ArticlePDF Available

Abstract and Figures

As the tide of obesity and its complications is on the rise, there is an urgent need for new drugs with weight-lowering and beneficial metabolic properties. Obesity-related disorders, such as metabolic syndrome, prediabetes, type 2 diabetes mellitus (T2DM), cardiovascular disease and non-alcoholic fatty liver disease (NAFLD) make this need more than mandatory. Sodium-Glucose Co-Transporter-2 (SGLT-2) inhibitors (empagliflozin, canagliflozin, dapagliflozin, and ertugliflozin) are the latest class of agents to receive approval for the treatment of T2DM. Not long after their marketing, a wide spectrum of target organ-protective and overall beneficial health effects associated with their use began to unveil. An increasing bulk of evidence points towards that these actions are to a great degree independent of glucose lowering, which has led to the broadening of the indications of SGLT-12 inhibitors outside the frame of antihyperglycemic therapy. Additionally, their unique mode of action including an increased renal glucose excretion and hence, net energy loss could render SGLT-2 inhibitors attractive candidates for the treatment of obesity. Very few reviews in the literature have appraised holistically the therapeutic potential of SGLT-2 inhibitors in obesity and its associated complications. Herein, we synopsize the currently available evidence regarding the effects of drugs of this class on body adiposity, together with considerations which accompany their potential use with the aim of weight loss. Furthermore, we attempt an overview of their actions and future perspectives of their use with respect to a range of obesity-related disorders, which include cardiovascular, renal, as well as NAFLD, malignancy, and ovarian dysfunction.
Content may be subject to copyright.
REVIEW ARTICLE SGLT ‑2 inhibitors in obesity and its comorbidities 1
those of obesity, and anestimated 422 million
individuals were affected as of2014. estrong
causal relationship between these2 conditions
is mediated through theinteraction of avariety
of genetic and environmental factors that culmi
nate in thedevelopment of systemic insulin resis
tance and eventually β ‑cell failure; hence there‑
cently coined term “diabesity.7-9
As thetide of obesity and its complications
is on therise, there is anurgent need for new
drugs with weight ‑lowering and beneficial met‑
abolic properties.
10
Obesity and obesity ‑related
Introduction
For thelast few decades obesity has
constituted agrowing worldwide public health is
sue that affects therisk and prognosis of sever‑
al conditions, including cardiovascular disease
(CVD), metabolic syndrome (MetS), type 2 di
abetes (T2D), non alcoholic fatty liver disease
(NAFLD), COVID ‑19, and cancer.
1-5
According to
theWorld Health Organization data, in 2016 more
than 1.9 billion adults were overweight (body
mass index [BMI] between 25 and 30kg/m
2
),
among whom more than 650 million were obese
(BMI>30kg/m2).6 Global trends in T2D parallel
REVIEW ARTICLE
Sodium ‑glucose co transporter ‑2 inhibitors
in obesity and associated cardiometabolic
disorders: where do we stand?
NataliaG.Vallianou
1*
, DimitriosTsilingiris
2*
, DimitrisKounatidis
1
,
IoannisG.Lempesis
3,4
, IreneKarampela
5
, MariaDalamaga
6
1 Department of Internal Medicine and Endocrinology, Evangelismos General Hospital, Athens, Greece
2 First Department of Propaedeutic Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
3 Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
4 Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, the Netherlands
5 Second Department of Critical Care, Medical School, University of Athens, Attikon General University Hospital, Chaidari, Athens, Greece
6 Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
Correspondence to:
Maria Dalamaga, MD, MSc, MPH,
PhD, National and Kapodistrian
University of Athens, Medical
School, Mikras Asias 27,
Goudi, 11527 Athens, Greece,
phone: +302 107 462 624,
email: madalamaga@med.uoa.gr
Received: August 23, 2022.
Accepted: September 12, 2022.
Published online:
September 12, 2022.
Pol Arch Intern Med. 2022;
132 (10): 16342
doi:10.20452/pamw.16342
Copyright by the Author(s), 2022
* NGV and DT equally contributed to
this work.
KEY WORDS
cardiometabolic
disorders,
cardiovascular
disease, diabetes,
obesity, SGLT ‑2
inhibitors
ABSTRACT
As the tide of obesity and its complications are on the rise, there is an urgent need for new drugs with
weight ‑lowering and beneficial metabolic properties. Obesity ‑related disorders, such as metabolic syn
drome, prediabetes, type 2 diabetes (T2D), cardiovascular disease, and nonalcoholic fatty liver disease
(NAFLD) make this need more than mandatory. Sodium ‑glucose cotransporter ‑2 (SGLT ‑2) inhibitors (em
pagliflozin, canagliflozin, dapagliflozin, and ertugliflozin) are the latest class of agents to receive approval
for the treatment of T2D. Not long after their marketing, a wide spectrum of target organ ‑protective and
overall beneficial health effects associated with their use began to unveil. An increasing bulk of evidence
indicates that these actions are to a great degree independent of glucose lowering, which has led to
the broadening of the indications for SGLT ‑2 inhibitors outside the frame of antihyperglycemic therapy.
Additionally, their unique mode of action including increased renal glucose excretion, and hence net
energy loss, could render SGLT ‑2 inhibitors attractive candidates for the treatment of obesity. Very few
reviews in the literature have holistically appraised the therapeutic potential of SGLT ‑2 inhibitors in obe
sity and its associated complications. Herein, we summarize the currently available evidence regarding
the effects of drugs of this class on body adiposity, together with considerations on their potential use
as weight loss agents. Furthermore, we attempt to overview their actions and future perspectives of
their use with respect to a range of obesity ‑related disorders, which include cardiovascular, renal, and
ovarian dysfunctions, as well as NAFLD and malignancy.
POLISH ARCHIVES OF INTERNAL MEDICINE 2022; 132 (10)2
chronic kidney disease (n = 20), type 1 diabetes
(n = 15), COVID ‑19 (n = 10), prediabetes (n = 9),
dual inhibition of SGLT ‑1 and SGLT ‑2 (n = 8),
merely heart failure (n = 6), merely hyperten
sion (n = 5), asthma and obstructive sleep apnea
(n = 3), or neurological diseases (n = 3), and there
maining 11 studies were not written in English
(3 studies in Japanese, 2 in Spanish, 2 in French,
1 in Polish, 1 in Russian, 1 in Swedish, and 1 in He
brew). In addition, there were 6 books and docu
ments that were excluded, leaving atotal of 601
studies included in this search.
Mechanisms of action of sodium -glucose cotransporter -2
inhibitors Animportant milestone in thecourse
of SGLT ‑2‑inhibition–based therapy was thedis
covery of theantihyperglycemic effects of phlori
zin, asubstance isolated from thebark of anap‑
ple tree by Josef von Mering in1886. Although
he additionally postulated that thekidneys are its
pharmacological target, it was not until the1970s
that inhibition of renal tubular glucose reabsorp
tion was specified as themechanism of action of
phlorizin,16 while Rossetti et al17 demonstrat
ed amelioration of insulin resistance and hyper‑
glycemia after phlorizin administration in1987.
edevelopment of thefirst orally absorbable,
phlorizin ‑derived SGLT ‑1 and SGLT ‑2 inhibitor
was followed by arapid discovery of more oral
ly active agents, and in 2013 canagliflozin was
thefirst SGLT‑2 inhibitor that received Food and
Drug Administration (FDA) approval for thetreat
ment of T2D, followed by dapagliflozin and em‑
pagliflozin in 2014.
18
SGLT cotransporters are di
vided into 2 categories: SGLT ‑1 and SGLT ‑2 co
transporters. SGLT ‑1 cotransporters are mainly
located in thesmall intestine and are responsi
ble for glucose absorption there and for there
absorption of approximately 10% of thefiltered
glucose in theupper part of therenal proximal tu
bule. eir major mechanism of action is to delay
glucose absorption in thesmall intestine, leading
to adecrease in theserum postprandial glucose
levels.19 SGLT ‑1 cotransporters are also located
in thekidneys, thebrain, theheart, thetrachea,
thetestis, and theprostate gland.19 On thecon‑
trary, SGLT ‑2 cotransporters are mainly found
in theproxy part of therenal proximal tubule,
where they act by reabsorbing approximately 90%
of thefiltered glucose. Apart from thekidneys,
SGLT ‑2 cotransporters are expressed in thebrain,
theheart, theliver, thethyroid gland, themus‑
cles, and theα pancreatic cells. eir major func
tion in thekidneys is to impede renal glucose re‑
absorption, causing glucosuria.2 0,21
Apart from lowering serum glucose levels,
SGLT ‑2 inhibitors have been documented to pro
vide significant cardiovascular benefits in patients
with T2D. Until now, there are 4 selective SGLT ‑2
inhibitors with demonstrated cardiovascular ben
efit: empagliflozin, canagliflozin, dapagliflozin,
and ertugliflozin.
22
eabovementioned drugs
have been documented to control blood glucose
levels, as well as to decrease body weight and
disorders, such as MetS, prediabetes, T2D,
NAFLD, and CVD complications, as well as thein
creased prevalence of certain types of cancer make
this need more than mandatory. Lifestyle modifi
cations, such as decreased calorie intake and in‑
creased physical activity play akey role in com‑
bating obesity but are not always very easy to pur
sue.11 ere are several weight‑lowering drugs,
mainly lorcaserine, phentermine, topiramate, and
glucagon ‑like peptide ‑1 (GLP ‑1) receptor agonists,
but their use is restricted by their adverse effects
and limited effectiveness. Bariatric surgery offers
amore drastic and more lasting weight‑lowering
potential, and it may reverse prediabetes and T2D
in asignificant proportion of patients.10 Howev
er, bariatric surgery, while offering theeffective
solution regarding severe obesity, is also associ‑
ated with severe adverse effects.12,13
Sodium ‑glucose cotransporter ‑2 (SGLT ‑2) in‑
hibitors are arelatively new class of antidiabetic
drugs, which act atthe level of therenal proximal
tubule, causing glucosuria. Apart from theglu
cosuric effects, they seem to exert pleiotropic bi
ological effects, which are not directly attributable
to thereduction of hyperglycemia, such as there
duction of cardiovascular mortality, heart failure
(HF) hospitalizations, and hard renal outcomes.
is has led to agradual generalization of thein
dications for individual SGLT ‑2 inhibitors outside
of theframe of antihyperglycemic therapy or car
diovascular risk reduction among patients with
T2D. ey are now indicated for patients with HF
with preserved (empagliflozin) and reduced ejec
tion fraction (EF) (empagliflozin, dapagliflozin),
as well as chronic kidney disease (CKD) of etiol‑
ogy other than diabetic kidney disease, such as
ischemic or immunoglobulin Anephropathy, fo‑
cal segmental glomerulosclerosis, chronic pyelo‑
nephritis, and chronic interstitial nephritis (dapa
gliflozin).
14
Furthermore, their unique mode of
action, which results in aglucosuria ‑induced net
caloric loss, renders theagents of these category
attractive candidates for obesity therapy.
Very few reviews in theliterature have dis
cussed holistically thetherapeutic potential of
SGLT ‑2 inhibitors in obesity and its associated
complications.
15
In this narrative review, we aim
to 1) present themechanisms of action of SGLT ‑2
inhibitors, with aspecial focus on obesity and its
associated disorders; 2) appraise their therapeutic
applications; 3) discuss adverse effects and toler
ability issues, and 4) review potential future per
spectives and challenges.
Literature search In August 2022, aliterature
search of 2 bibliographical databases (MEDLINE
and Scopus) was conducted to assess theeffects
of SGLT ‑2 inhibitors on obesity. is search used
thefollowing terms: “sglt2 inhibitors” and “obe‑
sity.” esearch for theabovementioned terms
yielded atotal of 697 papers, most of which (539
results) were published between 2017 and 2022
(during thepast 5 years). Of these 697 studies,
90 were excluded, as 79 dealt with issues such as
REVIEW ARTICLE SGLT ‑2 inhibitors in obesity and its comorbidities 3
clinically relevant loss of 0.8kg under SGLT ‑2 in
hibitor monotherapy, or up to 5.7kg when SGLT ‑2
inhibitors were combined with GLP ‑1 receptor
agonists or medications including sulfonylureas.
ese findings are consistent in patients with
and without T2D and are attributable to anet
fat mass loss in studies that included measures
of body composition estimates, most common
ly magnetic resonance tomography. Additional
ly, thetherapy with SGLT ‑2 inhibitors is well tol
erated and adverse events are scarce.
Furthermore, SGLT ‑2 inhibitors reduce body
weight by interfering with excess adipose tissue,
which is known to synthesize inflammatory ad‑
ipocytokines.28-32 In obesity and obesity ‑related
disorders, such as NAFLD and T2D, adipose tissue
macrophages exhibit polarization toward theM1
phenotype, which produces proinflammatory cy
tokines, such as tumor necrosis factor α (TNF ‑α)
and interleukin (IL)‑6, thus inducing alow ‑grade
inflammatory state.
