.30, 95% CI 0.86-1.97), hyperthyroidism (RR 1.19, 95% CI 0.61-
2.35), hypothyroidism (RR 1.22, 95% CI 0.80-1.87), thyroiditis (RR 1.83, 95% CI 0.51-
6.57), thyroid mass (RR 1.17, 95% CI 0.43-3.20), and goiter (RR 1.17, 95% CI 0.74-1.86).
Subgroup analyses and meta-regression analyses showed that underlying diseases, type
of control, and trial durations were not related to the effect of GLP-1 receptor agonists on
overall thyroid disorders (all P
subgroup
> 0.05). In conclusion, GLP-1 receptor agonists did
not increase or decrease the risk of thyroid cancer, hyperthyroidism, hypothyroidism,
thyroiditis, thyroid mass and goiter. However, due to the low incidence of these diseases,
these ndings need to be examined further.
Systematic Review Registration: PROSPERO https://www.crd.york.ac.uk/prospero/,
identier: CRD42021289121.
Keywords: GLP-1 receptor agonists, thyroid disorders, thyroid cancer, meta-analysis, randomized controlled trials
Frontiers in Endocrinology | www.frontiersin.org July 2022 | Volume 13 | Article 9278591
Edited by:
Johannes Wolfgang Dietrich,
Ruhr University Bochum, Germany
Reviewed by:
Daniel Quast,
St. Josef-Hospital Bochum, Germany
Simone Wajner,
Federal University of Rio Grande do
Sul, Brazil
Stephen Bain,
Swansea University, United Kingdom
Trevor Edmund Angell,
University of Southern California,
United States
*Correspondence:
Jing Liu
13934663905@163.com
Xing Li
13503504180@163.com
Specialty section:
This article was submitted to
Thyroid Endocrinology,
a section of the journal
Frontiers in Endocrinology
Received: 25 April 2022
Accepted: 13 June 2022
Published: 11 July 2022
Citation:
Hu W, Song R, Cheng R, Liu C, Guo R,
Tang W, Zhang J, Zhao Q, Li X and
Liu J (2022) Use of GLP-1 Receptor
Agonists and Occurrence of Thyroid
Disorders: a Meta-Analysis of
Randomized Controlled Trials.
Front. Endocrinol. 13:927859.
doi: 10.3389/fendo.2022.927859
SYSTEMATIC REVIEW
published: 11 July 2022
doi: 10.3389/fendo.2022.927859
INTRODUCTION
Thyroid diseases are common in some metabolic disorders, such
as diabetes mellitus (DM) and obesity. Thyroid dysfunction (TD)
and DM are closely linked. A high prevalence of TD has been
reported among both type 1 DM (T1DM) and type 2 DM
(T2DM) patients (1,2). Although the mechanism is unknown,
epidemiological studies have indicated that obesity and T2DM
are associated with increased risks of several cancers, including
thyroid cancer (35). Furthermore, insulin resistance and
hyperinsulinemia can lead to goiter, proliferation of thyroid
tissues, and an increased incidence of nodular thyroid disease
(6). In addition to the effects of the disease itself, some
antidiabetic drugs can impact the hypothalamicpituitary
thyroid (HPT) axis and thyroid function. For example,
multiple studies have demonstrated that metformin can inhibit
the growth of thyroid cells and different types of thyroid cancer
cells, and metformin therapy has been associated with a decrease
in the levels of serum thyroid-stimulating hormone (TSH) (7).
Thiazolidinediones can induce thyroid-associated
ophthalmopathy (8,9). Recently, the relationship between
glucagon-like peptide-1 (GLP-1) receptor agonists and thyroid
cancer has attracted attention, but there is still controversy.
GLP-1 is an amino acid peptide hormone secreted by L cells
of the gastrointestinal mucosa that promotes insulin secretion,
suppresses glucagon secretion, and delays gastric emptying (10).
Rodent studies have shown that the GLP-1 receptor agonist
liraglutide can activate the GLP-1 receptor on thyroid C cells,
leading to the release of calcitonin with a dose-dependent effect
on the pathology of C cells (11). Some animal models have
proven that exenatide or liraglutide treatment is related to the
abnormal appearance of thyroid C cells, with gradual
development of hyperplasia and adenomas (12,13). Moreover,
a study found that patients treated with exenatide had an
increased risk of thyroid cancer by examining the US Food
and Drug Administrations database of reported adverse events
(14). However, the results of A Long Term Evaluation
(LEADER) trial that followed for 3.5-5 years showed no effect
of GLP-1 receptor activation on human serum calcitonin levels,
C-cell proliferation or C-cell malignancy (15). Nevertheless,
GLP-1 receptor agonists are not recommended in patients with
a personal or family history of medullary thyroid cancer or type 2
multiple endocrine neoplasia.
GLP-1 receptor agonists, a new type of antidiabetic drug for
treating T2DM in recent years, with additional benets of weight
loss and blood pressure reduction (16). Although many large
randomized controlled trials (RCTs) of GLP-1 receptor agonists
have identied the obvious benets of GLP-1 receptor agonists
on cardiovascular and renal outcomes in patients with DM or
obesity (1720), the association between GLP-1 receptor agonists
and various thyroid disorders remains controversial. In addition,
considering that thyroid disorders are common in some
metabolic diseases such as DM and obesity, we conducted this
study. Thus, by comparing GLP-1 receptor agonists with placebo
or other antidiabetic drugs, we conducted a meta-analysis of all
available RCT data to evaluate the relationship between the use
of GLP-1 receptor agonists and the occurrence of various kinds
of thyroid disorders.
METHODS
Data Sources and Searches
We searched PubMed (MEDLINE), EMBASE, Cochrane Central
Register of Controlled Trials (CENTRAL) and Web of Science
from database inception to 31 October 2021 to identify eligible
RCTs without restriction of language or publication period. The
search terms used were glucagon-like peptide 1 receptor
agonist,exenatide,liraglutide,dulaglutide,lixisenatide,
semaglutide,albiglutide,taspoglutide,loxenatide,
diabetes mellitus,obesityand randomized controlled
trial. In addition, we manually scanned the ClinicalTrials.gov
web and reference lists from established trials and review articles.
Study Selection
The trials we included met the following criteria: (1) RCTs that
compared GLP-1 receptor agonist with a placebo or active control
(other antidiabetic drugs or insulin), (2) patients with type 2
diabetes, type 1 diabetes, prediabetes, overweight or obesity, (3)
with durations of at least 24 weeks,and (4) reported the occurrence
of at least one case of various thyroid disorders as adverse events.
We excluded duplicate reports, conference abstracts, letters, case
reports, editorials, articles without treatment-emergent adverse
events, and animal experimental studies.
Data Extraction and Quality Assessment
Two investigators (Hu and Song) independently extracted the
following data by reviewing the full text of each study: rst
author, year of publication, Clinical Trial Registration Number
(NCT ID), trial duration, patient characteristics, sample size,
intervention (type of GLP-1 receptor agonist), comparators, and
outcomes of interest. Any discrepancies were resolved by
consensus or by the third reviewer (Chen). The primary
outcome was the incidence of overall thyroid disorders, and
the secondary outcomes included the incidence of goiter,
hyperthyroidism, hypothyroidism, thyroiditis, thyroid mass,
and thyroid cancer. When multiple reports from the same
population were retrieved, the most complete or recently
reported data were used. If thyroid-related events were not
reported in publication, these data were extracted from the
Serious Adverse Eventsportion of ClinicalTrials.gov.
The quality of each included RCT was assessed by the
Cochrane Risk-of-Bias Tool 1.0. The Jadad scale was also used
to quantify the study quality. Two authors assessed the risk of
bias for each study through ve aspects: random sequence
generation, allocation concealment, blinding, incomplete
outcome data and selective reporting.
Statistical Analysis
Dichotomous outcomes were analyzed by risk ratios (RRs) and
95% condence intervals (CIs) using the DerSimonian and Laird
random-effects model. We assessed heterogeneity between the
Hu et al. GLP-1RAs and Thyroid Disorders
Frontiers in Endocrinology | www.frontiersin.org July 2022 | Volume 13 | Article 9278592
included studies using the statistic, where I
2
values of 25%,
50%, and 75% indicated low, medium, and high heterogeneity,
respectively. Subgroup analyses were conducted according to the
type of underlying diseases, type of control, and trial duration.
Between-subgroup heterogeneity was assessed by c
2
tests and
meta-regression. All of the above analyses were performed using
Stata software 13.0 (Stata Corp). A p value < 0.05 was considered
statistically signicant.
RESULT
Study Search and Study Characteristics
A total of 16,201 records were identied by retrieving the
aforementioned databases. Excluding duplicates and reviewing
titles and abstracts, 301 studies were read the full text. After
retrieving the full text and searching on ClinicalTrials.gov, the
nal analysis included 45 RCTs reported in 45 publications with
94063 participants (1761). Although the data from the two articles
were presented together on ClinicalTrials.gov (62), due to the
differences in population characteristics and follow-up time, we
considered them separately and regarded them as two independent
trials (24,25). The search and selection process is summarized in
Figure 1.The characteristicsof these includedstudies are detailedin
Table 1 and Table S1. Across the 45 trials, trial duration ranged
from 26 to 360 weeks. Of all the participants, 29,348 (55.8%) were
men in the experimental group, and 24121 (58.2%) were men in the
control group. The mean age of study participants ranged from41.6
to 66.2 yearsold in experimentalgroups and 41.4 to 66.2 years old in
control groups. Mean patient body mass index (BMI) ranged from
24.5 to 39.3kg/m2 in experimental groups and24.4 to 39.0 kg/m2 in
control groups.
Risk of Bias Evaluation
The studies included in this analysis provide information about
random sequence generation, allocation concealment,
participant blindness, personnel, outcome evaluation and
selective reporting. Figure S1 reports the risk details of
deviation assessment. (Figure S1 in Appendix) 29 trials had a
Jadad scale of 4 or 5, and others were scored 3.
Incidence of Thyroid Disorders With All
GLP-1 Receptor Agonists
As is shown in Figure 2, this meta-analysis included 52600
patients in the GLP-1 receptor agonist group and 41463 patients
in the control group. The event rate in the GLP-1 receptor
agonist group (0.39%) was higher than in the control group
(0.31%). Compared with placebo or other interventions, GLP-1
receptor agonist increased the risk of overall thyroid disorders by
28% (RR 1.28, 95% CI 1.03-1.60; p = 0.027), with no statistically
signicant between-study heterogeneity (I
2
= 0.0%). The funnel
plot for this analysis indicated no signicant publication bias
(Figure S2).
GLP-1 receptor agonists versus placebo or other interventions
had no signicant effects on the occurrence of thyroid cancer (RR
1.30, 95% CI 0.86-1.97, p = 0.212; I
2
=0.0%;Figure S3),
hyperthyroidism (RR 1.19, 95% CI 0.61-2.35, p = 0.608; I
2
=
0.0%; Figure S4), hypothyroidism (RR 1.22, 95% CI 0.80-1.87, p =
0.359; I
2
= 0.0%; Figure S5), thyroiditis (RR 1.83, 95% CI 0.51-
6.57, p = 0.353; I
2
= 0.0%; Figure S6), thyroid mass (RR 1.17, 95%
CI 0.43-3.20, p = 0.759; I
2
= 0.0%; Figure S7), and goiter (RR 1.17,
95% CI 0.74-1.86, p = 0.503; I
2
= 0.0%; Figure S8).
Incidence of Thyroid Disorders With
Different GLP-1 Receptor Agonists
Among all 45 enrolled trials, 18 trials including 24787 patients
used liraglutide as the experimental agent. Compared with
placebo or other interventions, treatment with liraglutide
increased the risk of overall thyroid disorders by 37% (RR
1.37, 95% CI 1.01-1.86, p = 0.044; Figure 3), and no
statistically signicant between-study heterogeneity was
observed (I
2
= 0.0%, p = 0.933).
Moreover, another 5 trials including 13281 patients provided
information about the risk of thyroid disorders in patients
treated with dulaglutide. This result showed that compared
with placebo or other interventions, dulaglutide signicantly
increased the incidence of overall thyroid disorders by 96%
(RR 1.96, 95% CI 1.11-3.45, p = 0.020; Figure 3), and no
statistically signicant between-study heterogeneity was
observed (I
2
= 0.0%, p = 0.965).
However, no effect against overall thyroid disorders was
found for other GLP-1 receptor agonists. There were 11 studies
including 15401 patients that regarded semaglutide as the
experimental agent, and the pooled RR of overall thyroid
disorders in patients receiving semaglutide versus other
interventions was 0.75 (95% CI 0.351.57; Figure 3). Whether
oral semaglutide or subcutaneous semaglutide, the results
showed that they had no signicant effects on the occurrence
of overall thyroid disorders (Figure S9 and Figure S10). There
were 5 studies including 8895 patients that regarded lixisenatide
as the experimental agent, and the pooled RR of overall thyroid
disorders in patients receiving lixisenatide versus other
FIGURE 1 | Summary of trial selection.
Hu et al. GLP-1RAs and Thyroid Disorders
Frontiers in Endocrinology | www.frontiersin.org July 2022 | Volume 13 | Article 9278593
TABLE 1 | Baseline characteristics of included studies.
Study Clinical Trial
Registration
Number
Trial Duration
(week)
Interventions Events/Patients (N) Age (years) Man (N, %) BMI (kg/m
2
) Jadad
score
Experimental Control Experimental Control Experimental Control Experimental Control Experimental Control
Unger et al., 2022 (21) NCT02730377 105 Liraglutide OAD 1/996 0/995 57.6 (11.0) 57.1
(10.7)
520 (52.2) 524
(52.7)
33.2 (7.2) 33.7
(7.6)
2
Garveyet al 2020 (22) NCT02963922 60 Liraglutide Placebo 1/198 0/198 55.9 (11.3) 57.6
(10.4)
90 (45.5) 99 (50.0) 35.9 (6.5) 35.3
(5.8)
4
Wadden et al., 2020
(23)
NCT02963935 60 Liraglutide Placebo 1/142 0/140 45.4 (11.6) 49.0
(11.2)
23 (16.2) 24 (17.1) 39.3 (6.8) 38.7
(7.2)
4
le et al., 2017 (24) NCT01272219 172 Liraglutide Placebo 3/1505 3/749 47.5 (11.7) 47.3
(11.8)
364 (24.0) 176
(23.0)
38.8 (6.4) 39.0
(6.3)
4
Pi-Sunyer et al., 2015
(25)
NCT01272219 68 Liraglutide Placebo 1/959 0/487 41.6 (11.7) 41.5
(11.5)
158 (16.5) 97 (19.9) 37.5 (6.2) 37.4
(6.2)
4
Zang et al., 2016 (26) NCT02008682 26 Liraglutide Sitagliptin 0/183 1/184 51.7 (10.7) 51.4
(11.0)
102 (55.7) 117
(63.6)
27.3 (3.4) 27.2
(4.0)
2
Ahren et al., 2016 (27) NCT02098395 26 Liraglutide Placebo 2/625 0/206 43.3 42.7 288 (46.1) 94 (45.6) 28.9 28.9 4
Mathieu et al., 2016
(28)
NCT01836523 52 Liraglutide Placebo 0/1042 1/347 43.7 43.4 496 (47.6) 167
(48.1)
29.4 29.8 4
Marso et al., 2016
(20)
NCT01179048 240 Liraglutide Placebo 77/4668 54/
4672
64.2 (7.2) 64.4
(7.2)
3011 (64.5) 2992
(64.0)
32.5 (6.3) 32.5
(6.3)
4
Davies et al., 2015
(29)
NCT01272232 68 Liraglutide Placebo 1/634 1/212 55.0 54.7 328 (51.7) 97 (45.8) 37.1 37.4 4
Gough et al., 2014
(30)
NCT01336023 52 Liraglutide
IDegLira
Degludec 2/414
0/833
0/413 55.0 (10.2)
55.1 (9.9)
54.9
(9.7)
208 (50.2)
435 (52.2)
200
(48.4)
31.3 (4.8)
31.2 (5.2)
31.2
(5.3)
3
Wadden et al., 2013
(31)
NCT00781937 56 Liraglutide Placebo 3/212 0/210 45.9 (11.9) 46.5
(11.0)
34 (16.0) 45 (21.4) 38.2 (6.2) 37.5
(6.2)
4
Seino et al., 2010 (32) NCT00393718 52 Liraglutide Glibenclamide 1/268 0/132 58.2 (10.4) 58.5
(10.4)
183 (68.3) 86 (65.2) 24.5 (3.7) 24.4
(3.8)
4
Pratley et al., 2010
(33)
NCT00700817 78 Liraglutide Sitagliptin 1/446 0/219 55.5 55.0 232 (52.0) 120
(55.0)
32.9 32.6 2
Nauck et al., 2009
(34)
NCT00318461 104 Liraglutide Glibenclamide
Placebo
6/724 2/242
0/121
56.7 57.3
56.0
422 (58.3) 139
(57.4)
72 (59.5)
30.8 31.2
31.6
4
Garber et al., 2009
(35)
NCT00294723 104 Liraglutide Glibenclamide 6/498 0/248 52.9 53.4 238 (47.8) 133
(53.6)
33.0 33.2 3
Hernandez et al.,
2018 (36)
NCT02465515 130 Albiglutide Placebo 0/4731 1/4732 64.1 (8.7) 64.2
(8.7)
3304 (70.0) 3265
(69.0)
32.3 (5.9) 32.3
(5.9)
5
Home et al., 2015 (37) NCT00839527 52 Albiglutide Pioglitazone
Placebo
5/271 9/277
2/115
54.5 (9.5) 55.7
(9.4)
55.7
(9.6)
135 (49.8) 148
(53.4)
70 (60.9)
32.4 (5.5) 32.2
(5.7)
31.8
(4.9)
3
Ahren et al., 2014 (38) NCT00838903 164 Albiglutide Sitagliptin
Glibenclamide
Placebo
1/302 2/302
0/307
0/101
54.3 (10.1) 54.3
(9.8)
54.4
(10.0)
56.1
(10.0)
135 (44.7) 139
(46.0)
158
(51.5)
50 (49.5)
32.7 (5.6) 32.5
(5.4)
32.5
(5.5)
32.8
(5.4)
2
Leiter et al., 2014 (19) NCT01098539 60 Albiglutide Sitagliptin 1/249 0/246 63.2 (8.4) 63.5
(9.0)
136 (54.6) 130
(52.8)
30.4 (5.5) 30.4
(5.8)
4
(Continued)
Hu et al. GLP-1RAs and Thyroid Disorders
Frontiers in Endocrinology | www.frontiersin.org July 2022 | Volume 13 | Article 9278594
TABLE 1 | Continued
Study Clinical Trial
Registration
Number
Trial Duration
(week)
Interventions Events/Patients (N) Age (years) Man (N, %) BMI (kg/m
2
) Jadad
score
Experimental Control Experimental Control Experimental Control Experimental Control Experimental Control
Holman et al., 2017
(18)
NCT01144338 360 Exenatide Placebo 23/7356 16/
7396
61.8 (9.4) 61.9
(9.4)
4562 (62) 4587(62) 31.8 31.7 5
Gallwitz et al., 2012
(39)
NCT00359762 216 Exenatide Glimepiride 0/490 4/487 56.0 (10.0) 56.0
(9.1)
272 (55.5) 252
(51.7)
32.6 (4.2) 32.3
(3.9)
2
Bergenstal et al., 2010
(40)
NCT00637273 26 Exenatide Sitagliptin
Pioglitazone
0/160 1/166
0/165
52.4 (10.4) 52.2
(10.5)
53.0
(9.9)
89 (55.6) 86 (51.8)
79 (47.9)
32.0 (5.0) 32.0
(5.0)
32.0
(6.0)
3
Wang et al., 2019 (41) NCT01648582 56 Dulaglutide Glargine 8/505 2/250 54.8 55.4 278 (55.0) 139
(55.6)
26.8 26.7 2
Gerstein et al., 2019
(42)
NCT01394952 336 Dulaglutide Placebo 26/4949 14/
4952
66.2 (6.5) 66.2
(6.5)
2643 (53·4) 2669
(53·9)
32.3 (5.7) 32.3
(5.8)
5
Chen et al., 2018 (43) NCT01644500 26 Dulaglutide Glimepiride 2/478 0/242 53.2 52.0 261 (54.6) 130
(53.7)
26.0 25.7 4
Weinstock et al., 2015
(44)
NCT00734474 104 Dulaglutide Sitagliptin
Placebo
3/606 0/315
0/177
54.0 54.0
55.0
280 (46.2) 151
(48.0)
90 (51.0)
31.0 31.0
31.0
5
Giorgino et al., 2015
(45)
NCT01075282 78 Dulaglutide Glargine 1/545 0/262 56.5 57.0 280 (51.4) 134
(51.0)
31.5 32.0 2
Rosenstock et al.,
2016 (46)
NCT02058147 30 Lixisenatide
iGlarLixi
Glargine 0/469
0/234
1/467 58.7 (8.7)
58.2 (9.5)
58.3
(9.4)
133 (56.8)
222 (47.3)
237
(50.7)
32.0 (4.4)
31.6 (4.4)
31.7
(4.5)
2
Pfeffer et al., 2015 (47) NCT01147250 225 Lixisenatide Placebo 2/3034 3/3034 59.9 (9.7) 60.6
(9.6)
2111 (69.6) 2096
(69.1)
30.1 (5.6) 30.2
(5.8)
5
Bolli et al., 2014 (48) NCT00763451 112 Lixisenatide Placebo 2/322 0/160 55.0 58.2 143 (44.4) 72 (45.0) 32.6 32.4 5
Ahren et al., 2013 (49) NCT00712673 76 Lixisenatide Placebo 1/510 1/170 54.7 55.0 212 (41.6) 81 (47.6) 32.9 33.1 4
Riddle et al., 2013
(50)
NCT00715624 125 Lixisenatide Placebo 1/328 0/167 57.4 (9.5) 56.9
(9.8)
146 (44.5) 82 (49.1) 31.9 (6.2) 32.6
(6.3)
5
Wilding et al., 2021
(51)
NCT03548935 75 Semaglutide Placebo 1/1306 0/655 46.0 (13.0) 47.0
(12.0)
351 (26.9) 157
(24.0)
37.8 (6.7) 38.0
(6.5)
4
Wadden et al., 2021
(52)
NCT03611582 75 Semaglutide Placebo 1/407 0/204 46.0 (13.0) 46.0
(13.0)
92 (22.6) 24 (11.8) 38.1 (6.7) 37.8
(6.9)
5
Yamada et al., 2020
(53)
NCT03018028 57 Semaglutide
Liraglutide
Placebo 1/146
0/48
0/49 59.7
59.0
59.0 112 (76.7)
39 (81.3)
40 (81.6) 25.8
26.9
25.1 5
Husain et al., 2019
(54)
NCT02692716 87 Semaglutide Placebo 2/1591 2/1592 66.0 (7.0) 66.0
(7.0)
1084 (68.1) 1092
(68.6)
32.3 (6.6) 32.3
(6.4)
5
Rosenstock et al.,
2019 (55)
NCT02607865 83 Semaglutide Sitagliptin 0/1396 1/467 58.0 58.0 746 (53.4) 238
(51.0)
32.5 32.5 3
Pratley et al., 2019
(56)
NCT02863419 57 Semaglutide
Liraglutide
Placebo 2/285
1/284
0/142 56.0 (10.0)
56.0 (10.0)
57.0
(10.0)
147 (51.6)
149 (52.5)
74 (52.1) 32.5 (5.9)
33.4 (6.7)
32.9
(6.1)
4
Aroda et al., 2019 (57) NCT02906930 31 Semaglutide Placebo 2/525 0/178 55.0 54.0 268 (51.0) 89 (50.0) 31.7 32.2 3
ONeil et al., 2018 (58) NCT02453711 59 Semaglutide
Liraglutide
Placebo 0/718
0/103
1/136 46.3
49.0
46.0 254 (35.4)
36 (35.0)
48 (35.0) 30.0
30.4
30.7 3
Ahren et al., 2017 (59) NCT01930188 56 Semaglutide Sitagliptin 3/818 0/407 55.4 54.6 412 (50.3) 208
(51.1)
32.5 32.5 4
Aroda et al., 2017 (60) NCT02128932 36 Semaglutide Glargine 0/722 1/360 56.6 56.2 379 (52.5) 195 (54) 33.1 33.0 3
(Continued)
Hu et al. GLP-1RAs and Thyroid Disorders
Frontiers in Endocrinology | www.frontiersin.org July 2022 | Volume 13 | Article 9278595
interventions was 0.69 (95% CI 0.222.20; Figure 3). There were
3 studies including 16220 patients that regarded exenatide as the
experimental agent, and the pooled RR of overall thyroid
disorders in patients receiving exenatide versus other
interventions was 0.82 (95% CI 0.213.29; Figure 3). There
were 3 studies including 11633 patients that regarded
albiglutide as the experimental agent, and the pooled RR of
overall thyroid disorders in patients receiving albiglutide versus
other interventions was 0.76 (95% CI 0.311.83; Figure 3). Most
of the above meta-analyses had no heterogeneity (I
2
= 0%), while
one had medium heterogeneity (I
2
= 33.1%).