33,34
On thecontrary, theM2
phenotype is restricted in obesity and obesity‑
‑related disorders, thereby resulting in mitigation
of theanti ‑inflammatory cytokines, such as IL ‑4
and IL ‑10.
35-37
SGLT ‑2 inhibitors have been docu
mented to reverse thepolarization of adipocytes
from type 1 macrophages (M1), which release pro‑
inflammatory cytokines, to type 2 macrophages
(M2), which produce anti ‑inflammatory cyto
kines.24,38 is increase in the M2 phenotype ac‑
counts for thebeneficial effects of SGLT ‑2 inhibi
tors regarding obesity, and is suggested to reduce
thechronic inflammatory state, which character
izes and promotes obesity. By bolstering theM2
phenotype, SGLT ‑2 inhibitors suppress this chron
ic inflammation, and thus induce weight loss.
36,39
reduce systolic and diastolic blood pressure.
23
FIGURE 1 summarizes themain biological actions
of SGLT‑2 inhibitors, which are considered to
contribute to their cardiorenal protective effects.
Effects on body wei ght and adiposity indexes SGLT ‑2
inhibitors cause glucosuria, and thus they alone
induce weight loss of approximately 1.5–2 kg.
is weight loss phenomenon is dose ‑dependent
and may be maximized, when combining oth
er types of anti diabetic drugs, especially GLP ‑1
analogs, which suppress appetite by acting di
rectly atthe hypothalamus level.24 Notably,
Ferrannini et al25 demonstrated adisproportion
ate decrease in body weight induced by SGLT ‑2
inhibitors used alone due to their glucosuric ef‑
fects. In particular, SGLT ‑2 inhibitors produce
asmaller weight loss than can be expected based
on their glucosuric potential. is discrepancy
could be attributed to anincrease in energy in
take as anadaptive mechanism of thebody to
prevent any further changes in body weight. Ac‑
cording to Ferrannini et al,
25
thehuman body
might have developed anadaptive enhancement
in appetite in aneffort to induce stability in body
weight to counterbalance theweight loss effects
of SGLT ‑2 inhibitors. is notion is also increas‑
ingly being supported by other researchers.
24-26
erefore, thecombined use of SGLT ‑2 inhibitors
with drugs suppressing theappetite atthe hypo
thalamus level, such as GLP ‑1 analogs, is gaining
much interest nowadays.
24,27
TABLE 1 presents main
studies linking SGLT ‑2 inhibitors with weight loss
among patients with and without T2D. In short,
theresults from different studies using awide se
lection of agents have demonstrated amodest but
FIGURE 1 Overview of
biological actions of
sodium ‑glucose
cotransporter ‑2 inhibitors,
which may drive their
cardiovascular and renal
protective effects based
on Braunwald23 and
Tuttle et al100
• Blood pressure normalization
• Increased erythrocyte mass
• Promotion of ketogenesis
• Amelioration of insulin
resistance
• Increased glucagon levels
• Plasma volume reduction
• Increased erythropoietin activity
• Reduced plasma glucose levels
• Weight loss
• Increased circulating fatty acids
• Increased urinary sodium
and glucose excretion
• Augmentation of
glomerulotubular reflex
• Reduced tubular workload
• Reduced left ventricular
wall stress
• Promotion of ketone
body oxidation
• Increased oxygen delivery
• Reduced sodium-hydrogen
exchanger-3 activity
POLISH ARCHIVES OF INTERNAL MEDICINE 2022; 132 (10)4
TABLE 1 List of studies associating overweight / obesity and the effects of sodium glucose cotransporter ‑2 inhibitors on body weight among patients with and without type 2 diabetes (continued on the next pages)
Research / year Population, type of study Treatment Main findings Remarks
List of studies among patients without T2D
Hussey et al,(S1)a 2010 18 overweight / obese patients without T2D;
A double ‑blind, placebo ‑controlled,
randomized, phase 1 trial
I) Sergliflozin etabonate 500 mg/tid
II) Sergliflozin etabonate 1000 mg/tid
III) Placebo
Sergliflozin induced significant weight
loss of approximately 1.55 kg for
sergluflozin 500 mg/tid to 1.74 kg for
sergluflozin 1000 mg/tid, from baseline
after 15 days of administration
Sergluflozin was generally well tolerated;
No major adverse effects reported
Bays et al,54 2014 376 overweight / obese patients without
T2D;
A double ‑blind, placebo ‑controlled, phase 2b
trial
I) Canagliflozin 50 mg
II) Canagliflozin 100 mg
III) Canagliflozin 300 mg
IV) Placebo
body weight by
I) 2.2%
II) 2.9%
III) 2.7%
IV) 1.3%
Canagliflozin body weight without any severe
adverse effects
Napolitano et al,(S2)
2014
30 overweight / obese patients without T2D;
A double ‑blind, placebo ‑controlled,
randomized, pilot trial
I) Sergliflozin etabonate 1000 mg/tid
II) Remogliflozin etabonate 250 mg/tid
III) Placebo
No changes in body weight reported in
the 3 groups;
All patients except 1 exhibited weight
loss
in the leptin / adiponectin ratio in the groups receiving
sergliflozin or remogliflozin when compared with
placebo
Lundkvist et al,(S3)
2017a
50 obese patients without T2D;
A randomized, placebo‑controlled, phase 2a
trial
I) Dapagliflozin 10 mg/d + exanetide 2 mg sc
every week for 24 weeks
II) Placebo
body weight by 4.13 kg (SD) (95% CI,
6.44–1.81 kg; P <0.001);
in total adipose tissue by 3.8 l
36% of the patients enrolled lost 5% of their body
weight;
The loss in body weight was attributed to in adipose
tissue with no in lean tissue, as estimated by MRI
Lundkvist et al,(S4)
2017b
50 obese adults without T2D;
Open ‑label extension trial
I) Dapagliflozin 10 mg/d + exanetide 2 mg sc
every week for 1 year
II) Placebo
body weight by 5.7 kg;
in total adipose tissue by 5.3 l
body weight lasting for 1 year;
in adipose tissue, as estimated by MRI
Hollander et al,(S5)
2017
335 overweight / obese patients without
T2D;
A double ‑blind, placebo ‑controlled, phase 2a
trial
I) Canagliflozin 300 mg/d
II) Phentermine 15 mg/d
III) Canagliflozin 300 mg/d + phentermine
15 mg/d
IV) Placebo
body weight by 6.9% [95% CI,
8.6%–5.2%], P <0.001 in
the combination group
body weight 5%, without any severe adverse
effects
Ramirez ‑Rodriguez
et al,(S6) 2020
24 patients with prediabetes;
A double ‑blind, placebo ‑controlled,
randomized trial
I) Dapagliflozin 10 mg/d
II) Placebo
body weight by 3.0 kg or 3.7% vs 1 kg
in placebo (P = 0.019)
body weight;
BMI;
WC
Faecher et al,(S7) 2021 120 overweight / obese patients with
prediabetes;
A controlled, randomized, parallel ‑arm,
non ‑blind, open label trial
I) Metformin 1700 mg/d
II) Dapagliflozin 10 mg/d
III) Placebo
IV) Exercise group
Dapagliflozin and exercise led to small
improvements in glycemic control
among patients with prediabetes, when
compared with metformin and placebo
The study revealed uncertainty about glycemic control
in prediabetic patients;
The study did not mention any differences in body
weight between the groups
List of studies in patients with T2D
Ferrannini et al(S8),
2010
485 patients with T2D;
A double ‑blind, placebo ‑controlled
randomized, phase 3 trial
I) Dapagliflozin 2.5 mg/d
II) Dapagliflozin 5 mg/d
III) Dapagliflozin 10 mg/d
IV) Placebo
in mean HbA1c of 0.58%, 0.77%, 0.89%
in group I, II, and III, respectively,
P <0.0005;
in mean HbA1c of 0.23% in the placebo
group
No severe adverse effects reported;
Noninsulin‑dependent mode of action
REVIEW ARTICLE SGLT ‑2 inhibitors in obesity and its comorbidities 5
TABLE 1 List of studies associating overweight / obesity and the effects of sodium glucose cotransporter ‑2 inhibitors on body weight among patients with and without type 2 diabetes (continued on the next pages)
Research / year Population, type of study Treatment Main findings Remarks
Strojek et al,(S9) 2011 597 patients with T2D;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial
I) Dapagliflozin 2.5 mg/d + glimepiride 4 mg/d
II) Dapagliflozin 5 mg/d + glimepiride 4 mg/d
III) Dapagliflozin 10 mg/d + glimepiride 4 mg/d
IV) Placebo + glimepiride 4 mg/d
in mean HbA1c of 0.58%, 0.63% and
0.82% in group I, II, and III, respectively,
P <0.0001
Dapagliflozin as an addition to therapy with glimepiride
↓↓ HbA1c and TBW;
in hypoglycemia reported
Nauck et al,(S10) 2011 814 patients with T2D;
A double ‑blind, placebo ‑controlled,
randomized noninferiority, phase 3 trial,
lasting for 1 year
I) Metformin 1500–2500 mg/d + dapagliflozin
5–10 mg/d
II) Metformin 1500–2500 mg/d+ glipizide
10–20 mg/d
in mean HbA1c of 0.52%, for both
dapagliflozin and glipizide;
Weight loss of 3.2 kg vs weight gain of
1.2 kg for glipizide, P <0.0001
in body weight of 5% in 33.33% of the patients on
dapagliflozin;
infections of the genitalia and UTIs on dapagliflozin
of hypoglycemia on glipizide
Bailey et al,(S11) 2012 282 patients with T2D;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial
I) Dapagliflozin 1 mg/d
II) Dapagliflozin 2.5 mg/d
III) Dapagliflozin 5 mg/d
IV) Placebo
in mean HbA1c of 0.68%, 0.72%, and
0.82%, in group I, II, and III, respectively,
P <0.0001
Insulin ‑independent mode of action;
No severe adverse effects reported
Bolinder et al,(S12)
2012
182 patients with T2D;
A double ‑blind, placebo ‑controlled,
randomized trial
I) Dapagliflozin 10 mg/d + metformin
II) Placebo + metformin
TBW of 2.08 kg (95% CI, 2.84–1.31 kg;
P <0.0001)
Dapagliflozin further TBW, of FM, VAT, and SAT
Bailey et al,(S13) 2013 546 patients with T2D;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial
I) Dapagliflozin 2.5 mg/d
+ metformin 1500 mg/d
II) Dapagliflozin 5 mg/d + metformin 1500 mg/d
III) Dapagliflozin 10 mg/d
+ metformin 1500 mg/d
IV) Placebo + metformin 1500 mg/d
in mean HbA1c of 0.48%, 0.58%, and
0.78% in group I, II, and III, respectively,
P <0.0008;
in mean HbA1c of 0.02% in the placebo
group
Dapagliflozin as an addition to the therapy with
metformin ↓↓ HbA1c and TBW;
No severe adverse effects reported
Lambers Heerspink
et al,(S14) 2013
75 patients with T2D;
A double ‑blind, placebo ‑controlled,
randomized, phase 2 trial
I) Dapagliflozin 10 mg/d
II) Hydrochlorothiazide 25 mg/d
III) Placebo
body weight 2.4 kg vs 0.1 kg in
the placebo group
Dapagliflozin and hydrochlorothiazide body weight;
Comparison of the diuretic and blood pressure
lowering effects of dapagliflozin and
hydrochlorothiazide
Kaku et al,(S15) 2014 261 patients with T2D;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial
I) Dapagliflozin 5 mg/d
II) Dapagliflozin 10 mg/d
III) Placebo
in mean HbA1c of 0.41% and 0.45% in
group I and II, respectively
Dapagliflozin ↓↓ HbA1c and TBW;
Hypoglycemia only in 2 patients on dapagliflozin
10 mg/d
Ji et al,(S16) 2014 393 patients with T2D;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial
I) Dapagliflozin 5 mg/d
II) Dapagliflozin 10 mg/d
III) Placebo
in mean HbA1c of 1.04% and 1.11% in
group I and II, respectively, P <0.0001
Dapagliflozin ↓↓ HbA1c and TBW;
Infrequent hypoglycemia
Nauck et al,(S17) 2014 814 patients with T2D;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial, lasting for 2 years
I) Metformin 1500–2500 mg/d + dapagliflozin
5–10 mg/d
II) Metformin 1500–2500 mg/d + glipizide
10–20 mg/d
in body weight of 5.1 kg (95% CI,
5.7–4.4 kg) after 2 years
in body weight and glycemic durability that lasted
longer on dapagliflozin than on glipizide;
Hypoglycemia much (10 ‑fold) less frequent on
dapagliflozin than on glipizide;
Genital infections and UTIs more frequent on