TABLE 1 | Continued
Study Clinical Trial
Registration
Number
Trial Duration
(week)
Interventions Events/Patients (N) Age (years) Man (N, %) BMI (kg/m
2
) Jadad
score
Experimental Control Experimental Control Experimental Control Experimental Control Experimental Control
Marso et al., 2016
(17)
NCT01720446 109 Semaglutide Placebo 4/1648 6/1649 64.7 64.6 1013 (61.5) 989
(60.0)
––4
Gerstein et al., 2021
(61)
NCT03496298 126 Efpeglenatide Placebo 5/2717 0/1359 64.7 64.4 1792 (66.0) 940
(69.2)
32.9 32.4 5
OAD, oral antidiabetic drugs; IDegLira, insulin degludec/liraglutide; IGlarLixi, insulin glargine/lixisenatide Fixed Ratio Combination.
FIGURE 2 | Forest plot of GLP-1 receptor agonists versus comparators on
risk of overall thyroid disorders. GLP-1RAs, GLP-1 receptor agonists; RR, risk
ratios; CI, condence interval.
FIGURE 3 | Forest plot of specic GLP-1 receptor agonists versus
comparators on risk of overall thyroid disorders. GLP-1RAs, GLP-1 receptor
agonists; RR, risk ratios; CI, condence interval.
Hu et al. GLP-1RAs and Thyroid Disorders
Frontiers in Endocrinology | www.frontiersin.org July 2022 | Volume 13 | Article 9278596
Subgroup Analyses and Meta-
Regression Analyses
Subgroup analyses based on type of underlying diseases, type of
control, trial durations and pharmacokinetics. The results
showed that the type of underlying diseases, type of control,
trial durations and pharmacokinetics did not signicantly affect
the effects of GLP-1 receptor agonists on overall thyroid
disorders (all P
subgroup
>0.05;Figure 4). The statistical
signicance of the results from the meta-regression was
consistent with the subgroup analyses.
DISCUSSION
This meta-analysis is the rst large sample study that was designed
to assess the relationship between the use of GLP-1 receptor
agonists and the occurrence of various thyroid disorders. As a
result, the following two major ndings were produced. First,
compared with placebo or other interventions, GLP-1 receptor
agonists signicantly increased the risk of overall thyroid disorders
by 28%. Second, among GLP-1 receptor agonists, only liraglutide
and dulaglutide showed increased trends in the risks of overall
thyroid disorders compared with placebo and other
antidiabetic drugs.
Despite the lack of consistent clinical and epidemiological
evidence, the potential link between GLP-1 receptor agonists and
thyroid cancer has received considerable attention. Rodent
studies have shown that treatment with liraglutide or once-
weekly exenatide is associated with thyroid C-cell proliferation
and the formation of thyroid C-cell tumors (11,63).
Therefore, the US Food and Drug Administration (FDA)
prohibits these therapies for patients with an individual or family
history of medullary thyroid carcinoma (MTC) or patients with
multiple endocrine neoplasia syndrome type 2 (MEN2).
However, these concerns are controversial in clinical trials. A
retrospective analysis of the FDAs AERS database found that the
incidence of thyroid cancer treated with exenatide was 4.7 times
that of the control drug (14). Similarly, analysis of data from the
EudraVigilance database has found evidence from spontaneous
reports that GLP-1 analogues are related to thyroid cancer in
diabetic patients (64). However, a meta-analysis involving 25
studies showed that liraglutide had no signicant correlation
with the increased risk of thyroid cancer (65). Although our
meta-analysis also showed that GLP-1 receptor agonists did not
increase the risk of thyroid cancer compared to placebo or other
interventions, in combination with previously available evidence,
patients at risk for thyroid cancer should be prescribed GLP-1
receptor agonists with caution.
To date, the potential mechanism of the unfavorable effects of
GLP-1 receptor agonists on thyroid disorders has not been
completely clear. The possible mechanisms are as follows. First, it
was reported that the mechanism of C-cell transformation in
rodents is by activation of the GLP-1 receptor on the C cell, and a
study has shown that GLP-1 receptor stimulation is a better
predictor of C-cell hyperplasia than plasma drug concentrations
of exenatide and liraglutide (66,67). Second, in addition to
medullary thyroid carcinoma and C-cell hyperplasia, the
expression of GLP-1 receptors in papillary thyroid carcinoma
(PTC) has been demonstrated. Gier et al. (68) reported positive
immunoreactivity for GLP-1 receptors in PTC tissues, detected
using a polyclonal anti-GLP-1 receptors antibody. Meanwhile, they
reported that GLP-1 receptors were expressed differently in non-
neoplastic thyroid tissues according to different inammatory
states. GLP-1 receptors were expressed in normal thyroid tissues
with inammation, but not in normal thyroid tissues without
inammation. In addition, another study also conrmed the
expression of GLP-1 receptors in PTC and the expression rate of
GLP-1 receptors in PTC, which was almost 30% (69). Korner et al.
(70) ascertained the expression of GLP-1 receptors in various
human thyroid tissues by scintigraphy and demonstrated that few
normal thyroid tissueexpressed GLP-1 receptors. Therefore, GLP-1
receptors may be abnormally induced in cells derived from thyroid
follicles through inammation, cell proliferation or tumorigenesis.
However, some of the mentioned studies used GLP-1 receptor
antibodies lacking specicity (71,72). Using another detection
method, Waser et al. found that neither normal nor hyperplastic
human thyroids containing parafollicular C cells express GLP-1
receptors (73). At present, the presence and importance of GLP-1
receptors in normal human thyroid remains controversial. Third,
GLP-1 might work through the phosphoinositol-3 kinase/AKT
serine/threonine kinase (PI3K/Akt) pathway and/or mitogen-
activated protein kinase/extracellular signal-regulated kinase
(MAPK/Erk) pathway. These two signaling pathways are also
critical in regulating cell growth and proliferation; accordingly,
they are closely related to cancer, including PTC. These two
signaling pathways are signicant pathways for regulating cell
growth and proliferation, and thus they are closely related to
cancer formation (74). Finally, the GlP-1 receptor may be
associated with triiodothyronine (T3) levels. GLP-1 stimulates
type 3 iodothyronine deiodinase (D3) expression through the
GLP-1 receptor, and the regulation of intracellular (T3)
concentration by D3 may be involved in the stimulation of
FIGURE 4 | Subgroup analyses of the effects of GLP-1 receptor agonists on
the risk of overall thyroid disorders. P value calculated by c
2
statistics is shown.
Statistical signicance of results from meta-regression was consistent.
Hu et al. GLP-1RAs and Thyroid Disorders
Frontiers in Endocrinology | www.frontiersin.org July 2022 | Volume 13 | Article 9278597
insulin secretion by GLP-1 (75). In addition, a clinical study showed
that exenatide treatment for 6 months signicantly reduced the
serum TSH concentration in diabetic patients without thyroid
disease (76). In conclusion, some animal studies have provided
evidencethat the use of GLP-1 receptor agonists increases the risk of
thyroid disease, but this evidence has not been conrmed in
humans. Therefore, we performed this meta-analysis to clarify the
association of GLP-1 receptor agonists with thyroid disease in
clinical studies and preparation for future studies in humans.
Further prospective studies should be carried out to determine
the potential effects of GLP-1 receptor agonists on thyroid disease.
In the analysis of different types of GLP-1 receptor agonists,
we found that liraglutide and dulaglutide were signicantly
associated with an increased risk of overall thyroid disorders.
However, individual tolerability and safety to GLP-1RA may
vary due to differences in molecular structures (77).
Furthermore, these different ndings could explain with an
imbalanced sample size. It is worth noting that the signicantly
increased risk of liraglutide is largely driven by the LEADER trial
(20) and that of dulaglutide is largely driven by the REWIND test
(42), both of which contributed more than 75% of the weight to
the overall results. Due to the lack of sufcient research, we
cannot draw a decisive conclusion until further research provides
more information. Among the included studies, only one was
related to short-acting exenatide (39), and two were long-acting
exenatide (18,40). Due to the small number of studies, we did
not separately analyze according to pharmacokinetics.
This review has two main strengths. First, this is the rst
meta-analysis to comprehensively assess the risks of various
thyroid diseases associated with the use of GLP-1 receptor
agonists. Moreover, all included studies were RCTs. Second, no
or only mild heterogeneity was found in any of the meta-analyses
conducted in the present study.
We acknowledge that our study has several limitations. First,
almost every included study did not consider thyroid events as the
main result, only regarded them as safety results and did not
monitor the changes in thyroid function at the same time. In
addition, only trials reporting thyroid events were included in this
analysis, leading to an unclear risk of reporting bias. Second,
although this analysis included 45 studies with a fairly large
sample size, the low incidence of thyroid events resulted in a wide
condence interval that reduced the certainty of our ndings.
Moreover, the study groups considerably differ in size (52600 vs.
41463). Considering the slight difference in the rate of thyroid
disorders (0.39 vs. 0.31%), a signicant inuence on the primary
endpoint cannot be ruled out. The third limitation is that there may
be the potential for numerous indirect effects or confounding. For
example, reduction in BMI in obesity patients, caloric restriction,
and illness are all associated with different thyroid function test
(TFT) changes. Patients may be more stringently screened,
particularly for thyroid nodules/cancer in patients receiving GLP-
1 receptor agonists. Another limitation is that for thyroid cancer,
reporting specically the cases of MTC vs. PTC would further the
goal of elucidating mechanisms of thyroid disease. However, we
found that some studies did not specify the type of thyroid cancer,
which would affect the accuracy of the results. Due to the lack of
standardization of adverse event reports and original data, we
cannot make comparisons according to different types. Finally,
although our meta-analysis showed that GLP-1 receptor agonists
increasedthe risk of overall thyroid disorder, due to the decrease in
sample size, it did not show statistically signicant results for
specic thyroid disorder. Future large long-term RCTs with
primary or secondary outcomes, including thyroid disorders and
real-world data, are needed to elucidate the association between
GLP-1 receptor agonists and the risk of various thyroid disorders,
particularly thyroid cancer.
CONCLUSION
In conclusion, compared with placebo or other interventions,
GLP-1 receptor agonists did not increase or decrease the risk
of thyroid cancer, hyperthyroidism, hypothyroidism, thyroiditis,
thyroid mass and goiter. Due to the low incidence of various
thyroid disorders, these ndings still need to be veried by
further studies.
DATA AVAILABILITY STATEMENT
The datasets presented in this study can be found in online
repositories. The names of the repository/repositories
and accession number(s) can be found in the article/
Supplementary Material.
AUTHOR CONTRIBUTIONS
JL and XL designed and outlined the work; WH, RS, RC, CL, RG,
WT, JZ and QZ drafted and revised the manuscript. Both authors
approved the nal version of the article and agree to be
accountable for all aspects of the work. All authors contributed
to the article and approved the submitted version.
FUNDING
This work was supported in part by National Natural Science
Foundation of China (No. 82000799), Research Project
Supported by Shanxi Scholarship Council of China (No. 2020-
187), Scientic Research Project of Shanxi Provincial Health
Committee (No.2021068), The Doctoral Foundation of the
Second Hospital of Shanxi Medical University (No. 20200112)
and Natural Science Foundation of Shanxi Province
(No. 202103021224243).
SUPPLEMENTARY MATERIAL
The Supplementary Material for this article can be found online at:
https://www.frontiersin.org/articles/10.3389/fendo.2022.927859/
full#supplementary-material
Hu et al. GLP-1RAs and Thyroid Disorders
Frontiers in Endocrinology | www.frontiersin.org July 2022 | Volume 13 | Article 9278598
REFERENCES
1. Gu Y, Li H, Bao X, Zhang Q, Liu L, Meng G, et al. The Relationship Between
Thyroid Function and the Prevalence of Type 2 Diabetes Mellitus in
Euthyroid Subjects. J Clin Endocrinol Metab (2017) 102(2):43442.
doi: 10.1210/jc.2016-2965
2. Nederstigt C, Corssmit EP, de Koning EJ, Dekkers OM. Incidence and
Prevalence of Thyroid Dysfunction in Type 1 Diabetes. JDiabetes
Complications (2016) 30(3):4205. doi: 10.1016/j.jdiacomp.2015.12.027
3. Vigneri P, Frasca F, Sciacca L, Pandini G, Vigneri R. Diabetes and Cancer.
Endocr Relat Cancer (2009) 16(4):110323. doi: 10.1677/ERC-09-0087
4. Aschebrook-Kilfoy B, Sabra MM, Brenner A, Moore SC, Ron E, Schatzkin A,
et al. Diabetes and Thyroid Cancer Risk in the National Institutes of Health-
AARP Diet and Health Study. Thyroid (2011) 21(9):95763. doi: 10.1089/
thy.2010.0396
5. Avgerinos KI, Spyrou N, Mantzoros CS, Dalamaga M. Obesity and Cancer
Risk: Emerging Biological Mechanisms and Perspectives. Metabolism (2019)
92:12135. doi: 10.1016/j.metabol.2018.11.001
6. Kalra S, Aggarwal S, Khandelwal D. Thyroid Dysfunction and Type 2
Diabetes Mellitus: Screening Strategies and Implications for Management.