dapagliflozin than on glipizide
POLISH ARCHIVES OF INTERNAL MEDICINE 2022; 132 (10)6
TABLE 1 List of studies associating overweight / obesity and the effects of sodium glucose cotransporter ‑2 inhibitors on body weight among patients with and without type 2 diabetes (continued on the next pages)
Research / year Population, type of study Treatment Main findings Remarks
Jabbour et al,(S18)
2014
432 patients with T2D;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial
I) Sitagliptin 100 mg/d ± metformin 1500 mg/d
+ dapagliflozin 10 mg/d
II) Sitagliptin 100 mg/d ± metformin 1500 mg/d
+ placebo
in body weight of 2.3 kg on
dapagliflozin vs 0.3 kg on placebo
in body weight and HbA1c on dapagliflozin vs placebo;
Genital infections more frequent on dapagliflozin, UTI
frequency almost the same on dapagliflozin and
placebo
Kovacs et al,(S19) 2014 498 patients with T2D;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial
I) Pioglitazone 30 mg/d ±
metformin 1500 mg/d + empagliflozin 10 mg/d
II) Pioglitazone 30 mg/d ±
metformin 1500 mg/d + empagliflozin 25 mg/d
III) Pioglitazone 30 mg/d ±
metformin 1500 mg/d + placebo
in mean body weight (SD) of 1.62 kg
(0.21) and 1.47 kg (0.21) on
empagliflozin 10 mg and 25 mg,
respectively, vs weight gain of 0.34 kg
(0.21) on placebo (both P <0.001)
Empagliflozin 10 mg/d or 25 mg/d resulted in in both
body weight and HbA1c;
Empagliflozin generally well tolerated
Strojek et al,(S20) 2014 597 patients with T2D;
A double ‑blind, parallel ‑group, randomized,
phase 3 trial
I) Glimepiride 4 mg/d
II) Glimepiride 4 mg/d + dapagliflozin 2.5 mg/d
III) Glimepiride 4 mg/d + dapagliflozin 5 mg/d
IV) Glimepiride 4 mg/d + dapagliflozin 10 mg/d
No significant changes in mean HbA1c
were observed;
in body weight of 1.36 kg, 1.54 kg, and
2.41 kg reported on dapagliflozin
2.5 mg/d, 5 mg/d, and 10 mg/d,
respectively, over 48 weeks
Addition of dapagliflozin led to sustained weight loss;
No severe adverse effects reported
Bolinder et al,(S21)
2014
182 patients with T2D;
A double ‑blind, placebo ‑controlled,
randomized, multi ‑arm, parallel ‑group, phase
3 trial
I) Metformin 1500 mg/d
II) Metformin 1500 mg/d +
dapagliflozin 10 mg/d
in mean HbA1c;
↓↓ in body weight of 4.54 kg and in WC
of 5 cm
↓↓ in body weight and ↓↓ in FM of 2.8 kg;
No severe adverse effects reported
DeFronzo et al,(S22)
2015
674 patients with T2D;
A double ‑blind, parallel ‑group, randomized,
phase 3 trial
I) Metformin 1500 mg/d + linagliptin 5 mg/d
II) Metformin 1500 mg/d + empagliflozin
10 mg/d
III) Metformin 1500 mg/d + empagliflozin
25 mg/d
IV) Metformin 1500 mg/d + linagliptin 5 mg/d
+ empagliflozin 10 mg/d
V) Metformin 1500 mg/d + linagliptin 5 mg/d
+ empagliflozin 25 mg/d
↓↓ in body weight and HbA1c No severe adverse effects reported attributable to
the use of empagliflozin;
Allergic reactions in 3 patients occurred when
linagliptin was used alone or in combination with
empagliflozin
Del Prato et al,(S23)
2015
814 patients with T2D;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial
I) Metformin 1500 mg/d + glipizide 10–20 mg/d
II) Metformin 1500 mg/d + dapagliflozin up to
20 mg/d
in mean HbA1c and ↓↓ in TBW of 4.38 kg
(95% CI, 5.31–3.46 kg), when comparing
dapagliflozin with glipizide at 4 years
↓↓ in mean HbA1c and TBW at 4 years on dapagliflozin
vs glipizide;
Genital infections and UTIs more frequent on
dapagliflozin but tended to disappear with time and
antibiotics
Bailey et al,(S24) 2015 274 patients with T2D;
A double ‑blind, placebo ‑controlled,
parallel ‑group, phase 3 trial
I) Placebo, ie patients who after completion of 24
weeks of the study received metformin 500 mg/d
(low dose), therefore:
II) Metformin 500 mg/d + dapagliflozin 2.5 mg/d
III) Metformin 500 mg/d + dapagliflozin 10 mg/d
IV) Metformin 500 mg/d + dapagliflozin 10 mg/d
in mean HbA1c and ↓↓ in TBW of
2.016 kg, P = 0.016
↓↓ in mean HbA1c and TBW at 2 years on dapagliflozin
vs low ‑dose metformin;
Genital infections and UTIs more frequent on
dapagliflozin;
Hypoglycemia uncommon on dapagliflozin
REVIEW ARTICLE SGLT ‑2 inhibitors in obesity and its comorbidities 7
TABLE 1 List of studies associating overweight / obesity and the effects of sodium glucose cotransporter ‑2 inhibitors on body weight among patients with and without type 2 diabetes (continued on the next page)
Research / year Population, type of study Treatment Main findings Remarks
Rosenstock et al,(S25)
2015
534 patients with T2D;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial
I) Metformin 1500 mg/d + saxagliptin 5 mg/d
+ placebo
II) Metformin 1500 mg/d + dapagliflozin
10 mg/d + placebo
III) Metformin 1500 mg/d + saxagliptin 5 mg/d
+ dapagliflozin 10 mg/d
↓↓ in mean HbA1c and ↓↓ in body weight in
patients on saxagliptin + dapagliflozin
apart from metformin
Triple combination of metformin + dapagliflozin +
saxagliptin resulted in ↓↓ in HbA1c and ↓↓ in body weight;
No episodes of severe hypoglycemia reported with
the triple combination
Fulcher et al,(S26) 2016 411 patients with T2D;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial
I) DPP ‑4 inhibitor + placebo
II) DPP ‑4 inhibitor + canagliflozin 100 mg/d
III) DPP ‑4 inhibitor + canagliflozin 300 mg/d
IV) GLP ‑1 analog + placebo
V) GLP ‑2 analog + canagliflozin 100 mg/d
VI) GLP ‑1 analog + canagliflozin 300 mg/d
in mean HbA1c and in body weight
with canagliflozin 100 mg/d or 300 mg/d
In the patients receiving incretin mimetics,
canagliflozin 100 mg/d or 300 mg/d body weight;
Incidence of hypoglycemia with canagliflozin addition
Frias et al,(S27) 2016 695 patients with T2D;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial
I) Metformin 1500 mg/d + exanetide
2 mg/weekly
II) Metformin 1500 mg/d + dapagliflozin
10 mg/d
III) Metformin 1500 mg/d + exanetide
2 mg/weekly + dapagliflozin 10 mg/d
in mean HbA1c and body weight on
the combination of exanetide +
dapagliflozin
As expected, exanetide + dapagliflozin were superior
regarding lowering mean HbA1c and weight loss, and
more patients with weight loss 5% of their body
weight;
No major adverse effects reported
Mathieu et al,(S28)
2016
294 patients with T2D;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial
I) Metformin + saxagliptin + placebo
II) Metformin + saxagliptin + dapagliflozin
10 mg/d
HbA1c, –0.74% vs 0.07%; FPG, –27 mg/dl
vs 10 mg/dl; body weight, –2.1 kg vs
–0.4 kg on dapaglifozin vs placebo
Triple combination of metformin + dapagliflozin +
saxagliptin resulted in ↓↓ in mean HbA1c and ↓↓ in body
weight;
Overall, the triple combination was well tolerated and
provided sustainable effects;
Only genital infections with Candida species were
more common in the dapagliflozin group
Neal et al,58 2017 10 142 patients with T2D and high CVD risk
receiving standard care;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial (CANVAS
program)
I) Canagliflozin 100 mg/d
II) Canagliflozin 300 mg/d
III) Placebo
Body weight loss of 1.46 kg risk of CVD event;
risk of lower limb amputation
Wanner et al,(S29)
2018
7020 patients with T2D and CVD receiving
standard care;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial (EMPAREG‑
‑OUTCOME trial)
I) Empagliflozin 10 mg/d
II) Empagliflozin 25 mg/d
III) Placebo
Body weight loss of 2 kg Empagliflozin addition to standard care led to in body
weight;
No severe adverse effects reported
Ludvik et al,(S30) 2018 424 patients with T2D;
A double ‑blind, placebo ‑controlled, parallel‑
‑arm, randomized, phase 3b trial
I) SGLT ‑2 inhibitor + placebo
II) SGLT ‑2 inhibitor + dulaglutide 0.75 mg/weekly
III) SGLT ‑2 inhibitor + dulaglutide 1.5 mg/weekly
Patients on dulaglutide instead of
placebo had ↓↓ in mean HbA1c and
↓↓ body weight
Although the addition of dulaglutide to SGLT ‑2 inhibitor
resulted in significant ↓↓ in body weight, adverse side
effects were more frequent in the groups that received
dulaglutide;
Nausea, diarrhea, vomiting were more frequent in
patients on dulaglutide instead of placebo
POLISH ARCHIVES OF INTERNAL MEDICINE 2022; 132 (10)8
TABLE 1 List of studies associating overweight / obesity and the effects of sodium glucose cotransporter ‑2 inhibitors on body weight among patients with and without type 2 diabetes (continued from the previous pages)
Research / year Population, type of study Treatment Main findings Remarks
Pratley et al,(S31) 2018 1233 patients with T2D;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial
I) Metformin 1500 mg/d + ertugliflozin 5 mg/d
II) Metformin 1500 mg/d + ertugliflozin 15 mg/d
III) Metformin 1500 mg/d + ertugliflozin 5 mg/d
+ sitagliptin 100 mg/d
IV) Metformin 1500 mg/d + ertugliflozin
15 mg/d + sitagliptin 100 mg/d
V) Metformin 1500 mg/d + sitagliptin 100 mg/d
in mean HbA1c and in body weight
with the combination of ertugliflozin
+ sitagliptin, when compared with
sitagliptin alone
in glucose levels and body weight after 52 weeks of
observation;
Only genital infections with Candida species more
common in patients on ertugliflozin
Perkovic et al,90 2019 4401 patients with T2D and albuminuric
CKD;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial (CREDENCE trial)
I) Canagliflozin 100 mg/d
II) Placebo
Canagliflozin induced weight loss of
0.8 kg
Canagliflozin ↓↓ the risk of CVD events and renal failure,
body weight;
No differences in the risk of amputations
Wiviott et al,57 2019 17 160 patients with T2D receiving standard
care;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial (DECLARE ‑TIMI
58)
I) Dapagliflozin 10 mg
II) Placebo
Weight loss of 1.8 kg Dapagliflozin as an additional treatment to standard
care was associated with ↓↓ risk of CVD events and
↓↓ risk of renal events;
DKA more common on dapagliflozin when compared
with placebo (0.3% vs 0.1%, P = 0.02);
Genital infections more common on dapagliflozin
Cannon et al,(S32) 2020 8246 patients with T2D and atherosclerotic
CVD receiving standard care;
A double ‑blind, placebo ‑controlled,
randomized, phase 3 trial (VERTIS CV)
I) Ertugliflozin 5 mg/d
II) Ertugliflozin 15 mg/d
III) Placebo
Weight loss of 2.4 kg on ertugliflozin
5 mg/d and 2.8 kg on ertugliflozin
15 mg/d
Ertugliflozin was not inferior to placebo regarding CVD
risk;
Amputations performed in 2% of patients on
ertugliflozin 5 mg/d and in 2.1% of patients on
ertugliflozin 15 mg/d, vs 1.6% in the placebo group
a References S1–S32 are listed in Supplementary material
Abbreviations: , decrease; ↓↓, strong decrease; , increase; BMI, body mass index; CKD, chronic kidney disease; CVD, cardiovascular disease; DKA, diabetic ketoacidosis; FM, fat mass; HbA1c, glycated hemoglobin; MRI,
magnetic resonance imaging; RCT, randomized controlled trials; SAT, subcutaneous adipose tissue; T2D, type 2 diabetes mellitus; TBW, total body weight; tid, thrice daily; UTIs, urinary tract infections; VAT, visceral adipose
tissue; WC, waist circumference
REVIEW ARTICLE SGLT ‑2 inhibitors in obesity and its comorbidities 9
effects on lipid metabolism and biomarkers will
most probably appear in thenear future.
Cardiovascular effects Despite the fact that
SGLT ‑2 inhibitors have been mainly introduced
for thetreatment of patients with T2D, they pos
sess pleiotropic properties over and beyond their
antidiabetic potential.