Diabetes Ther (2019) 10(6):203544. doi: 10.1007/s13300-019-00700-4
7. Meng X, Xu S, Chen G, Derwahl M, Liu C. Metformin and Thyroid Disease. J
Endocrinol (2017) 233(1):R4351. doi: 10.1530/JOE-16-0450
8. Lee S, Tsirbas A, Goldberg RA, McCann JD. Thiazolidinedione Induced
Thyroid Associated Orbitopathy. BMC Ophthalmol (2007) 4:7:8. doi: 10.1186/
1471-2415-7-8
9. Mimura LY, Villares SM, Monteiro ML, Guazzelli IC, Bloise W. Peroxisome
Proliferator-Activated Receptor-Gamma Gene Expression in Orbital
Adipose/Connective Tissues is Increased During the Active Stage of Graves'
Ophthalmopathy. Thyroid (2003) 13(9):84550. doi: 10.1089/
105072503322401032
10. Cho YM, Fujita Y, Kieffer TJ. Glucagon-Like Peptide-1: Glucose Homeostasis
and Beyond. Annu Rev Physiol (2014) 76:53559. doi: 10.1146/annurev-
physiol-021113-170315
11. Bjerre Knudsen L, Madsen LW, Andersen S, Almholt K, de Boer AS, Drucker
DJ, et al. Glucagon-Like Peptide-1 Receptor Agonists Activate Rodent
Thyroid C-Cells Causing Calcitonin Release and C-Cell Proliferation.
Endocrinology (2010) 151(4):147386. doi: 10.1210/en.2009-1272
12. Martı
n-Lacave I, Bernab R, Sampedro C, Conde E, Fernandez-Santos JM, San
Martı
n MV, et al. Correlation Between Gender and Spontaneous C-Cell
Tumors in the Thyroid Gland of the Wistar Rat. Cell Tissue Res (1999) 297
(3):4517. doi: 10.1007/s004410051371
13. Capen CC, Martin SL. The Effects of Xenobiotics on the Structure and
Function of Thyroid Follicular and C-Cells. Toxicol Pathol (1989) 17
(2):26693. doi: 10.1177/019262338901700205
14. Elashoff M, Matveyenko AV, Gier B, Elashoff R, Butler PC. Pancreatitis,
Pancreatic, and Thyroid Cancer With Glucagon-Like Peptide-1-Based
Therapies. Gastroenterology (2011) 141(1):1506. doi: 10.1053/j.gastro.
2011.02.018
15. Hegedüs L, Sherman SI, Tuttle RM, von Scholten BJ, Rasmussen S, Karsbøl
JD, et al. No Evidence of Increase in Calcitonin Concentrations or
Development of C-Cell Malignancy in Response to Liraglutide for Up to 5
Years in the LEADER Trial. Diabetes Care (2018) 41(3):6202. doi: 10.2337/
dc17-1956
16. Brown E, Heerspink HJL, Cuthbertson DJ, Wilding JPH. SGLT2
Inhibitors and GLP-1 Receptor Agonists: Established and Emerging
Indications. Lancet (2021) 398(10296):26276. doi: 10.1016/S0140-6736(21)
00536-5
17. Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jodar E, Leiter LA, et al.
Semaglutide and Cardiovascular Outcomes in Patients With Type 2 Diabetes.
N Engl J Med (2016) 375(19):183444. doi: 10.1056/NEJMoa1607141
18. Holman RR, Bethel MA, Mentz RJ, Thompson VP, Lokhnygina Y, Buse JB,
et al. Effects of Once-Weekly Exenatide on Cardiovascular Outcomes in Type
2Diabetes.NEnglJMed(2017) 377(13):122839. doi: 10.1056/
NEJMoa1612917
19. Leiter LA, Carr MC, Stewart M, Jones-Leone A, Scott R, Yang F, et al. Efcacy
and Safety of the Once-Weekly GLP-1 Receptor Agonist Albiglutide Versus
Sitagliptin in Patients With Type 2 Diabetes and Renal Impairment: A
Randomized Phase III Study. Diabetes Care (2014) 37(10):272330.
doi: 10.2337/dc13-2855
20. Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck
MA, et al. Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes. N
Engl J Med (2016) 375(4):31122. doi: 10.1056/NEJMoa1603827
21. Unger J, Allison DC, Kaltoft M, Lakkole K, Panda JK, Ramesh C, et al.
Maintenance of Glycaemic Control With Liraglutide Versus Oral Antidiabetic
Drugs as Add-on Therapies in Patients With Type 2 Diabetes Uncontrolled
With Metformin Alone: A Randomized Clinical Trial in Primary Care (LIRA-
PRIME). Diabetes Obes Metab (2022) 24(2):20411. doi: 10.1111/dom.14566
22. Garvey WT, Birkenfeld AL, Dicker D, Mingrone G, Pedersen SD, Satylganova
A, et al. Efcacy and Safety of Liraglutide 3.0 Mg in Individuals With
Overweight or Obesity and Type 2 Diabetes Treated With Basal Insulin:
The SCALE Insulin Randomized Controlled Trial. Diabetes Care (2020) 43
(5):108593. doi: 10.2337/dc19-1745
23. Wadden TA, Tronieri JS, Sugimoto D, Lund MT, Auerbach P, Jensen C, et al.
Liraglutide 3.0 Mg and Intensive Behavioral Therapy (IBT) for Obesity in
Primary Care: The SCALE IBT Randomized Controlled Trial. Obes (Silver
Spring) (2020) 28(3):52936. doi: 10.1002/oby.22726
24. le Roux CW, Astrup A, Fujioka K, Greenway F, Lau DCW, Van Gaal L, et al. 3
Years of Liraglutide Versus Placebo for Type 2 Diabetes Risk Reduction and
Weight Management in Individuals With Prediabetes: A Randomised,
Double-Blind Trial. Lancet (2017) 389(10077):1399409. doi: 10.1016/
S0140-6736(17)30069-7
25. Pi-Sunyer X, Astrup A, Fujioka K, Greenway F, Halpern A, Krempf M, et al. A
Randomized, Controlled Trial of 3.0 Mg of Liraglutide in Weight
Management. NEnglJMed(2015) 373(1):1122. doi: 10.1056/
NEJMoa1411892
26. Zang L, Liu Y, Geng J, Luo Y, Bian F, Lv X, et al. Efcacy and Safety of
Liraglutide Versus Sitagliptin, Both in Combination With Metformin, in
Chinese Patients With Type 2 Diabetes: A 26-Week, Open-Label,
Randomized, Active Comparator Clinical Trial. Diabetes Obes Metab (2016)
18(8):80311. doi: 10.1111/dom.12674
27. Ahren B, Hirsch IB, Pieber TR, Mathieu C, Gomez-Peralta F, Hansen TK,
et al. Efcacy and Safety of Liraglutide Added to Capped Insulin Treatment in
Subjects With Type 1 Diabetes: The ADJUNCT TWO Randomized Trial.
Diabetes Care (2016) 39(10):1693701. doi: 10.2337/dc16-0690
28. Mathieu C, Zinman B, Hemmingsson JU, Woo V, Colman P, Christiansen E,
et al. Efcacy and Safety of Liraglutide Added to Insulin Treatment in Type 1
Diabetes: The ADJUNCT ONE Treat-To-Target Randomized Trial. Diabetes
Care (2016) 39(10):170210. doi: 10.2337/dc16-0691
29. Davies MJ, Bergenstal R, Bode B, Kushner RF, Lewin A, Skjøth TV, et al.
Efcacy of Liraglutide for Weight Loss Among Patients With Type 2 Diabetes:
The SCALE Diabetes Randomized Clinical Trial. JAMA (2015) 314(7):68799.
doi: 10.1001/jama.2015.9676
30. Gough SC, Bode B, Woo V, Rodbard HW, Linjawi S, Poulsen P, et al. Efcacy
and Safety of a Fixed-Ratio Combination of Insulin Degludec and Liraglutide
(IDegLira) Compared With its Components Given Alone: Results of a Phase
3, Open-Label, Randomised, 26-Week, Treat-to-Target Trial in Insulin-Naive
Patients With Type 2 Diabetes. Lancet Diabetes Endocrinol (2014) 2(11):885
93. doi: 10.1016/S2213-8587(14)70174-3
31. Wadden TA, Hollander P, Klein S, Niswender K, Woo V, Hale PM, et al.
Weight Maintenance and Additional Weight Loss With Liraglutide After
Low-Calorie-Diet-Induced Weight Loss: The SCALE Maintenance
Randomized Study. Int J Obes (Lond) (2015) 39(1):187. doi: 10.1038/
ijo.2013.120
32. Seino Y, Rasmussen MF, Nishida T, Kaku K. Efcacy and Safety of the Once-
Daily Human GLP-1 Analogue, Liraglutide, vs Glibenclamide Monotherapy
in Japanese Patients With Type 2 Diabetes. Curr Med Res Opin (2010) 26
(5):101322. doi: 10.1185/03007991003672551
33. Pratley RE, Nauck M, Bailey T, Montanya E, Cuddihy R, Filetti S, et al.
Liraglutide Versus Sitagliptin for Patients With Type 2 Diabetes Who did Not
Have Adequate Glycaemic Control With Metformin: A 26-Week,
Randomised, Parallel-Group, Open-Label Trial. Lancet (2010) 375
(9724):144756. doi: 10.1016/S0140-6736(10)60307-8
34. Nauck M, Frid A, Hermansen K, Shah NS, Tankova T, Mitha IH, et al. Efcacy
and Safety Comparison of Liraglutide, Glimepiride, and Placebo, All in
Combination With Metformin, in Type 2 Diabetes: The LEAD (Liraglutide
Hu et al. GLP-1RAs and Thyroid Disorders
Frontiers in Endocrinology | www.frontiersin.org July 2022 | Volume 13 | Article 9278599
Effect and Action in Diabetes)-2 Study. Diabetes Care (2009) 32(1):8490.
doi: 10.2337/dc08-1355
35. Garber A, Henry R, Ratner R, Garcia-Hernandez PA, Rodriguez-Pattzi H,
Olvera-Alvarez I, et al. Liraglutide Versus Glimepiride Monotherapy for Type
2 Diabetes (LEAD-3 Mono): A Randomised, 52-Week, Phase III, Double-
Blind, Parallel-Treatment Trial. Lancet (2009) 373(9662):47381.
doi: 10.1016/S0140-6736(08)61246-5
36. Hernandez AF, Green JB, Janmohamed S, D'Agostino RB, Granger CB, Jones
NP, et al. Albiglutide and Cardiovascular Outcomes in Patients With Type 2
Diabetes and Cardiovascular Disease (Harmony Outcomes): A Double-Blind,
Randomised Placebo-Controlled Trial. Lancet (2018) 392(10157):151929.
doi: 10.1016/S0140-6736(18)32261-X
37. Home PD, Shamanna P, Stewart M, Yang F, Miller M, Perry C, et al. Efcacy
and Tolerability of Albiglutide Versus Placebo or Pioglitazone Over 1 Year in
People With Type 2 Diabetes Currently Taking Metformin and Glimepiride:
HARMONY 5. Diabetes Obes Metab (2015) 17(2):17987. doi: 10.1111/
dom.12414
38. Ahren B, Johnson SL, Stewart M, Cirkel DT, Yang F, Perry C, et al.
HARMONY 3: 104-Week Randomized, Double-Blind, Placebo- and Active-
Controlled Trial Assessing the Efcacy and Safety of Albiglutide Compared
With Placebo, Sitagliptin, and Glimepiride in Patients With Type 2 Diabetes
Taking Metformin. Diabetes Care (2014) 37(8):21418. doi: 10.2337/dc14-
0024
39. Gallwitz B, Guzman J, Dotta F, Guerci B, SimoR, Basson BR, et al. Exenatide
Twice Daily Versus Glimepiride for Prevention of Glycaemic Deterioration in
Patients With Type 2 Diabetes With Metformin Failure (EUREXA): An
Open-Label, Randomised Controlled Trial. Lancet (2012) 379(9833):22708.
doi: 10.1016/S0140-6736(12)60479-6
40. Bergenstal RM, Wysham C, Macconell L, Malloy J, Walsh B, Yan P, et al.
Efcacy and Safety of Exenatide Once Weekly Versus Sitagliptin or
Pioglitazone as an Adjunct to Metformin for Treatment of Type 2 Diabetes
(DURATION-2): A Randomised Trial. Lancet (2010) 376(9739):4319.
doi: 10.1016/S0140-6736(10)60590-9
41. Wang W, Nevarez L, Filippova E, Song KH, Tao B, Gu L, et al. Efcacy and
Safety of Once-Weekly Dulaglutide Versus Insulin Glargine in Mainly Asian
Patients With Type 2 Diabetes Mellitus on Metformin and/or a
Sulphonylurea: A 52-Week Open-Label, Randomized Phase III Trial.