49
Among these pleiotropic
effects, their cardioprotective ability is of theut
most importance. In particular, theEMPA ‑REG
OUTCOME study55 was thefirst to show asig
nificant decrease in therate of death from CVD
causes, nonfatal infarction, or nonfatal stroke
among 7020 patients with T2D athigh risk of
anadverse CVD effect. eCANVAS study56 fol
lowed, which enrolled 10 142 patients with T2D
and high CVD risk, and confirmed alower risk
of thesame adverse CVD effects. eDECLARE‑
‑TIMI 58 study,
57
also published, like prior trials,
in New England Journal of Medicine, has document
ed asignificant reduction in therate of hospital‑
ization due to HF among 10 186 patients with
T2D and lowered risk of atherosclerotic CVD in
thepatients followed for amedian of 4.2 years.
Furthermore, theEMPEROR ‑Reduced study,
58
which enrolled 3730 patients with HF and thelat
est EMPEROR ‑Preserved study,59 which investi‑
gated 5988 patients with HF, have shown ben
eficial cardiovascular effects associated with
theuse of empagliflozin. In particular, asignif
icant decrease in death or hospitalization rate
due to HF was noted in both studies, regardless
of thepresence of T2D. eDAPA ‑HF study
60
in
cluding 4744 patients with HF and reduced EF
has also confirmed theabovementioned findings
for dapagliflozin.
ese remarkable cardioprotective effects of
SGLT ‑2 inhibitors have led to their use in patients
even without T2D but with established HF. Re
garding their beneficial effects in patients with
HF, these may be attributed to theincreased pro
duction of ketones by SGLT ‑2 inhibitors, which,
in turn, ameliorate mitochondrial dysfunction
observed in HF and increase adenosine triphos‑
phate production, resulting in animproved ven‑
tricular contractility.23 In particular, SGLT ‑2 in‑
hibitors may lead to changes in intracellular sodi
um and calcium concentrations, resulting in im‑
proved ventricular contractility and fewer cardiac
arrhythmias.
23
In addition, cardiac inflamma
tion and thesubsequent cardiac fibrosis are at
tenuated by theuse of SGLT ‑2 inhibitors. is
effect is mainly attributed to decreased produc‑
tion of free radicals in thecardiomyocytes, as
these antidiabetic drugs induce anantioxidative
and anti ‑inflammatory milieu that supports cor
onary endothelial function. It is also important
to highlight that theepicardial fat surrounding
theheart, characterized by increased production
of proinflammatory cytokines, is decreased fol‑
lowing theuse of SGLT ‑2 inhibitors, thus result‑
ing in areduction of proinflammatory cytokines
and amelioration of thesurrounding environ
ment.
61
Besides, due to theweight loss induced by
In addition, it is widely known that brown
adipose tissue (BAT) plays akey role in obesity
and obesity ‑related disorders. More specifically,
browning of thewhite adipose tissue (WAT) has
been suggested to be acrucial factor in combating
obesity.
40-43
is is mainly achieved by increasing
energy expenditure by means of increased expres
sion of uncoupling protein 1 (UCP ‑1) in BAT, as
well as anenhancement of cells expressing UCP ‑1
in WAT. ese brown ‑like adipocytes, often called
beige cells, are characterized by anincreased ex‑
pression of UCP ‑1, and thus by their transforma
tion into brown adipocytes.39 SGLT ‑2 inhibitors
have been documented to increase body energy
expenditure by increasing theexpression of UCP ‑1
in WAT and BAT, leading to browning of thead‑
ipose tissue. In this process, adiponectin and fi‑
broblast growth factor 21 have been found to be
elevated as aresult of chronic administration of
SGLT ‑2 inhibitors.44-48
Overall, SGLT ‑2 inhibitors seem to be associ‑
ated with weight loss via their glucosuric effects,
polarization toward theM2 phenotype of macro
phages, and browning of theWAT.
Effects on glycemic control
SGLT ‑2 inhibitors in
duce glucosuria, thereby ameliorating theserum
glucose levels, while their ability to promote fat
utilization and browning of theWAT improves
insulin sensitivity.24 SGLT ‑2 inhibitors used as
monotherapy have been shown to lower fasting
plasma glucose (FPG) by 20–46mg/dl and gly
cated hemoglobin (HbA1c) by 0.54% to 1.45% in
patients with baseline HbA1c of 7% to 9.1%, as
compared with placebo. Notably, theaddition of
SGLT ‑2 inhibitors as anadd ‑on therapy to met‑
formin may lower FPG by 15–40mg/dl and HbA
1c
by 0.54%–0.77% as compared with placebo in
patients with baseline HbA1c between 7.9% and
8.2%.49
Although SGLT ‑2 inhibitors are widely used
for thetreatment of patients with T2D, individ
uals with T1D may also benefit from their pleio
tropic properties, apart from theglucose low
ering effects.
49-51
In fact, sotagliflozin, adual
SGLT ‑1 and SGLT ‑2 inhibitor, is authorized in
theEuropean Union only for thetreatment of
patients with both T1D and obesity.
52
Howev
er, theuse of SGLT ‑2 inhibitors or even sota
gliflozin among patients with T1D has been as
sociated with higher rates of euglycemic diabetic
ketoacidosis (EDKA), as these patients are more
prone to ketoacidosis than patients with T2D.
Besides, there is always ahigher risk of hypogly
cemia among patients with T1D due to thecon
current use of insulin.53
Regarding theeffects of SGLT ‑2 inhibitors on
lipid parameters, it has been suggested that this
class of antidiabetic drugs may lead to asmall
increase in serum high‑density lipoprotein cho‑
lesterol levels, whereas other reports have also
shown aslight increase in low‑density lipopro
tein cholesterol levels.
10,54
As these drugs are now
extensively used, more research in terms of their
POLISH ARCHIVES OF INTERNAL MEDICINE 2022; 132 (10)10
theuse of canagliflozin with kidney function as
aprimary end point. is trial showed that the
relative risk of death from renal causes among
theenrolled 4401 patients was reduced by 34%,
while therelative risk of ESRD was reduced by
32% during themedian follow ‑up of 2.62 years.
68
eDAPA ‑CKD study,
69
which dealt with thead
ministration of another SGLT ‑2 inhibitor, dapa‑
gliflozin, in 4304 patients with kidney dysfunc‑
tion and with or without T2D, has also demon
strated aslower decline in eGFR in thelong term,
especially among patients with higher HbA
1c
and
higher urinary to creatinine ratio atthe begin
ning of thestudy. econsistent results demon‑
strating areduction of adverse renal outcomes
across abroad selection of different SGLT ‑2 in
hibitors point toward aclass renoprotective ef
fect of SGLT ‑2 inhibitors. erefore, theusage
of SGLT ‑2 inhibitors has been expanded even in
lower eGFR, where their antidiabetic potential is
compromised but their nephroprotective as well
as cardioprotective properties are still sustained.
In particular, atGFRbelow 45ml/min/1.73m2,
SGLT ‑2 inhibitors have lower glycemic efficacy
and usually another agent should be added to
achieve glycemic goals. However, depending on
thelabeling of representative drugs, they should
be initiated even when eGFR is between 30 and
45ml/min/1.73m2 due to their dual beneficial car
diorenal effects. eresults of DAPA ‑CKD study
7
generalized these findings outside of theframe
of diabetic nephropathy, and led to approvals by
theFDA and theEuropean Medicines Agency, re
garding theuse of dapagliflozin among patients
with CKD of nondiabetic etiology.
Regarding CKD and theobesity paradox, it
should be noted that thephenomenon of “re
verse epidemiology” is observed in CKD patients,
whereas increased BMI has been associated with
better survival outcomes.
70
However, this phe
nomenon, which apart from CKD has been ob
served in heart disease, liver cirrhosis, and chron
ic obstructive lung disease, may be attributed to
protein wasting and cachexia, which characterize
theadvanced stages of theabovementioned dis‑
orders.70 SGLT ‑2 inhibitors act mainly by caus
ing glucosuria and natriuresis, thereby inducing
weight loss without leading to sarcopenia and
cachexia.71 In addition, it should be pointed out
that SGLT ‑2 inhibitors, by interfering with thein
flammasome pathway, may result in reduced re‑
lease of IL ‑1β, which is widely known for its key
role in thepathogenesis of CKD.72
Effects on hepatic function and nonalcoholic fatty
liver disease Nowadays, NAFLD has been as
sessed to affect almost 70% to 80% of patients
with T2D,
73,74
while in obesity its prevalence rang
es from 50% to 90%, depending on thedegree of
excess adiposity.
76
NAFLD, which is character
ized by excess fat accumulation in thehepato
cytes, may be associated with nonalcoholic ste
atohepatitis (NASH), cirrhosis, and even hepato
cellular carcinoma.
76,77
Apart from these severe
SGLT ‑2 inhibitors, there is also ageneral decrease
in theproduction of proinflammatory cytokines
by theadipose tissue, which accounts for abetter
and more functional cardiac microenvironment.
62
Effects on arterial blood pressure Due to their na
triuretic effects, SGLT ‑2 inhibitors slightly reduce
arterial blood pressure (3–7mmHg for systolic
and 2mmHg for diastolic blood pressure).
63
is
anti hypertensive potential is reported to be pres
ent regardless of theuse of other antihypertensive
drugs, such as loop diuretics. However, it has been
demonstrated that SGLT ‑2 inhibitors and loop di
uretics may share asynergistic natriuretic effect.
64
Notably, theantihypertensive potential of SGLT ‑2
inhibitors does not induce further release of re‑
nin by themacula densa. edecreased intravas
cular volume resulting from thenatriuretic prop
erties of SGLT ‑2 inhibitors does not seem to ac‑
tivate therenin ‑angiotensin ‑aldosterone system
or thesympathetic activity tone. is unique fea
ture of SGLT ‑2 inhibitors may be thecornerstone
of thebeneficial effects of SGLT ‑2 inhibitors re‑
garding blood pressure.
64
Apart from their natri
uretic effect, SGLT ‑2 inhibitors decrease thearte
rial stiffness, thus reducing thearterial tone and
decreasing thearterial blood pressure. is de
crease in arterial stiffness may be attributed to
reduction in theperivascular fat caused by SGLT ‑2
inhibitors, as reported by Batzias et al65 in their
meta ‑analysis. Furthermore, theweight loss ef‑
fect may also account for thereduction in arterial
blood pressure.65 Overall, thenatriuretic effects
together with theweight loss and thereduction
in arterial stiffness all result in decreased arteri‑
al blood pressure.
Effects on renal function Among patients with
T2D, theincreased reabsorption of sodium and
glucose by theSGLT ‑2 cotransporters accounts
for thestate of hyperfiltration noted in thevery
early stages of diabetic kidney disease.
10
is phe
nomenon is mainly attributed to anenhanced re
absorption of sodium leading to vasoconstriction
of theafferent arteriole. euse of SGLT ‑2 inhib
itors may result in reduced hyperfiltration as well
as lowered intraglomerular pressure, and there‑
fore ameliorated renal function. Interestingly, in
theEMPA ‑REG Outcome study,
55
theadministra
tion of empagliflozin was associated with areduc
tion in therate of doubling of serum creatinine
levels, areduction in albuminuria, progression to
end‑stage renal disease (ESRD), and death related
to kidney dysfunction. eEMPA ‑REG Outcome
study
55
was thefirst to report these favorable re
nal effects associated with theuse of aSGLT ‑2 in
hibitor. eCANVAS trial ensued,66 which con
firmed that theuse of another SGLT ‑2 inhibitor,
canagliflozin, resulted in reduction in theprogres
sion of albuminuria and a40% decline in dete
rioration of estimated glomerular filtration rate
(eGFR), theneed for renal replacement treat
ment, and death of renal origin causes. eCRE
DENCE study
67
was thefirst one investigating
REVIEW ARTICLE SGLT ‑2 inhibitors in obesity and its comorbidities 11
are among themost beneficial features regard
ing their use in women with PCOS.81 Notably,
1 current RCT studying theadministration of
25mg of empagliflozin or 1500mg of metfor
min in 39 women with PCOS, has showed are
duction in body weight, BMI, waist circumfer
ence, and total fat, but no significant differences
in insulin resistance and androgen levels.
82
An
other RCT is ongoing regarding theadministra‑
tion of canagliflozin vs metformin among wom‑
en with PCOS [NCT04700839]. Overall, due to
theweight loss and blood pressure lowering ef‑
fects of SGLT ‑2 inhibitors, there is anongoing
interest in theadministration of SGLT ‑2 inhibi‑
tors in women with PCOS to also improve vari‑
ous CVD risk parameters.
Adverse events and tolerability emost com
mon adverse effect of SGLT ‑2 inhibitors is geni‑
tal candidiasis. ephenomenon of glucosuria,
which is themain mechanism of action in this
category of drugs, confers afavorable environ
ment for thegrowth of Candida species. Apart
from mycotic genitalia infections, which are re‑
ported to be 4 to 6 times increased, bacterial
infections of theurinary system may also de
velop, although less frequently. ese urinary
tract infections are rarely severe enough to cause
pyelonephritis.
83,84
Another adverse effect of this
class of drugs is that they may provoke EDKA,
which, however, is highly preventable and may
be managed by discontinuation of thedrug and
intravenous administration of fluids together
with insulin and potassium supplementation, if
needed. Nevertheless, if apatient is adequate
ly hydrated, this adverse effect occurs very rare‑
ly.
23,85
Atransient increase in serum creatinine
levels, albeit of questionable clinical relevance,
is observed frequently following theinitiation
of SGLT‑2 inhibitor therapy.86 is is in contrast
with documented long ‑term nephroprotective ef
fects of SGLT ‑2 inhibitors (see section Effects on
renal function), while therapy with dapagliflozin
appears to be safe even in patients with stage
4 CKD.
87
Regarding canagliflozin, theCANVAS
study
56
reported that canagliflozin therapy was
associated with anincreased risk of limb ampu‑
tation and bone fractures. However, these results
were not confirmed in theCREDENCE study,
88
while they have not been reported with theuse
of other SGLT ‑2 inhibitors. Furthermore, ather
apy with SGLT ‑2 inhibitors does not seem to in‑
crease fracture risk in patients with CKD, regard
less of baseline eGFR.89
Perspectives and challenges According to
theAmerican Diabetes Association and theAmeri
can Heart Association recommendations for 2022,
in patients with T2D and established CVD or re‑
nal disease, SGLT ‑2 inhibitors or GLP ‑1 analogs
or both are recommended.
23,90,91
Regarding obesi
ty treatment, despite thefact that SGLT ‑2 inhibi
tors induce weight loss, their weight ‑lowering ef
fects are counterbalanced by anincreased appetite
liver consequences, NAFLD, especially in patients
with obesity and T2D, calls for action in terms of
CVD, as it is related to increased cardiovascular
adverse effects.78,79 In their recent meta ‑analysis,
Mantovani et al
80
have concluded that SGLT ‑2 in
hibitors have beneficial effects regarding NAFLD.
In particular, they have documented asignificant
decrease in liver fat content as estimated by mag
netic resonance techniques as well as reductions
in liver enzymes, such as alanine aminotrans
ferase (ALT) and gamma ‑glutamyl transferase
(γ ‑GT). eir meta ‑analysis included random
ized controlled trials (RCTs) involving theuse of
empagliflozin, dapagliflozin, canagliflozin, and
ipragliflozin in patients with NAFLD defined by
magnetic resonance techniques and not by liver
biopsy, as there were no eligible RCTs regarding
SGLT ‑2 inhibitors and NAFLD assessed by liv
er biopsy. e participants in theincluded stud‑
ies were overweight or obese and 90% had T2D.
When compared with theplacebo group, theuse
of SGLT ‑2 inhibitors for 24 weeks resulted in ame
lioration of serum ALT and γ‑GT levels as well as
improvement in liver fat content (pooled weight
ed mean differences, −2.05%; 95% CI, −2.61% to
−1.48%), and reduction in body weight of approx
imately 3.5kg.
80
Overall, evidence points toward
abeneficial class effect of SGLT ‑2 inhibitors in liv
er steatosis among patients with T2D. More stud
ies in thefield are needed to generalize thecur‑
rent findings in patients without overt dysgly
cemia. Additionally, in order to assess potential
synergistic effects of SGLT ‑2 inhibition and oth‑
er treatment approaches, atleast 1 ongoing study
aims to compare theeffects of combined SGLT ‑2
inhibitor (empagliflozin 10mg) and GLP‑1 ago‑
nist (1mg semaglutide weekly) vs empagliflozin
monotherapy or placebo in NASH among T2D
patients, with invasive (liver biopsy) and non‑
invasive (elastography) measures to asses liver
steatosis, fibrosis, and inflammation (clinical
trials.gov registration number: NCT04639414)
Effects on ovarian function Polycystic ovary syn‑
drome (PCOS) affects approximately 20% of wom
en of reproductive age.
28
It is characterized by ab
normalities in ovulation, hyperandrogenemia,
and / or pathologic ovarian ultrasound morpholo
gy, and may be associated with insulin resistance
and subsequent hyperinsulinemia. elatter fea
ture is responsible for themetabolic derangement
related to PCOS and theincreased CVD risk. In
this case, insulin sensitizers, such as metformin
or thiazolidinediones may be alternatives to oral
contraceptives.81 Approximately 60% to 70% of
women with PCOS finally develop insulin resis‑
tance and hyperinsulinemia. euse of other an
tidiabetic agents, especially with weight‑lower
ing effects is also under investigation regarding
thetreatment options for women with PCOS. De
spite thelack of SGLT ‑2 cotransporters in theova
ries, theeffects of SGLT ‑2 inhibitors, which in
duce glucosuria leading to weight loss and natri‑
uresis resulting in reduction of blood pressure,
POLISH ARCHIVES OF INTERNAL MEDICINE 2022; 132 (10)12
associated with theuse of these agents continue
to be elucidated, broadening of their clinical in‑
dications outside of thestrict frame of diabetes
will not come as asurprise.
SUPPLEMENTARY MATERIAL
Supplementary material is available at www.mp.pl/paim.
ARTICLE INFORMATION
ACKNOWLEDGMENTS None.
FUNDING This work did not receive any funding.
CONFLICT OF INTEREST None declared.
OPEN ACCESS This is an Open Access article distributed under the terms
of the Creative Commons Attribution‑NonCommercial‑ShareAlike 4.0 Inter‑
national License (CC BY ‑NC ‑SA 4.0), allowing third parties to copy and re‑
distribute the material in any medium or format and to remix, transform, and
build upon the material, provided the original work is properly cited, distrib‑
uted under the same license, and used for noncommercial purposes only. For
commercial use, please contact the journal office at pamw@mp.pl.
HOW TO CITE Vallianou NG, Tsilingiris D, Kounatidis D, et al. Sodium‑glu‑
cose cotransporter‑2 inhibitors in obesity and its associated cardiometa
bolic disorders: where do we stand? Pol Arch Intern Med. 2022; 132:
16342. doi:10.20452/pamw.16342
REFERENCES
1 Dalamaga M, Karampela I, Mantzoros CS. Commentary: phosphodiester
ase 4 inhibitors as potential adjunct treatment targeting the cytokine storm
in COVID ‑19. Metabolism. 2020; 109: 154282.
2 Hroussalas G, Kassi E, Dalamaga M, et al. Leptin, soluble leptin receptor,
adiponectin and resistin in relation to OGTT in overweight / obese postmeno
pausal women. Maturitas. 2008; 59: 339‑349.
3 Karampela I, Chrysanthopoulou E, Christodoulatos GS, Dalamaga M. Is
there an obesity paradox in critical illness? Epidemiologic and metabolic con
siderations. Curr Obes Rep. 2020; 9: 231‑244.
4 Marouga A, Dalamaga M, Kastania AN, et al. Correlates of serum resis‑
tin in elderly, non ‑diabetic patients with chronic kidney disease. Clin Lab.
2013; 59: 1121‑1128.
5 Marouga A, Dalamaga M, Kastania AN, et al. Circulating resistin is a sig
nificant predictor of mortality independently from cardiovascular comorbid‑
ities in elderly, non ‑diabetic subjects with chronic kidney disease. Biomark‑
ers. 2016; 21: 73‑79.
6 World Health Organization. Obesity. https://www.who.int/health ‑topics/
obesity#tab=tab_1. Accessed August 8, 2022.
7 World Health Organization. Global Report on Diabetes. April 21, 2016.
https://www.who.int/publications/i/item/9789241565257. Accessed Au
gust 8, 2022.
8 Koziel P, Jankowski P, Mirek ‑Bryniarska E, et al. Obesity in patients with
established coronary artery disease over a 20 ‑year period (1997‑2017). Pol
Arch Intern Med. 2021; 131: 26‑32.
9 Rajca A, Wojciechowska A, Smigielski W, et al. Increase in the prev‑
alence of metabolic syndrome in Poland: comparison of the results of
the WOBASZ (2003‑2005) and WOBASZ II (2013‑2014) studies. Pol Arch
Intern Med. 2021; 131: 520‑526.
10 Pereira MJ, Eriksson JW. Emerging role of SGLT ‑2 Inhibitors for
the treatment of obesity. Drugs. 2019; 79: 219‑230.
11 Fogarasi A, Gonzalez K, Dalamaga M, Magkos F. The impact of the rate
of weight loss on body composition and metabolism. Curr Obes Rep. 2022;
11: 33‑44.
12 Argyrakopoulou G, Konstantinidou SK, Dalamaga M, Kokkinos A. Nu‑
tritional deficiencies before and after bariatric surgery: prevention and treat‑
ment. Curr Nutr Rep. 2022; 11: 95‑101.
13 Liu J, Tsilingiris D, Dalamaga M. The non ‑linear relationship between
muscle mass and BMI calls into question the use of BMI as a major criteri‑
on for eligibility for bariatric surgery. Metabol Open. 2022; 13: 100164.
14 Yau K, Dharia A, Alrowiyti I, Cherney DZI. Prescribing SGLT2 inhibitors
in patients with CKD: expanding indications and practical considerations.
Kidney Int Rep. 2022; 7: 1463‑1476.
15 Zheng H, Liu M, Li S, et al. Sodium ‑glucose co ‑transporter ‑2 inhibitors
in non ‑diabetic adults with overweight or obesity: a systematic review and
meta ‑analysis. Front Endocrinol (Lausanne). 2021; 12: 706914.
16 Vick H, Diedrich DF, Baumann K. Reevaluation of renal tubular glu
cose transport inhibition by phlorizin analogs. Am J Physiol. 1973; 224:
552‑557.
17 Rossetti L, Smith D, Shulman GI, et al. Correction of hyperglycemia
with phlorizin normalizes tissue sensitivity to insulin in diabetic rats. J Clin
Invest. 1987; 79: 1510‑1515.
18 Valdes ‑Socin H, Scheen AJ, Jouret F, et al. From the discovery of phlo‑
rizin (a Belgian story) to SGLT2 inhibitors [in French]. Rev Med Liege. 2022;
77: 175‑180.
and calorie intake. erefore, they are not prob‑
able to be used alone in obesity treatment. How
ever, their combination with drugs suppressing
appetite and / or inducing satiety would be much
more feasible and welcome. Indeed, acombina‑
tion of aSGLT ‑2 inhibitor with aGLP ‑1 analog,
especially aonce ‑weekly administered GLP ‑1 an
alog, would be more convenient and more potent
in this regard. In addition, it would be interest‑
ing to combine aSGLT ‑2 inhibitor with adual
GLP ‑1 analog and glucose ‑dependent insulino
tropic polypeptide, which may be even more ef‑
ficient in suppressing appetite and in promoting
reduced gastric emptying. Regarding thealready
used weight ‑lowering drugs, such as lorcaserine,
GLP ‑1 analogs, phentermine and topiramate, and
bupropione and naltrexone combinations, they
are known to act centrally atthe central nervous
system level to suppress appetite.
10,23
erefore,
a combination with a SGLT‑2 inhibitor that may
increase appetite would be really intriguing in
terms of the weight‑lowering efficacy.
Combinations of SGLT ‑2 inhibitors with
other drugs for thetreatment of T2D, such as
dipeptidyl ‑peptidase inhibitors and insulin se
cretagogues, have been studied much better than
theweight ‑lowering combinations.92 In particu‑
lar, there is growing evidence in favor of thecom
bination of SGLT ‑2 inhibitors with GLP ‑1 analogs
in T2D, if thecost is not abarrier.93 In addition,
thecombination of SGLT ‑2 inhibitors with insulin
has been suggested to lower HbA
1c
levels and body
weight gain caused by insulin treatment. How
ever, anadjustment of theinsulin dose to avoid
hypoglycemia and theadvent of EDKA should
be borne in mind by theattending physicians.94
Conclusions SGLT ‑2 inhibitors, theclass of anti
diabetic agents named “the new kids on theblock”
in 2015 by Cefalu and Riddle,20 seem to possess
pleiotropic properties ranging from glycosuria
to weight loss, cardioprotection and renoprotec‑
tion.10,20,23,95 Apart from their glucose ‑lowering
effects, SGLT ‑2 inhibitors exhibit renoprotective
properties, as they significantly improve theintra
glomerular pressure, thereby ameliorating eGFR,
while also reducing albuminuria.10,2 3 In addition,
they confer cardioprotection, especially in terms
of improving HF, as documented by thedecrease
in thenumber of hospitalizations due to HF.