Diabetes Obes Metab (2019) 21(2):23443. doi: 10.1111/dom.13506
42. Gerstein HC, Colhoun HM, Dagenais GR, Diaz R, Lakshmanan M, Pais P,
et al. Dulaglutide and Cardiovascular Outcomes in Type 2 Diabetes
(REWIND): A Double-Blind, Randomised Placebo-Controlled Trial. Lancet
(2019) 394(10193):12130. doi: 10.1016/S0140-6736(19)31149-3
43. Chen YH, Huang CN, Cho YM, Li P, Gu L, Wang F, et al. Efcacy and Safety
of Dulaglutide Monotherapy Compared With Glimepiride in East-Asian
Patients With Type 2 Diabetes in a Multicentre, Double-Blind,
Randomized, Parallel-Arm, Active Comparator, Phase III Trial. Diabetes
Obes Metab (2018) 20(9):212130. doi: 10.1111/dom.13340
44. Weinstock RS, Guerci B, Umpierrez G, Nauck MA, Skrivanek Z, Milicevic Z.
Safety and Efcacy of Once-Weekly Dulaglutide Versus Sitagliptin After
2 Years in Metformin-Treated Patients With Type 2 Diabetes (AWARD-5):
A Randomized, Phase III Study. Diabetes Obes Metab (2015) 17(9):84958.
doi: 10.1111/dom.12479
45. Giorgino F, Benroubi M, Sun JH, Zimmermann AG, Pechtner V. Efcacy and
Safety of Once-Weekly Dulaglutide Versus Insulin Glargine in Patients With
Type 2 Diabetes on Metformin and Glimepiride (AWARD-2). Diabetes Care
(2015) 38(12):22419. doi: 10.2337/dc14-1625
46. Rosenstock J, Aronson R, Grunberger G, Hanefeld M, Piatti P, Serusclat P,
et al. Benets of LixiLan, a Titratable Fixed-Ratio Combination of Insulin
Glargine Plus Lixisenatide, Versus Insulin Glargine and Lixisenatide
Monocomponents in Type 2 Diabetes Inadequately Controlled on Oral
Agents: The LixiLan-O Randomized Trial. Diabetes Care (2016) 39
(11):202635. doi: 10.2337/dc16-0917
47. Pfeffer MA, Claggett B, Diaz R, Dickstein K, Gerstein HC, Køber LV, et al.
Lixisenatide in Patients With Type 2 Diabetes and Acute Coronary Syndrome.
N Engl J Med (2015) 373(23):224757. doi: 10.1056/NEJMoa1509225
48. Bolli GB, Munteanu M, Dotsenko S, Niemoeller E, Boka G, Wu Y, et al.
Efcacy and Safety of Lixisenatide Once Daily vs. Placebo in People With Type
2 Diabetes Insufciently Controlled on Metformin (GetGoal-F1). Diabetes
Med (2014) 31(2):17684. doi: 10.1111/dme.12328
49. Ahren B, Leguizamo Dimas A, Miossec P, Saubadu S, Aronson R. Efcacy and
Safety of Lixisenatide Once-Daily Morning or Evening Injections in Type 2
Diabetes Inadequately Controlled on Metformin (GetGoal-M). Diabetes Care
(2013) 36(9):254350. doi: 10.2337/dc12-2006
50. Riddle MC, Aronson R, Home P, Marre M, Niemoeller E, Miossec P, et al.
Adding Once-Daily Lixisenatide for Type 2 Diabetes Inadequately Controlled
by Established Basal Insulin: A 24-Week, Randomized, Placebo-Controlled
Comparison (GetGoal-L). Diabetes Care (2013) 36(9):248996. doi: 10.2337/
dc12-2454
51. Wilding JPH, Batterham RL, Calanna S, Davies M, Van Gaal LF, Lingvay I,
et al. Once-Weekly Semaglutide in Adults With Overweight or Obesity. N
Engl J Med (2021) 384(11):989. doi: 10.1056/NEJMoa2032183
52. Wadden TA, Bailey TS, Billings LK, Davies M, Frias JP, Koroleva A, et al.
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):140313.
doi: 10.1001/jama.2021.1831
53. Yamada Y, Katagiri H, Hamamoto Y, Deenadayalan S, Navarria A, Nishijima
K, et al. Dose-Response, Efcacy, and Safety of Oral Semaglutide
Monotherapy in Japanese Patients With Type 2 Diabetes (PIONEER 9): A
52-Week, Phase 2/3a, Randomised, Controlled Trial. Lancet Diabetes
Endocrinol (2020) 8(5):37791. doi: 10.1016/S2213-8587(20)30075-9
54. Husain M, Birkenfeld AL, Donsmark M, Dungan K, Eliaschewitz FG, Franco
DR, et al. Oral Semaglutide and Cardiovascular Outcomes in Patients With
Type 2 Diabetes. NEnglJMed(2019) 381(9):84151. doi: 10.1056/
NEJMoa1901118
55. Rosenstock J, Allison D, Birkenfeld AL, Blicher TM, Deenadayalan S, Jacobsen
JB, et al. Effect of Additional Oral Semaglutide vs Sitagliptin on Glycated
Hemoglobin in Adults With Type 2 Diabetes Uncontrolled With Metformin
Alone or With Sulfonylurea: The PIONEER 3 Randomized Clinical Trial.
JAMA (2019) 321(15):146680. doi: 10.1001/jama.2019.2942
56. Pratley R, Amod A, Hoff ST, Kadowaki T, Lingvay I, Nauck M, et al. Oral
Semaglutide Versus Subcutaneous Liraglutide and Placebo in Type 2 Diabetes
(PIONEER 4): A Randomised, Double-Blind, Phase 3a Trial. Lancet (2019)
394(10192):3950. doi: 10.1016/S0140-6736(19)31271-1
57. Aroda VR, Rosenstock J, Terauchi Y, Altuntas Y, Lalic NM, Morales Villegas
EC, et al. PIONEER 1: Randomized Clinical Trial Comparing the Efcacy and
Safety of Oral Semaglutide Monotherapy With Placebo in Patients With Type
2 Diabetes. Diabetes Care (2019) 42(9):172432. doi: 10.2337/dc19-0749
58. O'Neil PM, Birkenfeld AL, McGowan B, Mosenzon O, Pedersen SD, Wharton
S, et al. Efcacy 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):63749. doi: 10.1016/S0140-6736(18)31773-2
59. Ahren B, Masmiquel L, Kumar H, Sargin M, Karsbøl JD, Jacobsen SH, et al.
Efcacy 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):34154.
doi: 10.1016/S2213-8587(17)30092-X
60. Aroda VR, Bain SC, Cariou B, PileticM, Rose L, Axelsen M, et al. Efcacy 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):35566. doi: 10.1016/S2213-8587(17)30085-2
61. Gerstein HC, Sattar N, Rosenstock J, Ramasundarahettige C, Pratley R, Lopes
RD, et al. Cardiovascular and Renal Outcomes With Efpeglenatide in Type 2
Diabetes. NEnglJMed(2021) 385(10):896907. doi: 10.1056/
NEJMoa2108269
62. Effect of Liraglutide on Body Weight in Non-Diabetic Obese Subjects or
Overweight Subjects With Co-Morbidities: SCALE-Obesity and Pre-
Diabetes (2011). Available at: https://ClinicalTrials.gov/show/NCT01272219.
63. Bulchandani D, Nachnani JS, Herndon B, Molteni A, Pathan MH, Quinn T,
et al. Effect of Exendin (Exenatide)GLP 1 Receptor Agonist on the Thyroid
Hu et al. GLP-1RAs and Thyroid Disorders
Frontiers in Endocrinology | www.frontiersin.org July 2022 | Volume 13 | Article 92785910
and Parathyroid Gland in a Rat Model. Eur J Pharmacol (2012) 691(1-3):292
6. doi: 10.1016/j.ejphar.2012.07.024
64. Mali G, Ahuja V, Dubey K. Glucagon-Like Peptide-1 Analogues and Thyroid
Cancer: An Analysis of Cases Reported in the European Pharmacovigilance
Database. J Clin Pharm Ther (2021) 46(1):99105. doi: 10.1111/jcpt.13259
65. Alves C, Batel-Marques F, Macedo AF. A Meta-Analysis of Serious Adverse
Events Reported With Exenatide and Liraglutide: Acute Pancreatitis and
Cancer. Diabetes Res Clin Pract (2012) 98(2):27184. doi: 10.1016/
j.diabres.2012.09.008
66. Madsen LW, Knauf JA, Gotfredsen C, Pilling A, Sjögren I, Andersen S, et al.
GLP-1 Receptor Agonists and the Thyroid: C-Cell Effects in Mice are
Mediated via the GLP-1 Receptor and Not Associated With RET
Activation. Endocrinology (2012) 153(3):153847. doi: 10.1210/en.2011-1864
67. van den Brink W, Emerenciana A, Bellanti F, Della Pasqua O, van der Laan
JW. Prediction of Thyroid C-Cell Carcinogenicity After Chronic
Administration of GLP1-R Agonists in Rodents. Toxicol Appl Pharmacol
(2017) 320:519. doi: 10.1016/j.taap.2017.02.010
68. Gier B, Butler PC, Lai CK, Kirakossian D, DeNicola MM, Yeh MW. Glucagon
Like Peptide-1 Receptor Expression in the Human Thyroid Gland. J Clin
Endocrinol Metab (2012) 97(1):12131. doi: 10.1210/jc.2011-2407
69. Jung MJ, Kwon SK. Expression of Glucagon-Like Peptide-1 Receptor in
Papillary Thyroid Carcinoma and its Clinicopathologic Signicance.
Endocrinol Metab (Seoul) (2014) 29(4):53644. doi: 10.3803/
EnM.2014.29.4.536
70. Körner M, Stöckli M, Waser B, Reubi JC. GLP-1 Receptor Expression in
Human Tumors and Human Normal Tissues: Potential for In Vivo Targeting.
J Nucl Med (2007) 48(5):73643. doi: 10.2967/jnumed.106.038679
71. Pyke C, Knudsen LB. The Glucagon-Like Peptide-1 Receptoror Not?
Endocrinology (2013) 154(1):48. doi: 10.1210/en.2012-2124
72. McLean BA,Wong CK, CampbellJE, Hodson DJ,Trapp S, DruckerDJ. Revisiting
the Complexity of GLP-1 Action From Sites of Synthesis toReceptor Activation.
Endocr Rev (2021) 42(2):10132. doi: 10.1210/endrev/bnaa032
73. Waser B, Blank A, Karamitopoulou E, Perren A, Reubi JC. Glucagon-Like-
Peptide-1 Receptor Expression in Normal and Diseased Human Thyroid and
Pancreas. Mod Pathol (2015) 28(3):391402. doi: 10.1038/modpathol.
2014.113
74. He L, Zhang S, Zhang X, Liu R, Guan H, Zhang H. Effects of Insulin Analogs
and Glucagon-Like Peptide-1 Receptor Agonists on Proliferation and Cellular
Energy Metabolism in Papillary Thyroid Cancer. Onco Targets Ther (2017)
10:562131. doi: 10.2147/OTT.S150701
75. Akiyama S, Ogiwara T, Aoki T, Tsunekawa K, Araki O, Murakami M.
Glucagon-Like Peptide-1 Stimulates Type 3 Iodothyronine Deiodinase
Expression in a Mouse Insulinoma Cell Line. Life Sci (2014) 115(1-2):228.
doi: 10.1016/j.lfs.2014.09.004
76. Sencar ME, Sakiz D, Calapkulu M, Hepsen S, Kizilgul M, Ozturk IU, et al. The
Effect of Exenatide on Thyroid-Stimulating Hormone and Thyroid Volume.
Eur Thyroid J (2019) 8(6):30711. doi: 10.1159/000501895
77. Gentilella R, Pechtner V, Corcos A, Consoli A. Glucagon-Like Peptide-1
Receptor Agonists in Type 2 Diabetes Treatment: Are They All the Same?
Diabetes Metab Res Rev (2019) 35(1):e3070. doi: 10.1002/dmrr.3070
Conict of Interest: The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be construed as a
potential conict of interest.
Publishers Note: All claims expressed in this article are solely those of the authors
and do not necessarily represent those of their afliated organizations, or those of
the publisher, the editors and the reviewers. Any product that may be evaluated in
this article, or claim that may be made by its manufacturer, is not guaranteed or
endorsed by the publisher.