23
Furthermore, they seem to ameliorate NAFLD
indices and promote weight loss, particularly in
conjunction with theuse of GLP ‑1 analogs.
96,97
eir utilization in obesity and obesity ‑related
disorders seems beneficial, as they are also safe
with rare severe adverse effects.98 While therole
of SGLT ‑2 inhibitors in thetreatment of T2D and
lately cardiac failure and chronic renal disease of
non diabetic etiology is already firmly established,
their utility in thetherapy of obesity, NAFLD,
and other indications, alone or in combination
with other agents will continue to be researched
in theforthcoming years. Since themultifac
eted mode of action and thepotential benefits
REVIEW ARTICLE SGLT ‑2 inhibitors in obesity and its comorbidities 13
45 Inagaki T, Dutchak P, Zhao G, et al. Endocrine regulation of the fasting
response by PPARalpha ‑mediated induction of fibroblast growth factor 21.
Cell Metab. 2007; 5: 415‑425.
46 Liu J, Dalamaga M. Emerging roles for stress kinase p38 and stress
hormone fibroblast growth factor 21 in NAFLD development. Metabol Open.
2021; 12: 100153.
47 Reitman ML. FGF21: a missing link in the biology of fasting. Cell Metab.
2007; 5: 405‑407.
48 Wu P, Wen W, Li J, et al. Systematic review and meta ‑analysis of ran‑
domized controlled trials on the effect of SGLT2 inhibitor on blood leptin and
adiponectin level in patients with type 2 diabetes. Horm Metab Res. 2019;
51: 487‑494.
49 Vallianou NG, Geladari E, Kazazis CE. SGLT ‑2 inhibitors: their pleiotropic
properties. Diabetes Metab Syndr. 2017; 11: 311‑315.
50 Vallianou NG, Stratigou T, Geladari E, et al. Diabetes type 1: can it be
treated as an autoimmune disorder? Rev Endocr Metab Disord. 2021; 22:
859‑876.
51 Karamanakos G, Kokkinos A, Dalamaga M, Liatis S. Highlighting
the role of obesity and insulin resistance in type 1 diabetes and its associ‑
ated cardiometabolic complications. Curr Obes Rep. 2022; 11: 180‑202.
52 Brosius FC, Vandvik PO. Cardioprotection with yet another SGLT2 in‑
hibitor ‑ an embarrassment of riches. N Engl J Med. 2021; 384: 179‑181.
53 Garg SK, Henry RR, Banks P, et al. Effects of sotagliflozin added to insu
lin in patients with type 1 diabetes. N Engl J Med. 2017; 377: 2337‑2348.
54 Bays HE, Weinstein R, Law G, Canovatchel W. Canagliflozin: effects
in overweight and obese subjects without diabetes mellitus. Obesity (Silver
Spring). 2014; 22: 1042‑1049.
55 Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, cardiovascu
lar outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015; 373:
2117‑2128.
56 Neal B, Perkovic V, Mahaffey KW, et al. Canagliflozin and cardio
vascular and renal events in type 2 diabetes. N Engl J Med. 2017; 377:
644‑657.
57 Wiviott SD, Raz I, Bonaca MP, et al. Dapagliflozin and cardiovascular
outcomes in type 2 diabetes. N Engl J Med. 2019; 380: 347‑357.
58 Anker SD, Butler J, Filippatos G, et al. Empagliflozin in heart failure with
a preserved ejection fraction. N Engl J Med. 2021; 385: 1451‑1461.
59 Packer M, Anker SD, Butler J, et al. Cardiovascular and renal outcomes
with empagliflozin in heart failure. N Engl J Med. 2020; 383: 1413‑1424.
60 McMurray JJV, Solomon SD, Inzucchi SE, et al. Dapagliflozin in pa
tients with heart failure and reduced ejection fraction. N Engl J Med. 2019;
381: 1995‑2008.
61 Masson W, Lavalle ‑Cobo A, Nogueira JP. Effect of SGLT2 ‑inhibitors on
epicardial adipose tissue: a meta ‑analysis. Cells. 2021; 10: 2150.
62 Neeland IJ, McGuire DK, Chilton R, et al. Empagliflozin reduces body
weight and indices of adipose distribution in patients with type 2 diabetes
mellitus. Diab Vasc Dis Res. 2016; 13: 119‑126.
63 Oliva RV, Bakris GL. Blood pressure effects of sodium ‑glucose co‑
‑transport 2 (SGLT2) inhibitors. J Am Soc Hypertens. 2014; 8: 330‑339.
64 Verma A, Patel AB, Waikar SS. SGLT2 inhibitor: not a traditional diuret‑
ic for heart failure. Cell Metab. 2020; 32: 13‑14.
65 Batzias K, Antonopoulos AS, Oikonomou E, et al. Effects of newer anti
diabetic drugs on endothelial function and arterial stiffness: a systematic re
view and meta ‑analysis. J Diabetes Res. 2018; 2018: 1232583.
66 Jardine MJ, Zhou Z, Mahaffey KW, et al. Renal, cardiovascular, and
safety outcomes of canagliflozin by baseline kidney function: a secondary
analysis of the CREDENCE randomized trial. J Am Soc Nephrol. 2020; 31:
1128‑1139.
67 Jardine MJ, Mahaffey KW, Neal B, et al. The canagliflozin and renal
endpoints in diabetes with established nephropathy clinical evaluation (CRE
DENCE) study rationale, design, and baseline characteristics. Am J Nephrol.
2017; 46: 462‑472.
68 Schernthaner G, Groop PH, Kalra PA, et al. Sodium ‑glucose linked
transporter ‑2 inhibitor renal outcome modification in type 2 diabetes: evi
dence from studies in patients with high or low renal risk. Diabetes Obes
Metab. 2020; 22: 1024‑1034.
69 Heerspink HJL, Jongs N, Chertow GM, et al. Effect of dapagliflozin on
the rate of decline in kidney function in patients with chronic kidney disease
with and without type 2 diabetes: a prespecified analysis from the DAPA‑
‑CKD trial. Lancet Diabetes Endocrinol. 2021; 9: 743‑754.
70 Kalantar ‑Zadeh K, Block G, Humphreys MH, Kopple JD. Reverse epide‑
miology of cardiovascular risk factors in maintenance dialysis patients. Kid‑
ney Int. 2003; 63: 793‑808.
71 Schork A, Saynisch J, Vosseler A, et al. Effect of SGLT2 inhibitors on
body composition, fluid status and renin ‑angiotensin ‑aldosterone system in
type 2 diabetes: a prospective study using bioimpedance spectroscopy. Car
diovasc Diabetol. 2019; 18: 46.
72 Lei Y, Devarapu SK, Motrapu M, et al. Interleukin ‑1beta inhibition for
chronic kidney disease in obese mice with type 2 diabetes. Front Immu
nol. 2019; 10: 1223.
73 Vallianou N, Christodoulatos GS, Karampela I, et al. Understanding
the role of the gut microbiome and microbial metabolites in non ‑alcoholic
19 Powell DR, Zambrowicz B, Morrow L, et al. Sotagliflozin decreases
postprandial glucose and insulin concentrations by delaying intestinal glu‑
cose absorption. J Clin Endocrinol Metab. 2020; 105: e1235 ‑e1249.
20 Cefalu WT, Riddle MC. SGLT2 inhibitors: the latest “new kids on
the block”! Diabetes Care. 2015; 38: 352‑354.
21 Dalamaga M, Christodoulatos GS, Karampela I, et al. Understanding
the co ‑epidemic of obesity and COVID ‑19: current evidence, comparison
with previous epidemics, mechanisms, and preventive and therapeutic per‑
spectives. Curr Obes Rep. 2021; 10: 214‑243.
22 Tentolouris A, Vlachakis P, Tzeravini E, et al. SGLT2 inhibitors: a review
of their antidiabetic and cardioprotective effects. Int J Environ Res Public
Health. 2019; 16: 2965.
23 Braunwald E. Gliflozins in the management of cardiovascular disease.
N Engl J Med. 2022; 386: 2024‑2034.
24 Xu L, Ota T. Emerging roles of SGLT2 inhibitors in obesity and insulin
resistance: focus on fat browning and macrophage polarization. Adipocyte.
2018; 7: 121‑128.
25 Ferrannini G, Hach T, Crowe S, et al. Energy balance after sodium‑
‑glucose cotransporter 2 inhibition. Diabetes Care. 2015; 38: 1730‑1735.
26 Polidori D, Iijima H, Goda M, et al. Intra‑ and inter ‑subject variability
for increases in serum ketone bodies in patients with type 2 diabetes treat‑
ed with the sodium glucose co ‑transporter 2 inhibitor canagliflozin. Diabetes
Obes Metab. 2018; 20: 1321‑1326.
27 Rajeev SP, Sprung VS, Roberts C, et al. Compensatory changes in ener
gy balance during dapagliflozin treatment in type 2 diabetes mellitus: a ran‑
domised double ‑blind, placebo ‑controlled, cross over trial (ENERGIZE)‑study
protocol. BMJ Open. 2017; 7: e013539.
28 Dalamaga M, Papadavid E, Basios G, et al. Ovarian SAHA syndrome
is associated with a more insulin ‑resistant profile and represents an inde‑
pendent risk factor for glucose abnormalities in women with polycystic ova
ry syndrome: a prospective controlled study. J Am Acad Dermatol. 2013;
69: 922‑930.
29 Kassi E, Dalamaga M, Hroussalas G, et al. Adipocyte factors, high‑
‑sensitive C ‑reactive protein levels and lipoxidative stress products in over‑
weight postmenopausal women with normal and impaired OGTT. Maturi
tas. 2010; 67: 72‑77.
30 Koliaki C, Liatis S, Dalamaga M, Kokkinos A. The implication of gut hor
mones in the regulation of energy homeostasis and their role in the patho‑
physiology of obesity. Curr Obes Rep. 2020; 9: 255‑271.
31 Sotiropoulos GP, Dalamaga M, Antonakos G, et al. Chemerin as a bio‑
marker at the intersection of inflammation, chemotaxis, coagulation, fibrino‑
lysis and metabolism in resectable non ‑small cell lung cancer. Lung Cancer.
2018; 125: 291‑299.
32 Karampela I, Christodoulatos GS, Kandri E, et al. Circulating eNampt
and resistin as a proinflammatory duet predicting independently mortality
in critically ill patients with sepsis: a prospective observational study. Cyto‑
kine. 2019; 119: 62‑70.
33 Dalamaga M, Liu J. Targeting gasdermin D and neutrophil mobilization
for cardioprotection. Metabol Open. 2021; 12: 100152.
34 Dalamaga M, Liu J. A chromatin remodeling checkpoint of diet‑
‑induced macrophage activation in adipose tissue. Meatbol Open. 2022; 15:
100204.
35 LaMarche NM, Lynch L. Adipose dendritic cells come out of hiding. Cell
Metab. 2018; 27: 485‑486.
36 Macdougall CE, Wood EG, Loschko J, et al. Visceral adipose tissue im
mune homeostasis is regulated by the crosstalk between adipocytes and
dendritic cell subsets. Cell Metab. 2018; 27: 588‑601.e4.
37 Stratigou T, Muscogiuri G, Kotopouli M, et al. Lower circulating
omentin ‑1 is independently linked to subclinical hypothyroidism reflecting
cardiometabolic risk: an observational case ‑control and interventional, lon‑
gitudinal study. Panminerva Med. 2022; Jun 17. [Epub ahead of print]
38 Tsilingiris D, Dalamaga M, Liu J. SARS ‑CoV ‑2 adipose tissue infec
tion and hyperglycemia: a further step towards the understanding of severe
COVID ‑19. Metabol Open. 2022; 13: 100163.
39 Tsigalou C, Vallianou N, Dalamaga M. Autoantibody production in obe‑
sity: is there evidence for a link between obesity and autoimmunity? Curr
Obes Rep. 2020; 9: 245‑254.
40 Arany Z. Taking a BAT to the chains of diabetes. N Engl J Med. 2019;
381: 2270‑2272.
41 Kim SH, Plutzky J. Brown fat and browning for the treatment of obesi‑
ty and related metabolic disorders. Diabetes Metab J. 2016; 40: 12‑21.
42 Cheng L, Wang J, Dai H, et al. Brown and beige adipose tissue: a nov‑
el therapeutic strategy for obesity and type 2 diabetes mellitus. Adipocyte.
2021; 10: 48‑65.
43 van Marken Lichtenbelt WD, Vanhommerig JW, Smulders NM, et al.
Cold ‑activated brown adipose tissue in healthy men. N Engl J Med. 2009;
360: 1500‑1508.
44 Hui X, Gu P, Zhang J, et al. Adiponectin enhances cold ‑induced brown‑
ing of subcutaneous adipose tissue via promoting M2 macrophage prolifer‑
ation. Cell Metab. 2015; 22: 279‑290.