Copyright © 2022 Hu, Song, Cheng, Liu, Guo, Tang, Zhang, Zhao, Li and Liu. This is
an open-access article distributed under the terms of the Creative Commons
Attribution License (CC BY). The use, distribution or reproduction in other forums
is permitted, provided the original author(s) and the copyright owner(s) are credited
and that the original publication in this journal is cited, in accordance with accepted
academic practice. No use, distribution or reproduction is permitted which does not
comply with these terms.
Hu et al. GLP-1RAs and Thyroid Disorders
Frontiers in Endocrinology | www.frontiersin.org July 2022 | Volume 13 | Article 92785911
... Thyroid-related adverse events, including hypothyroidism and thyroiditis, have been reported in pharmacovigilance databases and systematic reviews. A recent meta-analysis of randomized controlled trials reported a 28% increased risk of overall thyroid disorders with GLP-1 receptor agonist use, though the association with hypothyroidism or thyroiditis specifically was not statistically significant [2]. Additionally, pharmacovigilance analyses using the FDA Adverse Event Reporting System (FAERS) noted an increased number of thyroid-related reports, including hyperplasia and neoplasms, associated with GLP-1 receptor agonists. ...
... Drug-induced thyroiditis is a recognized but relatively rare condition, often associated with medications such as interferons, amiodarone, lithium, and tyrosine kinase inhibitors [2,3]. While the mechanism in these cases may involve immune modulation or cytotoxic effects on thyroid follicular cells, GLP-1 receptor agonists have primarily been linked to C-cell effects in animal models. ...
... For example, a meta-analysis of 45 randomized controlled trials observed a trend toward increased risk of thyroid dysfunction in patients treated with GLP-1 receptor agonists, although the findings did not reach statistical significance. Therefore, while these data indicate a possible correlation, they do not confirm a causal relationship, and further studies are needed to clarify this potential link [2]. ...
... Именно этим можно объяснить, что в недавнем метаанализе отмечено увеличение на 28% общего риска заболеваний щитовидной железы при использовании арГПП-1 по сравнению с плацебо или другими вмешательствами, но не было выявлено существенной корреляции с частотой рака щитовидной железы [68]. Это кажется вполне убедительным, если принять во внимание, что само по себе ожирение -значимый фактор онкологического риска [69] и стратегии снижения массы тела должны способствовать снижению онкологической заболеваемости, что уже находит свое подтверждение [70]. ...
Article
The rapid progress in the development of highly effective weekly incretin-based medications offers increasingly broad opportunities for comprehensive correction of cardiometabolic disorders in patients with type 2 diabetes and/or obesity. This article aims to summarize existing research that confirms the efficacy and safety of one of the most prescribed medications from the class of glucagon-like peptide-1 receptor agonists – weekly semaglutide. In addition to presenting the main results of randomized clinical trials involving semaglutide, special emphasis is placed on experimental and clinical studies related to the drug’s effectiveness in real-world conditions and during specific life periods for patients with type 2 diabetes and/or obesity, such as surgical and endoscopic interventions, bariatric surgery, intermittent fasting, and religious dietary restrictions. Based on this evidence base and their own clinical experience, the interdisciplinary author team proposes practical approaches to adjusting hypoglycemic therapy in patients with type 2 diabetes when combined with semaglutide and switching to other therapies. Practical recommendations for the use of the drug in patients with obesity during the active weight loss phase and the weight maintenance phase are also formulated. Key considerations supporting long-term obesity treatment are presented; however, trial de-escalation therapy schemes are also provided for patients who have successfully modified their lifestyle while achieving target weight outcomes. The reasons and mechanisms behind the most common adverse events associated with semaglutide use, which represent a potential barrier to its utilization, are separately discussed. The most effective measures for their prevention and correction are outlined, which will enable the realization of the therapeutic potential of weekly semaglutide and thus improve patient outcomes in the long-term management of obesity and type 2 diabetes.
... 123 An older analysis by Hu et al. did not find a significant relationship between thyroid cancer and the use of GLP-1a. 114 The current carcinogenic studies suggest that GLP-1a may stimulate the proliferation of thyroid cells, potentially increasing the risk of thyroid cancer. 115 Given the conflicting results, additional studies are needed to clarify the relationship between GLP-1a and thyroid cancer. ...
Article
Full-text available
Background Novel antidiabetic medications (SGLT-2 inhibitors, DPP-4 inhibitors, and GLP-1 agonists) are commonly used worldwide; however, the available research lacks definitive conclusions on their protective effects or potential risks on cancer. Objectives Compared to other antidiabetics, our systematic review and network meta-analysis (NMA) aims to use real-world studies to assess the potential cancer risks or protective effects of these novel antidiabetics. Methods We comprehensively searched PubMed, CINAHL, and Web of Science from their inception until November 30, 2023. We included observational studies examining at least one novel antidiabetics in the systematic review. The novel antidiabetics include sodium-glucose cotransporter-2 inhibitors (SGLT-2i), dipeptidyl peptidase-4 inhibitors (DPP-4i), and glucagon-like peptide-1 agonists (GLP-1a). Design We focused on cohort studies that provided data on cancer incidence and sample size in the NMA. Using NetMetaXL®, the random effects model with informative priors was used in the NMA to estimate the pooled odds ratio (OR) with 95% credible intervals (CrI). Results The systematic review included 62 studies, of which 22 met the inclusion criteria for the NMA. SGLT-2i users had lower overall cancer risk compared to sulfonylureas (OR: 0.54; 95% CrI: 0.40–0.74, low certainty), GLP-1a (OR: 0.70; 95% CrI: 0.53–0.92, low certainty), and DPP-4i users (OR: 0.72; 95% CrI: 0.57–0.92, very low certainty). DPP-4i users also had a lower cancer risk than sulfonylureas users (OR: 0.76; 95% CrI: 0.60–0.96, low certainty). No other statistically significant ORs were found in other direct comparisons. Conclusion SGLT-2i users have a lower risk of developing cancers than sulfonylureas, GLP-1a, and DPP-4i users. These results may improve patient safety by guiding future clinical practice and medication choices. Future studies should investigate the mechanisms behind these observed associations. Trial registration This NMA was registered in PROSPERO (CRD42023469941).
... Furthermore, in a meta-analysis study, a moderate increase in the relative risk and a slight increase in absolute risk of thyroid cancer with the use of GLP-1 agonists was reported 48 . On the other hand, other studies have reported no association between the use of GLP-1 agonists and the risk of thyroid cancer 49,50 . In sum, GLP-1 agonists associate with a reduced risk in CRC and a modest relative increase in thyroid cancer risk; however, further study is warranted to conclude whether and to what extent GLP-1 agonists elevate thyroid cancer risk. ...
Article
Full-text available
Medical therapeutics for weight loss are changing the landscape of obesity but impacts on obesity-associated cancer remain unclear. We report that in pre-clinical models with significant retatrutide (RETA, LY3437943)-induced weight loss, pancreatic cancer engraftment was reduced, tumor onset was delayed, and progression was attenuated resulting in a 14-fold reduction in tumor volume compared to only 4-fold reduction in single agonist semaglutide-treated mice. Despite weight re-gain after RETA withdrawal, the anti-tumor benefits of RETA persisted. Remarkably, RETA-induced protection extends to a lung cancer model with 50% reduced tumor engraftment, significantly delayed tumor onset, and mitigated tumor progression, with a 17-fold reduction in tumor volume compared to controls. RETA induced immune reprogramming systemically and in the tumor microenvironment with durable anti-tumor immunity evidenced by elevated circulating IL-6, increased antigen presenting cells, reduced immunosuppressive cells, and activation of pro-inflammatory pathways. In sum, our findings suggest that patients with RETA-mediated weight loss may also benefit from reduced cancer risk and improved outcomes.
... and goitre (RR 1.17, 95% CI 0.74-1.86) [27]. ...
Article
Full-text available
Aims GLP‐1 receptor agonists, such as semaglutide (Ozempic) and tirzepatide (Monjaro), have gained significant popularity for obesity management, but concerns have arisen about their potential link to thyroid cancer. This study investigates the association between thyroid cancer and weight‐loss medications. Materials and Methods A disproportionality analysis was conducted using data from the FDA Adverse Event Reporting System (FAERS) from 2004 to Q1 2024. Reporting odds ratios (RORs) were used to identify associations between thyroid cancer and weight‐loss drugs, including anti‐diabetic medications. Results Significant positive associations with thyroid cancer were found for GLP‐1 receptor agonists: semaglutide (ROR = 7.61, 95% CI: 6.37–9.08), dulaglutide (ROR = 3.59, 95% CI: 3.03–4.27), liraglutide (ROR = 15.59, 95% CI: 13.94–17.44) and tirzepatide (ROR = 2.09, 95% CI: 1.51–2.89). A weak inverse association was observed for metformin (ROR = 0.58, 95% CI: 0.36–0.93). No significant associations were found for other drugs, such as topiramate, dapagliflozin and insulin glargine. Conclusion The study, based on data from the FAERS database, suggests a potential association between GLP‐1 receptor agonists and an increased thyroid cancer risk. These findings underscore the importance of further research and continuous safety monitoring when prescribing these medications for obesity management.
Article
Background: Incretin mimetics, including glucagon-like peptide-1 receptor agonists (GLP-1 receptor agonist) and dipeptidyl peptidase-4 (DPP-4) inhibitors, have been increasingly utilized for glycemic control in patients with type 2 diabetes (T2D). Studies have demonstrated additional improvements in weight loss, cardiovascular health, and renal outcomes. Animal studies have shown an association between GLP-1 receptor agonists and C-cell proliferation and elevated calcitonin, resulting in an FDA black box. Insulin resistance in patients with T2D, along with the use of other glucose control medications, confounds the relationship between incretin mimetics and thyroid cancers. The true effect of incretin mimetics on thyroid cancer remains uncertain and speculative due to this confounding. Methods: This retrospective cohort study compared patients with T2D, who were new users of incretin mimetics, to new users of metformin. Study patients used no other anti-diabetes medications beyond the study medications. The risks of incident thyroid cancer and subsequent thyroidectomy were quantified using Cox proportional hazards regression models fitted with adjustments for demographic and medical covariates over a three-year study period. Medullary thyroid cancer (MTC) and multiple endocrine neoplasia type II (MEN2) cases were quantified. Results: Of the 91,394 patients, 28 incretin mimetic users had a diagnosis of thyroid cancer, and nine of these patients underwent a subsequent thyroidectomy procedure. No incretin mimetic user was diagnosed with MTC or MEN2. There was no statistically significant effect on the overall incretin mimetic category (1.28 aHR, 0.83–1.96), the incretin mimetic subcategories of GLP-1 receptor agonists (1.35 aHR, 0.80–2.29), or DPP-4 inhibitor (0.62 aHR, 0.33–1.17) users in developing thyroid cancer within three years of drug initiation. Similarly, no association was found between the overall incretin mimetic category (1.02 aHR, 0.49–2.10), the subcategories of GLP-1 receptor agonists (1.26 aHR, 0.54–2.96), or DPP-4 inhibitors (0.32 aHR, 0.08–1.37) and a subsequent thyroidectomy. Conclusions: In this real-world cohort study, exposure to incretin mimetics overall or through the incretin mimetic subcategories of GLP-1 receptor agonists and DPP-4 inhibitors was not associated with risks of thyroid cancer or thyroidectomy compared to metformin users.
Article
Purpose The purpose of this review is to highlight the role of glucagon-like peptide-1 receptor agonists (GLP-1 RAs) and dual GLP-1/glucose-dependent insulinotropic polypeptide (GIP) receptor agonists (GLP-1/GIP RAs) in managing cardiovascular-kidney-metabolic (CKM) syndrome, focusing on their cardiovascular (CV) and kidney-protective effects beyond glycemic control. Summary In multiple randomized controlled trials, GLP-1 RAs were demonstrated to confer significant benefits in reducing CV events and preserving kidney function in patients with preexisting atherosclerotic cardiovascular disease (ASCVD) and those at high CV risk. Current guidelines, including those from the Kidney Disease: Improving Global Outcomes (KDIGO) initiative and the American Diabetes Association (ADA), underscore the therapeutic potential of these agents for managing chronic kidney disease (CKD), type 2 diabetes mellitus (T2DM), and metabolic syndrome. Additionally, emerging data suggests their utility beyond T2DM. This review summarizes the evidence supporting these guidelines, along with newer findings not yet fully integrated into clinical practice. It also examines the role of pharmacists and multidisciplinary teams, safety considerations, and practical strategies for managing common adverse effects. Conclusion The integration of GLP-1 RAs and dual GLP-1/GIP RAs into clinical practice offers substantial benefits for patients, both with and without diabetes. Pharmacists play a pivotal role in recommending evidence-based treatments for those at high CV and kidney risk, educating patients, addressing social determinants of health, and bridging gaps across multidisciplinary care teams.
Article
Full-text available
Glucagon‐like peptide‐1 receptor agonists (GLP‐1RAs) have emerged as key agents for weight management, based on their marked efficacy as observed in randomized controlled trials. While still limited, real‐world studies on GLP‐1RA use in populations with obesity are increasingly available. This narrative review discusses contemporary real‐world evidence demonstrating the utilization, clinical and comparative effectiveness, and adverse effects of the currently approved GLP‐1RA‐based weight‐loss therapies, that is, liraglutide, semaglutide and tirzepatide. The observed weight reduction in clinical practice overall tends to be lower than in randomized controlled trials; however, outcomes approach those seen in trials when focusing on highly adherent patients. Real‐world studies demonstrate high discontinuation rates of GLP‐1RAs (20%–50%) within the first year, and the use of much lower doses than those evaluated in clinical trials. Evidence from observational studies within type 2 diabetes or obesity populations suggests frequent gastrointestinal disturbances in GLP‐1RA users, as also observed in trials, but no clear increase in risks of severe events like pancreatitis or pancreatic cancer, thyroid disorders, or depression and self‐harm. Further evidence is needed to understand possible real‐world associations of GLP‐1RAs with eye disease and other rare outcomes. We provide 10 areas of particular importance for further research on GLP‐1RA within the real‐world space, including improved understanding of the exact drivers of early discontinuation and suboptimal dosing, studies of the effects of stopping GLP‐1RA treatment, and investigations of clinical and cost‐effectiveness for hard clinical outcomes in real‐world settings, including not only cardio‐reno‐metabolic outcomes but also obesity‐induced diseases like neuropsychiatric disease, cancer, musculoskeletal disease, and infections. Plain Language Summary Recent advancements in weight‐loss medications have sparked a lot of interest. The so‐called GLP‐1 receptor agonist medications (GLP‐1RAs) have gained a lot of attention, because they have shown to be very effective, leading to significant weight loss in patients participating in clinical trials. GLP‐1RAs, like liraglutide, semaglutide, and tirzepatide, help manage weight by mimicking hormones that control blood sugar and appetite. However, how these medications perform in real life can be different from the controlled settings of clinical trials, in which patients are carefully selected and their treatment plans closely followed. This literature review looks at how these medications are used and their effectiveness and safety in real‐world settings. In real‐life practice, GLP‐1RAs are often less effective than in clinical trial conditions. This is usually because patients don't follow their medication plans as strictly as in trials. Real‐world data shows that many patients use lower doses and do not stick to their treatment as strictly as participants in a controlled trial might, leading to less weight loss. However, those who do follow their plans closely can achieve results similar to those in trials. A major issue with GLP‐1RAs is that many patients stop using them within the first year due to side effects or high costs of the medications, especially if not covered by insurance. Common side effects include nausea and digestive problems, which are the main reasons patients stop taking these treatments. These side effects are often manageable and decrease over time, and this reviews found no strong real‐world evidence that GLP‐1RAs cause severe side effects in many users. Despite these challenges, when GLP‐1RAs are used effectively and consistently, they show substantial benefits in weight loss, most so the newest medications semaglutide and tirzepatide. These medications are also likely to help manage and prevent weight‐related health conditions like type 2 diabetes and cardiovascular disease, but evidence for these beneficial outcomes is still scarce in real‐world settings. The review emphasizes the need for more research to understand why many patients stop using these medications and how to improve dosing. It also calls for studies on the long‐term effects of these therapies on various health outcomes, including mental health, cardiometabolic health, cancer, and rare conditions like eye diseases. Overall, while GLP‐1RAs are a valuable tool for weight management, their real‐world use requires careful consideration of individual patient factors, such as the ability to stick to treatment plans, manage side effects, and afford the medications. Further research will help make these treatments more effective for a wider range of people that need them.
Article
In rats, thyroid tumors are common age‐related findings with reported incidence rates up to 8.1% and 11.86% for follicular and C‐cell adenomas, respectively. Increases of thyroid follicular neoplasms in rodents via the induction of hepatic UDP‐glucuronosyltransferase (UGT) enzymes, resulting in elevated thyroid hormone (TH) metabolism, excretion, and subsequent follicular cell proliferation are generally accepted to have little or no relevance to humans due to species differences in sensitivity to this pathophysiologic process. In this analysis, we reviewed approved drugs that resulted in thyroid tumors in 2‐year rat carcinogenicity studies and summarized the positioning of these findings in product labeling language and human risk assessments in the United States and Europe. Overall, although thyroid follicular cell tumors are commonly observed, the labels reviewed listed no suspected human risk or directly state the absence of human relevance for these findings. Like follicular cell tumors, thyroid C‐cell tumors are common background findings in rats but comparatively are not as commonly increased in frequency as drug‐related findings in 2‐year rodent carcinogenicity studies. These findings are most notably observed with GLP‐1 agonists and their human relevance is a topic of ongoing clinical safety surveillance analysis. Thyroid follicular cell hyperplasia, when specifically occurring through hepatic enzyme induction and/or enhanced TH clearance, should be evaluated for anticipated human translational relevance using nonclinical and clinical data. If no human relevance is anticipated, this rationale should be incorporated into a weight of evidence approach for carcinogenicity studies as outlined in the ICH S1B addendum.
Article
Full-text available
Aim To compare (in the LIRA‐PRIME [NCT02730377], a randomized open‐label trial), the efficacy of liraglutide in controlling glycaemia versus an oral antidiabetic drug (OAD) in patients with uncontrolled type 2 diabetes (T2D), despite metformin use in a primary care setting (n = 219 sites, n = 9 countries). Materials and Methods Adults (n = 1991) with T2D (HbA1c 7.5%‐9.0%) receiving metformin were randomized 1:1 to liraglutide (≤1.8 mg/d) or one OAD, selected by the investigator, added to metformin, for up to 104 weeks. Primary endpoint: time to inadequate glycaemic control (HbA1c > 7.0%) at two scheduled consecutive visits after week 26. Outcomes were assessed for liraglutide versus a pooled OAD group, and (post hoc) liraglutide versus sodium‐glucose co‐transporter‐2 inhibitors, dipeptidyl peptidase‐4 inhibitors, and sulphonylureas individually. Results Among randomized patients (liraglutide, n = 996; OAD, n = 995), 47.6% were female, mean age was 57.4 years and mean HbA1c was 8.2%. Median time to inadequate glycaemic control was 44 weeks longer with liraglutide versus OAD (109 weeks [25% percentile, 38; 75% percentile, not available] vs. 65 weeks [25% percentile, 35; 75% percentile, 107], P < .0001). Changes in HbA1c and body weight at week 104 or at premature treatment discontinuation significantly favoured liraglutide over OAD. Hypoglycaemia rates were comparable between groups and few patients discontinued because of adverse events (liraglutide, 7.9% [n = 79]; OAD, 4.1% [n = 41]). Similar results were observed in the post hoc analysis for liraglutide versus individual OAD classes. Conclusions Glycaemic control was better maintained with liraglutide versus OAD, supporting liraglutide use when intensifying therapy in primary care patients with T2D.
Article
Full-text available
Importance Weight loss improves cardiometabolic risk factors in people with overweight or obesity. Intensive lifestyle intervention and pharmacotherapy are the most effective noninvasive weight loss approaches. Objective To compare the effects of once-weekly subcutaneous semaglutide, 2.4 mg vs placebo for weight management as an adjunct to intensive behavioral therapy with initial low-calorie diet in adults with overweight or obesity. Design, Setting, and Participants Randomized, double-blind, parallel-group, 68-week, phase 3a study (STEP 3) conducted at 41 sites in the US from August 2018 to April 2020 in adults without diabetes (N = 611) and with either overweight (body mass index ≥27) plus at least 1 comorbidity or obesity (body mass index ≥30). Interventions Participants were randomized (2:1) to semaglutide, 2.4 mg (n = 407) or placebo (n = 204), both combined with a low-calorie diet for the first 8 weeks and intensive behavioral therapy (ie, 30 counseling visits) during 68 weeks. Main Outcomes and Measures The co–primary end points were percentage change in body weight and the loss of 5% or more of baseline weight by week 68. Confirmatory secondary end points included losses of at least 10% or 15% of baseline weight. Results Of 611 randomized participants (495 women [81.0%], mean age 46 years [SD, 13], body weight 105.8 kg [SD, 22.9], and body mass index 38.0 [SD, 6.7]), 567 (92.8%) completed the trial, and 505 (82.7%) were receiving treatment at trial end. At week 68, the estimated mean body weight change from baseline was –16.0% for semaglutide vs –5.7% for placebo (difference, −10.3 percentage points [95% CI, −12.0 to −8.6]; P < .001). More participants treated with semaglutide vs placebo lost at least 5% of baseline body weight (86.6% vs 47.6%, respectively; P < .001). A higher proportion of participants in the semaglutide vs placebo group achieved weight losses of at least 10% or 15% (75.3% vs 27.0% and 55.8% vs 13.2%, respectively; P < .001). Gastrointestinal adverse events were more frequent with semaglutide (82.8%) vs placebo (63.2%). Treatment was discontinued owing to these events in 3.4% of semaglutide participants vs 0% of placebo participants. Conclusions and Relevance Among adults with overweight or obesity, once-weekly subcutaneous semaglutide compared with placebo, used as an adjunct to intensive behavioral therapy and initial low-calorie diet, resulted in significantly greater weight loss during 68 weeks. Further research is needed to assess the durability of these findings. Trial Registration ClinicalTrials.gov Identifier: NCT03611582
Article
Full-text available
Objective Previous studies have shown additive weight loss when intensive behavioral therapy (IBT) was combined with weight‐loss medication. The present multisite study provides the first evaluation, in primary care, of the effect of the Centers for Medicare and Medicaid Services–based IBT benefit, delivered alone (with placebo) or in combination with liraglutide 3.0 mg. Methods The Satiety and Clinical Adiposity—Liraglutide Evidence in individuals with and without diabetes (SCALE) IBT was a 56‐week, randomized, double‐blind, placebo‐controlled, multicenter trial in individuals with obesity who received liraglutide 3.0 mg (n = 142) or placebo (n = 140) as an adjunct to IBT. Results At week 56, mean weight loss with liraglutide 3.0 mg plus IBT was 7.5% and 4.0% with placebo combined with IBT (estimated treatment difference [95% CI]–3.4% [–5.3% to –1.6%], P = 0.0003). Significantly more individuals on liraglutide 3.0 mg than placebo achieved ≥ 5% weight loss (61.5% vs. 38.8%; odds ratio [OR] 2.5% [1.5% to 4.1%], P = 0.0003), > 10% weight loss (30.5% vs. 19.8%; OR 1.8% [1.0% to 3.1%], P = 0.0469), and > 15% weight loss (18.1% vs. 8.9%; OR 2.3% [1.1% to 4.7%], P = 0.0311). Liraglutide 3.0 mg in combination with IBT was well tolerated, with no new safety signals identified. Conclusions In a primary care setting, Centers for Medicare and Medicaid Services–based IBT produced clinically meaningful weight loss at 56 weeks, enhanced by the addition of liraglutide 3.0 mg.
Article
BACKGROUND Four glucagon-like peptide-1 (GLP-1) receptor agonists that are structurally similar to human GLP-1 have been shown to reduce the risk of adverse cardiovascular events among persons with type 2 diabetes. The effect of an exendin-based GLP-1 receptor agonist, efpeglenatide, on cardiovascular and renal outcomes in patients with type 2 diabetes who are also at high risk for adverse cardiovascular events is uncertain. METHODS In this randomized, placebo-controlled trial conducted at 344 sites across 28 countries, we evaluated efpeglenatide in participants with type 2 diabetes and either a history of cardiovascular disease or current kidney disease (defined as an estimated glomerular filtration rate of 25.0 to 59.9 ml per minute per 1.73 m2 of body-surface area) plus at least one other cardiovascular risk factor. Participants were randomly assigned in a 1:1:1 ratio to receive weekly subcutaneous injections of efpeglenatide at a dose of 4 or 6 mg or placebo. Randomization was stratified according to use of sodium–glucose cotransporter 2 inhibitors. The primary outcome was the first major adverse cardiovascular event (MACE; a composite of nonfatal myocardial infarction, nonfatal stroke, or death from cardiovascular or undetermined causes). RESULTS A total of 4076 participants were enrolled; 2717 were assigned to receive efpeglenatide and 1359 to receive placebo. During a median follow-up of 1.81 years, an incident MACE occurred in 189 participants (7.0%) assigned to receive efpeglenatide (3.9 events per 100 person-years) and 125 participants (9.2%) assigned to receive placebo (5.3 events per 100 person-years) (hazard ratio, 0.73; 95% confidence interval [CI], 0.58 to 0.92; P<0.001 for noninferiority; P = 0.007 for superiority). A composite renal outcome event (a decrease in kidney function or macroalbuminuria) occurred in 353 participants (13.0%) assigned to receive efpeglenatide and in 250 participants (18.4%) assigned to receive placebo (hazard ratio, 0.68; 95% CI, 0.57 to 0.79; P<0.001). Diarrhea, constipation, nausea, vomiting, or bloating occurred more frequently with efpeglenatide than with placebo. CONCLUSIONS In this trial involving participants with type 2 diabetes who had either a history of cardiovascular disease or current kidney disease plus at least one other cardiovascular risk factor, the risk of cardiovascular events was lower among those who received weekly subcutaneous injections of efpeglenatide at a dose of 4 or 6 mg than among those who received placebo.