POLISH ARCHIVES OF INTERNAL MEDICINE 2022; 132 (10)14
fatty liver disease: current evidence and perspectives. Biomolecules. 2021;
12: 56.
74 Younossi ZM. Non ‑alcoholic fatty liver disease – a global public health
perspective. J Hepatol. 2019; 70: 531‑544.
75 Divella R, Mazzocca A, Daniele A, et al. Obesity, nonalcoholic fatty liv‑
er disease and adipocytokines network in promotion of cancer. Int J Biol Sci.
2019; 15: 610‑616.
76 Diehl AM, Day C. Cause, pathogenesis, and treatment of nonalcoholic
steatohepatitis. N Engl J Med. 2017; 377: 2063‑2072.
77 Fotis D, Liu J, Dalamaga M. Could gut mycobiome play a role in NAFLD
pathogenesis? Insights and therapeutic perspectives. Metabol Open. 2022;
14: 100178.
78 Al KF, Bisanz JE, Gloor GB, et al. Evaluation of sampling and stor
age procedures on preserving the community structure of stool microbio
ta: a simple at ‑home toilet ‑paper collection method. J Microbiol Methods.
2018; 144: 117‑121.
79 Dalamaga M, Liu J. DRAK2 ‑SRSF6 ‑regulated RNA alternative splicing
is a promising therapeutic target in NAFLD / NASH. Metabol Open. 2022;
13: 100157.
80 Mantovani A, Petracca G, Csermely A, et al. Sodium ‑glucose
cotransporter ‑2 inhibitors for treatment of nonalcoholic fatty liver disease:
a meta ‑analysis of randomized controlled trials. Metabolites. 2020; 11:
22.
81 Marinkovic ‑Radosevic J, Cigrovski Berkovic M, Kruezi E, et al. Explor‑
ing new treatment options for polycystic ovary syndrome: review of a novel
antidiabetic agent SGLT2 inhibitor. World J Diabetes. 2021; 12: 932‑938.
82 Javed Z, Papageorgiou M, Deshmukh H, et al. Effects of empagliflozin
on metabolic parameters in polycystic ovary syndrome: a randomized con‑
trolled study. Clin Endocrinol (Oxf). 2019; 90: 805‑813.
83 Palmer BF, Clegg DJ, Taylor SI, Weir MR. Diabetic ketoacidosis, sodi‑
um glucose transporter ‑2 inhibitors and the kidney. J Diabetes Complica
tions. 2016; 30: 1162‑1166.
84 Taylor SI, Blau JE, Rother KI. SGLT2 inhibitors may predispose to keto‑
acidosis. J Clin Endocrinol Metab. 2015; 100: 2849‑2852.
85 Halimi S, Verges B. Adverse effects and safety of SGLT ‑2 inhibitors. Di‑
abetes Metab. 2014; 40: S28 ‑S34.
86 Meraz ‑Munoz AY, Weinstein J, Wald R. eGFR decline after SGLT2 in‑
hibitor initiation: the tortoise and the hare reimagined. Kidney360. 2021; 2:
1042‑1047.
87 Chertow GM, Vart P, Jongs N, et al. Effects of dapagliflozin in stage 4
chronic kidney disease. J Am Soc Nephrol. 2021; 32: 2352‑2361.
88 Perkovic V, Jardine MJ, Neal B, et al. Canagliflozin and renal out
comes in type 2 diabetes and nephropathy. N Engl J Med. 2019; 380:
2295‑2306.
89 Cowan A, Jeyakumar N, Kang Y, et al. Fracture risk of sodium ‑glucose
cotransporter ‑2 inhibitors in chronic kidney disease. Clin J Am Soc Nephrol.
2022; 17: 835‑842.
90 American Diabetes Association Professional Practice C. 2. Classifica‑
tion and diagnosis of diabetes: standards of medical care in diabetes ‑2022.
Diabetes Care. 2022; 45: S17 ‑S38.
91 Heidenreich PA, Bozkurt B, Aguilar D, et al. 2022 AHA/ACC/HFSA
guideline for the management of heart failure: a report of the American Col‑
lege of Cardiology / American Heart Association Joint Committee on Clinical
Practice Guidelines. Circulation. 2022; 145: e895 ‑e1032.
92 Kadowaki T, Yamamoto F, Taneda Y, et al. Effects of anti diabetes med‑
ications on cardiovascular and kidney outcomes in Asian patients with type
2 diabetes: a rapid evidence assessment and narrative synthesis. Expert
Opin Drug Saf. 2021; 20: 707‑720.
93 Goldenberg RM, Ahooja V, Clemens KK, et al. Practical considerations
and rationale for glucagon ‑like peptide ‑1 receptor agonist plus sodium‑
‑dependent glucose cotransporter ‑2 inhibitor combination therapy in type 2
diabetes. Can J Diabetes. 2021; 45: 291‑302.
94 Yang Y, Zhao C, Ye Y, et al. Prospect of sodium ‑glucose co ‑transporter
2 inhibitors combined with insulin for the treatment of type 2 diabetes. Front
Endocrinol (Lausanne). 2020; 11: 190.
95 Patoulias D, Papadopoulos C, Doumas M. Sodium ‑glucose cotransport
er 2 inhibitors and heart failure decompensation: considerations on body
composition and skeletal mass. Pol Arch Intern Med. 2021; 131: 16168.
96 Brown E, Heerspink HJL, Cuthbertson DJ, Wilding JPH. SGLT2 inhibi‑
tors and GLP ‑1 receptor agonists: established and emerging indications. Lan
cet. 2021; 398: 262‑276.
97 Brown E, Wilding JPH, Barber TM, et al. Weight loss variability with
SGLT2 inhibitors and GLP ‑1 receptor agonists in type 2 diabetes mellitus and
obesity: mechanistic possibilities. Obes Rev. 2019; 20: 816‑828.
98 Tuttle KR, Brosius FC 3rd, Cavender MA, et al. SGLT2 Inhibition for
CKD and cardiovascular disease in type 2 diabetes: report of a scientific
workshop sponsored by the National Kidney Foundation. Diabetes. 2021;
70: 1‑16.
... Nevertheless, current guidelines do not distinguish between obesity/MAFLD-related HCC and HCC associated with other modalities, e.g., chronic viral hepatitis [65]. Patients with obesity-related HCC usually present with other obesity-associated cardiometabolic comorbidities, affecting their general health status and influencing therapeutic decision-making [65,[122][123][124][125]. These patients are usually diagnosed at an older age at stages BCLC C or BCLC D, even though without the obvious manifestations of cirrhosis [65,126]. ...
... Another example is the relatively new class of anti-diabetic drugs, sodium-glucose cotransporter-2 inhibitors (SGLT2i), which promote weight loss [124], are effective against NAFLD and have recently shown promising results in individuals with T2DM and HCC [134]. In addition to improving metabolic markers, such as glycated hemoglobin and BMI, the possible benefits of SGLT2i in HCC could be mediated by the wealth of their pleiotropic actions, as these drugs are known to alleviate systemic inflammation, improve cell bioenergetics, and down-regulate oxidative stress [135]. ...
Article
Full-text available
The escalating global prevalence of obesity and its intricate association with the development of hepatocellular carcinoma (HCC) pose a substantial challenge to public health. Obesity, acknowledged as a pervasive epidemic, is linked to an array of chronic diseases, including HCC, cat-alyzing the need for a comprehensive understanding of its molecular underpinnings. Notably, HCC has emerged as a leading malignancy with rising incidence and mortality. The transition from viral etiologies to the prominence of metabolic dysfunction-associated fatty liver disease (MAFLD)-related HCC underscores the urgent need to explore the intricate molecular pathways linking obesity and hepatic carcinogenesis. This review delves into the interwoven landscape of molecular carcinogenesis in the context of obesity-driven HCC while also navigating using the current therapeutic strategies and future prospects for combating obesity-related HCC. We underscore the pivotal role of obesity as a risk factor and propose an integrated approach encompassing lifestyle interventions, pharma-cotherapy, and the exploration of emerging targeted therapies. As the obesity-HCC nexus continues to challenge healthcare systems globally, a comprehensive understanding of the intricate molecular mechanisms and innovative therapeutic strategies is imperative to alleviate the rising burden of this dual menace.
... These improvements were reported with the use of SGLT-2 inhibitors in this interesting meta-analysis [37]. The favorable effects of SGLT-2 inhibitors among patients with HF could be attributed to the increased production of ketones, which, in turn, ameliorates the mitochondrial dysfunction observed in HF and increases Adenosine triphosphate (ATP) production, thus leading to improved ventricular contractility [38][39][40][41]. More specifically, SGLT-2 inhibitors, via changes in intracellular sodium and intracellular calcium concentrations, may result in improved ventricular contractility and fewer cardiac arrhythmias [38][39][40][41]. ...
... The favorable effects of SGLT-2 inhibitors among patients with HF could be attributed to the increased production of ketones, which, in turn, ameliorates the mitochondrial dysfunction observed in HF and increases Adenosine triphosphate (ATP) production, thus leading to improved ventricular contractility [38][39][40][41]. More specifically, SGLT-2 inhibitors, via changes in intracellular sodium and intracellular calcium concentrations, may result in improved ventricular contractility and fewer cardiac arrhythmias [38][39][40][41]. Furthermore, cardiac inflammation and the subsequently observed cardiac fibrosis are attenuated by the use of SGLT-2 inhibitors. ...
Article
Full-text available
The nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome in the kidney and the heart is increasingly being suggested to play a key role in mediating inflammation. In the kidney, NLRP3 activation was associated with the progression of diabetic kidney disease. In the heart, activation of the NLRP3 inflammasome was related to the enhanced release of interleukin-1β (IL-1β) and the subsequent induction of atherosclerosis and heart failure. Apart from their glucose-lowering effects, SGLT-2 inhibitors were documented to attenuate activation of the NLRP3, thus resulting in the constellation of an anti-inflammatory milieu. In this review, we focus on the interplay between SGLT-2 inhibitors and the inflammasome in the kidney, the heart and the neurons in the context of diabetes mellitus and its complications.
... The distinctive mechanism of action of SGLT2i, which involves enhanced renal glucose excretion resulting in a net energy loss, could also make SGLT2i good candidates for managing obesity, especially given their [??Do you mean "its" (obesity's)?] relationship with CAD and diabetes [18]. However, there are no sufficient data on the safety of using SGLT2i before and during pPCI in diabetic STEMI patients who have a high risk of CI-AKI. ...
Article
Full-text available
Background: It has been demonstrated that there is a significant reduction in the incidence of cardiovascular events, mortality rates, and worsening kidney disease in patients using sodium-glucose cotransporter 2 inhibitors (SGLT2i). However, there is limited information about the effect of SGLT2i on the incidence of contrast-induced acute kidney injury (CI-AKI) in patients undergoing primary percutaneous intervention (pPCI). Aims: Our research was focused on examining how SGLT2i exposure impacts CI-AKI occurrence in patients with ST-segment elevation myocardial infarction (STEMI) and undergoing pPCI. Results: This retrospective, single-center, case-control study included diabetic patients diagnosed with STEMI who underwent pPCI in a tertiary healthcare center between 2021 and 2022. The study population included SGLT2i users (n = 130) and non-SGLT2i users (n = 165). Inverse probability propensity score weighting and doubly robust estimation were performed to decrease bias and to balance covariate distribution for estimating average treatment for those treated. In a doubly robust inverse probability weighted regression model, in which covariates were balanced, CI-AKI risk was also found to be lower in the SGLT2i-user group (OR: 0.86 [0.76-0.98]; 95% CI; P = 0.028). In addition, ejection fraction, admission creatinine, albumin, and volume of contrast media were found to be independent predictors of CI-AKI in patients presenting with STEMI and undergoing pPCI. Conclusion: Our study provides evidence supporting the potential protective effect of SGLT2i against CI-AKI in diabetic patients presenting with STEMI and undergoing pPCI.
... SGLT2i, also known as gliflozins, comprise a relatively new class of medications initially approved to treat T2DM that increase urinary glucose excretion by inhibiting glucose reabsorption in the proximal tubule of the nephron. 23,102 Beyond treating T2DM, however, SGLT2i were found to exhibit significant cardiovascular and renal benefits. 103,104 Various major trials showed significant prevention of MACE, to varying degrees, in those with T2D and either established cardiovascular disease or high cardiovascular risk. ...