Article
SGLT2 inhibitors and GLP-1 receptor agonists are used in patients with type 2 diabetes as glucose lowering therapies, with additional benefits of weight loss and blood pressure reduction. Data from cardiovascular outcome trials have highlighted that these drugs confer protection against major cardiovascular disease in those with established atherosclerotic cardiovascular disease, reduce the risk of admission to hospital for heart failure, and reduce cardiovascular and all-cause mortality. Ongoing research using hard renal endpoints such as end stage kidney disease rather than surrogate markers might clarify the renoprotective benefits of both agents. When used for glucose lowering, SGLT2 inhibitors are most effective if the estimated glomerular filtration rate is more than 60 ml per min per 1·73m² at initiation and should be avoided where there is a risk of diabetic ketoacidosis. GLP-1 receptor agonists are contraindicated in those with a history of medullary thyroid cancer and used with caution in patients with a history of pancreatitis of a known cause. These drugs are now second-line, or even arguably first-line, glucose lowering therapies in patients with cardiorenal disease, irrespective of glycaemic control. If an SGLT2 inhibitor or GLP-1 receptor agonist is considered suitable in patients with type 2 diabetes, treatment should be prioritised according to existing evidence: GLP-1 receptor agonists should be considered in patients at a high risk of, or with established, cardiovascular disease and SGLT2 inhibitors considered for patients with heart failure (with reduced ejection fraction) or chronic kidney disease (with or without established cardiovascular disease). There is now compelling data on the benefits of these drugs for a range of other clinical indications even without type 2 diabetes, including for GLP-1 receptor agonists in patients with obesity and overweight with weight-related comorbidities.
Article
Background Obesity is a global health challenge with few pharmacologic options. Whether adults with obesity can achieve weight loss with once-weekly semaglutide at a dose of 2.4 mg as an adjunct to lifestyle intervention has not been confirmed. Methods In this double-blind trial, we enrolled 1961 adults with a body-mass index (the weight in kilograms divided by the square of the height in meters) of 30 or greater (≥27 in persons with ≥1 weight-related coexisting condition), who did not have diabetes, and randomly assigned them, in a 2:1 ratio, to 68 weeks of treatment with once-weekly subcutaneous semaglutide (at a dose of 2.4 mg) or placebo, plus lifestyle intervention. The coprimary end points were the percentage change in body weight and weight reduction of at least 5%. The primary estimand (a precise description of the treatment effect reflecting the objective of the clinical trial) assessed effects regardless of treatment discontinuation or rescue interventions. Results The mean change in body weight from baseline to week 68 was −14.9% in the semaglutide group as compared with −2.4% with placebo, for an estimated treatment difference of −12.4 percentage points (95% confidence interval [CI], −13.4 to −11.5; P<0.001). More participants in the semaglutide group than in the placebo group achieved weight reductions of 5% or more (1047 participants [86.4%] vs. 182 [31.5%]), 10% or more (838 [69.1%] vs. 69 [12.0%]), and 15% or more (612 [50.5%] vs. 28 [4.9%]) at week 68 (P<0.001 for all three comparisons of odds). The change in body weight from baseline to week 68 was −15.3 kg in the semaglutide group as compared with −2.6 kg in the placebo group (estimated treatment difference, −12.7 kg; 95% CI, −13.7 to −11.7). Participants who received semaglutide had a greater improvement with respect to cardiometabolic risk factors and a greater increase in participant-reported physical functioning from baseline than those who received placebo. Nausea and diarrhea were the most common adverse events with semaglutide; they were typically transient and mild-to-moderate in severity and subsided with time. More participants in the semaglutide group than in the placebo group discontinued treatment owing to gastrointestinal events (59 [4.5%] vs. 5 [0.8%]). Conclusions In participants with overweight or obesity, 2.4 mg of semaglutide once weekly plus lifestyle intervention was associated with sustained, clinically relevant reduction in body weight. (Funded by Novo Nordisk; STEP 1 ClinicalTrials.gov number, NCT03548935).
Article
Glucagon-like peptide-1 (GLP-1) is produced in gut endocrine cells and in the brain, and acts through hormonal and neural pathways to regulate islet function, satiety, and gut motility, supporting development of GLP-1 receptor (GLP-1R) agonists for the treatment of diabetes and obesity. Classic notions of GLP-1 acting as a meal-stimulated hormone from the distal gut are challenged by data supporting production of GLP-1 in the endocrine pancreas, and by the importance of brain-derived GLP-1 in the control of neural activity. Moreover, attribution of direct vs. indirect actions of GLP-1 is difficult, as many tissue and cellular targets of GLP-1 action do not exhibit robust or detectable GLP-1R expression. Furthermore, reliable detection of the GLP-1R is technically challenging, highly method-dependent, and subject to misinterpretation. Here we revisit the actions of GLP-1, scrutinizing key concepts supporting gut vs. extra-intestinal GLP-1 synthesis and secretion. We discuss new insights refining cellular localization of GLP-1R expression and integrate recent data to refine our understanding of how and where GLP-1 acts to control inflammation, cardiovascular function, islet hormone secretion, gastric emptying, appetite, and body weight. These findings update our knowledge of cell types and mechanisms linking endogenous vs. pharmacological GLP-1 action to activation of the canonical GLP-1R, and the control of metabolic activity in multiple organs.
Article
What is known and objective: The use of glucagon-like peptide-1 (GLP-1) analogues has been linked to the risk of thyroid cancer. Spontaneous reports can provide information about rare adverse events occurring after the time of marketing. Our objective was to detect, from the European pharmacovigilance database (EudraVigilance), a signal of thyroid cancer during GLP-1 analogues treatment in patients with diabetes. Methods: Herein, we analysed all reports of thyroid cancer reported with GLP-1 analogues in EudraVigilance database from their first marketing authorization till 30 January 2020. A case/non-case method was used to assess the association between thyroid cancer and GLP-1 analogues, calculating proportional reporting ratios (PRRs) and their 95% confidence interval (CI) as a measure of disproportionality. The cases were identified with Medical Dictionary for Regulatory Activities (MedDRA) version 22.1. Results and discussion: There were 11 243 cases of thyroid cancer and related preferred terms (PTs) in the 6 665 794 reports recorded in EudraVigilance during the study period. GLP-1 analogues were involved in 236 cases. Exenatide, liraglutide and dulaglutide met the criteria to generate a safety signal, suggesting that thyroid cancer is reported relatively more frequently in association with these drugs than with other medicinal products. The association was strongest for liraglutide followed by exenatide with PRR of 27.5 (95% CI, 22.7-33.3) and 22.5 (95% CI, 17.9-28.3), respectively. Disproportionality was also observed for GLP-1 analogues and individual identified preferred term, that is thyroid cancer (N = 111), medullary thyroid cancer (N = 64) and thyroid neoplasm (N = 46) with PRR of 14.4 (95% CI, 11.8-17.4), 221.5 (95% CI, 155.7-315.1) and 35.5 (95% CI, 25.9-48.5), respectively. What is new and conclusions: Our findings showed disproportionality for thyroid cancer, medullary thyroid cancer and thyroid neoplasm in patients treated with GLP-1 analogues. We have found evidence from spontaneous reports that GLP-1 analogues are associated with thyroid cancer in patients with diabetes.
Article
Background Given the unique phenotype of type 2 diabetes in Japanese patients, novel therapies such as oral semaglutide require evaluation in this population. PIONEER 9 aimed to assess the dose-response of oral semaglutide and to compare the efficacy and safety of oral semaglutide with placebo and a subcutaneous GLP-1 receptor agonist in a Japanese population. Methods PIONEER 9 was a 52-week, phase 2/3a, randomised, controlled trial done at 16 sites (clinics and university hospitals) in Japan. Japanese patients aged 20 years or older with uncontrolled type 2 diabetes managed by diet or exercise or with oral glucose-lowering drug monotherapy (washed out) were randomly assigned (1:1:1:1:1) to receive double-blind once-daily oral semaglutide (3 mg, 7 mg, or 14 mg) or placebo, or open-label subcutaneous once-daily liraglutide 0·9 mg. The primary endpoint was change in HbA1c from baseline to week 26 with the trial product (primary) estimand (which assumes all patients remained on trial product without rescue medication use) in all randomly assigned patients. This trial is registered with ClinicalTrials.gov, NCT03018028. Findings Between Jan 10, and July 11, 2017, 243 patients were randomly assigned to oral semaglutide 3 mg (n=49), 7 mg (n=49), or 14 mg (n=48), or placebo (n=49), or to liraglutide 0·9 mg (n=48). Changes in HbA1c from baseline (mean 8·2%) to week 26 were dose-dependent with oral semaglutide (mean change −1·1% [SE 0·1] for oral semaglutide 3 mg, −1·5% [0·1] for 7 mg, and −1·7% [0·1] for 14 mg), −0·1% (0·1) with placebo, and −1·4% (0·1) with liraglutide 0·9 mg. Estimated treatment differences for change in HbA1c compared with placebo were −1·1 percentage points (95% CI −1·4 to −0·8; p<0·0001) for oral semaglutide 3 mg, −1·5 percentage points (–1·7 to −1·2; p<0·0001) for oral semaglutide 7 mg, and −1·7 percentage points (–2·0 to −1·4; p<0·0001) for oral semaglutide 14 mg. Estimated treatment differences for change in HbA1c compared with liraglutide 0·9 mg were 0·3 percentage points (95% CI −0·0 to 0·6; p=0·0799) for oral semaglutide 3 mg, −0·1 percentage points (–0·4 to 0·2; p=0·3942) for oral semaglutide 7 mg, and −0·3 percentage points (–0·6 to −0·0; p=0·0272) for oral semaglutide 14 mg. Gastrointestinal events, predominantly of mild or moderate severity, were the most frequently reported class of adverse event with oral semaglutide: constipation was most common, occurring in five to six (10–13%) patients with oral semaglutide, three (6%) with placebo, and nine (19%) with liraglutide 0·9 mg. Interpretation This study showed that oral semaglutide provides significant reductions in HbA1c compared with placebo in a dose-dependent manner in Japanese patients with type 2 diabetes, and has a safety profile consistent with that of GLP-1 receptor agonists. Funding Novo Nordisk.
Article
Objective: Most individuals with type 2 diabetes also have obesity, and treatment with some diabetes medications, including insulin, can cause further weight gain. No approved chronic weight-management medications have been prospectively investigated in individuals with overweight or obesity and insulin-treated type 2 diabetes. The primary objective of this study was to assess the effect of liraglutide 3.0 mg versus placebo on weight loss in this population. Research design and methods: Satiety and Clinical Adiposity-Liraglutide Evidence (SCALE) Insulin was a 56-week, randomized, double-blind, placebo-controlled, multinational, multicenter trial in individuals with overweight or obesity and type 2 diabetes treated with basal insulin and less than or equal to two oral antidiabetic drugs. Results: Individuals were randomized to liraglutide 3.0 mg (n = 198) or placebo (n = 198), combined with intensive behavioral therapy (IBT). At 56 weeks, mean weight change was -5.8% for liraglutide 3.0 mg versus -1.5% with placebo (estimated treatment difference -4.3% [95% CI -5.5; -3.2]; P < 0.0001). With liraglutide 3.0 mg, 51.8% of individuals achieved ≥5% weight loss versus 24.0% with placebo (odds ratio 3.41 [95% CI 2.19; 5.31]; P < 0.0001). Liraglutide 3.0 mg was associated with significantly greater reductions in mean HbA1c, mean daytime glucose values, and less need for insulin versus placebo, despite a treat-to-glycemic target protocol. More hypoglycemic events were observed with placebo than liraglutide 3.0 mg. No new safety or tolerability issues were observed. Conclusions: In individuals with overweight or obesity and insulin-treated type 2 diabetes, liraglutide 3.0 mg as an adjunct to IBT was superior to placebo regarding weight loss and improved glycemic control despite lower doses of basal insulin and without increases in hypoglycemic events.