Article
Polycystic ovary syndrome (PCOS) is a complex endocrinopathy affecting many women of reproductive age. Although its physiology is poorly understood, hyper-androgenemia and insulin resistance play a pivotal role in this complex syndrome, predisposing patients to a variety of cardiovascular and metabolic modalities. Current therapeutic options, including lifestyle modifications and medications, often do not satisfactorily improve clinical outcomes. SGLT2 inhibitors (SGLT-2i) are a novel option which can potentially improve many hormonal and metabolic parameters for patients with PCOS, though the net cardiovascular effects remain under investigation in this population of patients with PCOS. Overall, the use of SGLT-2i may be associated with beneficial somatometric, metabolic and hormonal outcomes of PCOS. To date, all available studies have recorded body mass index, waist and hip circumference, and fat mass reductions, improved insulin and androgen levels, and reduced blood pressure. The aim of the present review is to summarise PCOS-related manifestations and mechanisms leading to cardiovascular disease, to explore the cardiometabolic impact of SGLT2i on PCOS, and to critically analyse the cardiometabolic and hormonal outcomes of the recent studies on the use of SGLT2i in women with PCOS.
... Thrombosis has a significant role in the pathogenesis of PAD as opposed to CAD, where weight, atherosclerosis is the predominant factor [97]. Another potential explanation is that patients with obesity and PAD receive more aggressive medical treatment and have better outcomes in some of the studies in comparison to patients with normal weight who might be less aggressively treated because of their overall lower risk factor profile [98][99][100]. Other cardinal pathophysiologic mechanisms related to obesity that could explain this phenomenon include higher energy reserves, inflammatory preconditioning, an anti-inflammatory immune profile, endotoxin neutralization, adrenal steroid synthesis, renin-angiotensin system activation, cardioprotective metabolic effects, and prevention of muscle wasting [25]. ...
Article
Full-text available
Purpose of Review Obesity is a significant public health problem and a major risk factor for the development and progression of atherosclerosis and its cardiovascular manifestations. Lower extremity peripheral artery disease (PAD) affects 3%–10% of the Western population and, if left untreated, can lead to devastating outcomes with both an increased risk of morbidity and mortality. Interestingly, the association between obesity and PAD remains debatable. Whereas it is well known that PAD and obesity frequently overlap in the same patients, many studies have demonstrated a negative association between obesity and PAD and a protective effect of obesity on disease development and progression, a phenomenon described as the “obesity paradox.” Possible mechanisms for this paradox may include genetic background, as assessed by mendelian randomization studies, adipose tissue dysfunction, and body fat distribution rather than adiposity, while other factors, such as sex, ethnicity, sarcopenia in the elderly population, or aggressive treatment of co-existing metabolic conditions in individuals with obesity compared to those with normal weight, could have some impact as well. Recent Findings Few reviews and meta-analyses examining systematically the relationship between obesity and PAD exist. The impact of PAD development due to the presence of obesity remains largely controversial. However, the most current evidence, backed by a recent meta-analysis, suggests a potential protective role of a higher body mass index on PAD-related complications and mortality. Summary In this review, we discuss the association between obesity and PAD development, progression, and management, and the potential pathophysiologic mechanisms linking the two diseases.
... As the obesity epidemic is on the rise worldwide, novel treatment strategies are being developed [12,17]. Apart from classical treatment options, such as glucagon-like peptide-1 (GLP-1) receptor agonists [18][19][20], researchers have also focused on the microbiome as a potential target to combat obesity [12,[20][21][22][23]. ...
Article
Full-text available
Obesity and obesity-associated disorders pose a major public health issue worldwide. Apart from conventional weight loss drugs, next-generation probiotics (NGPs) seem to be very promising as potential preventive and therapeutic agents against obesity. Candidate NGPs such as Akkermansia muciniphila, Faecalibacterium prausnitzii, Anaerobutyricum hallii, Bacteroides uniformis, Bacteroides coprocola, Parabacteroides distasonis, Parabacteroides goldsteinii, Hafnia alvei, Odoribacter laneus and Christensenella minuta have shown promise in preclinical models of obesity and obesity-associated disorders. Proposed mechanisms include the modulation of gut flora and amelioration of intestinal dysbiosis, improvement of intestinal barrier function, reduction in chronic low-grade inflammation and modulation of gut peptide secretion. Akkermansia muciniphila and Hafnia alvei have already been administered in overweight/obese patients with encouraging results. However, safety issues and strict regulations should be constantly implemented and updated. In this review, we aim to explore (1) current knowledge regarding NGPs; (2) their utility in obesity and obesity-associated disorders; (3) their safety profile; and (4) their therapeutic potential in individuals with overweight/obesity. More large-scale, multicentric and longitudinal studies are mandatory to explore their preventive and therapeutic potential against obesity and its related disorders.
Article
Full-text available
Purpose of Review To examine the epidemiological data on obesity and leukemia; evaluate the effect of obesity on leukemia outcomes in childhood acute lymphoblastic leukemia (ALL) survivors; assess the potential mechanisms through which obesity may increase the risk of leukemia; and provide the effects of obesity management on leukemia. Preventive (diet, physical exercise, obesity pharmacotherapy, bariatric surgery) measures, repurposing drugs, candidate therapeutic agents targeting oncogenic pathways of obesity and insulin resistance in leukemia as well as challenges of the COVID-19 pandemic are also discussed. Recent Findings Obesity has been implicated in the development of 13 cancers, such as breast, endometrial, colon, renal, esophageal cancers, and multiple myeloma. Leukemia is estimated to account for approximately 2.5% and 3.1% of all new cancer incidence and mortality, respectively, while it represents the most frequent cancer in children younger than 5 years. Current evidence indicates that obesity may have an impact on the risk of leukemia. Increased birthweight may be associated with the development of childhood leukemia. Obesity is also associated with worse outcomes and increased mortality in leukemic patients. However, there are several limitations and challenges in meta-analyses and epidemiological studies. In addition, weight gain may occur in a substantial number of childhood ALL survivors while the majority of studies have documented an increased risk of relapse and mortality among patients with childhood ALL and obesity. The main pathophysiological pathways linking obesity to leukemia include bone marrow adipose tissue; hormones such as insulin and the insulin-like growth factor system as well as sex hormones; pro-inflammatory cytokines, such as IL-6 and TNF-α; adipocytokines, such as adiponectin, leptin, resistin, and visfatin; dyslipidemia and lipid signaling; chronic low-grade inflammation and oxidative stress; and other emerging mechanisms. Summary Obesity represents a risk factor for leukemia, being among the only known risk factors that could be prevented or modified through weight loss, healthy diet, and physical exercise. Pharmacological interventions, repurposing drugs used for cardiometabolic comorbidities, and bariatric surgery may be recommended for leukemia and obesity-related cancer prevention.
Article
Full-text available
Overweight, obesity, and type 2 diabetes mellitus pose global health problems that are ever-increasing. A chronic low-grade inflammatory status and the presence of various pro-inflammatory markers either in circulation or within dysfunctional metabolic tissues are well established. The presence of these factors can, to some extent, predict disease development and progression. A central role is played by the presence of dysfunctional adipose tissue, liver dysfunction, and skeletal muscle dysfunction, which collectively contribute to the increased circulatory levels of proinflammatory factors. Weight loss and classical metabolic interventions achieve a decrease in many of these factors’ circulating levels, implying that a better understanding of the processes or even the modulation of inflammation may alleviate these diseases. This review suggests that inflammation plays a significant role in the development and progression of these conditions and that measuring inflammatory markers may be useful for assessing disease risk and development of future treatment methods.
Article
Full-text available
Obesity is characterized by excessive adipose tissue accumulation, which impacts physiological, metabolic, and immune functions. Several respiratory infections, including bacterial pneumonia, influenza, and coronavirus disease 2019, appear to be linked to unfavorable results in individuals with obesity. These may be attributed to the direct mechanical/physiological effects of excess body fat on the lungs’ function. Notably, adipose tissue dysfunction is associated with a low-grade chronic inflammatory status and hyperleptinemia, among other characteristics. These have all been linked to immune system dysfunction and weakened immune responses to these infections. A better understanding and clinical awareness of these risk factors are necessary for better disease outcomes.
Article
Full-text available
Purpose of Review This narrative review appraises research data on the potentially harmful effect of obesity and insulin resistance (IR) co-existence with type 1 diabetes mellitus (T1DM)-related cardiovascular (CVD) complications and evaluates possible therapeutic options. Recent Findings Obesity and IR have increasingly been emerging in patients with T1DM. Genetic, epigenetic factors, and subcutaneous insulin administration are implicated in the pathogenesis of this coexistence. Accumulating evidence implies that the concomitant presence of obesity and IR is an independent predictor of worse CVD outcomes. Summary The prevalence of obesity and IR has increased in patients with T1DM. This increase can be partly attributed to general population trends but, additionally, to iatrogenic weight gain caused by insulin treatment. This association might be the missing link explaining the excess CVD burden observed in patients with T1DM despite optimal glycemic control. Data on newer agents for type 2 diabetes mellitus (T2DM) treatment are unraveling novel ways to challenge this aggravating coexistence.
Article
Full-text available
The interplay between the environment and the immune cells is linked to metabolic homeostasis under pathophysiologic conditions. Diabetes mellitus type 2 (T2D) is considered an immune-related inflammatory disease, in which the adipose tissue macrophages (ATMs) are key players orchestrating metabolic chronic meta-inflammation and contributing to the pathogenesis of metabolic disease. However, the molecular regulators that integrate the environmental signals to control ATM activation and adipose inflammation during obesity and T2D remain unclear. Epigenetics mechanisms constitute important parameters in metabolic homeostasis, obesity and T2D via the integration of the environmental factors to the transcriptional regulation of gene programs. In a very recent study published in Diabetes by Kong et al. BAF60a has been identified as a key chromatin remodeling checkpoint factor that associates obesity-associated stress signals with meta-inflammation and systemic homeostasis. Furthermore, this work uncovers Atf3 as an important downstream effector in BAF60a-mediated chromatin remodeling and transcriptional reprogramming of macrophage activation in adipose tissue. The findings of this research may contribute to the development of new therapeutic approaches for obesity-induced metabolic inflammation and associated metabolic disorders.
Article
Full-text available
The entire spectrum of nonalcoholic fatty liver disease (NAFLD) ranging from fatty liver to cirrhosis has been considered as the result of specific metabolic pathways and mediators, gut barrier function alterations and inflammatory responses. Previous studies have associated intestinal microbiota with NAFLD pathogenesis, focusing mostly on bacteria. In a recent study by Demir et al. in the Journal of Hepatology, researchers characterized the fecal mycobiome of patients with NAFLD and controls. NAFLD severity was linked to a specific fecal mycobiome signature, particularly in patients without obesity, highlighting previously undescribed aspects of the non obese phenotype of NAFLD. There has recently been a growing interest in the pathophysiology and progression of non obese NAFLD, as its actual incidence seems to be higher than previously described. Moreover, the authors demonstrated that in subjects with NAFLD and advanced fibrosis, there was an augmented systemic immune response to Candida albicans. Amphotericin B, which has been widely regarded as an antifungal with a good safety profile, low rate of resistance and high efficacy, has already been shown to prevent liver injury and steatosis in mice. Similarly in this study when germ-free mice colonized with feces from patients with NASH were fed with a Western diet, treatment with amphotericin B protected against steatohepatitis and liver fibrosis. In conclusion, this study has provided novel insights into the fecal mycobiome composition in advanced NAFLD especially in the non obese population while suggesting a role for antifungal therapy in the management of NAFLD.
Article
Full-text available
Purpose of Review Weight loss has multiple beneficial effects on body composition and metabolism, but whether these depend on the rate at which body weight is lost is not clear. We analyzed data from studies in which the same amount of weight loss was induced rapidly or gradually. Recent Findings Thirteen studies were included in which the same percentage weight loss was achieved at slow or fast rates (range: 0.2 to 3.2 kg/week) by means of dietary calorie restriction, exercise, and bariatric surgery. Faster rates of weight loss may result in more fat-free mass and less fat mass being lost during the dynamic phase of weight reduction compared with slower rates of weight loss, in conjunction with greater declines in resting energy expenditure. However, these differences are attenuated after 2–4 weeks of stabilization at the new, lower body weight, and do not affect the rate and amount of weight regain 9–33 months later (nor the tissue composition of regained weight). Differences in waist circumference, visceral and liver fat contents, resting blood pressure, fasting blood lipid profile, and insulin and adipokine concentrations in response to different rates of weight loss are trivial. The decline in fasting glucose concentration and the improvement in insulin sensitivity after 6–11% weight loss are both greater with rapid than gradual weight loss, but not different after 18–20% weight loss. Summary Changes in body composition and metabolism after losing the same amount of body weight at different rates are largely similar, and occasional differences are likely not meaningful clinically for the long-term management of obesity and cardiometabolic diseases.
Article
Background: Omentin-1, a newly discovered adipokine, is implicated in the modulation of the adipose phenotype, ameliorating systemic metabolism and exhibiting anti-atherogenic, antioxidative, cardioprotective, anti-inflammatory and insulin-sensitizing properties. Our goal was to explore circulating omentin-1 in subclinical hypothyroidism (SH) and determine its correlations with cardiometabolic risk factors. Methods: In a large case-control and interventional longitudinal study, serum omentin-1, metabolic and lipid parameters, inflammatory biomarkers, classic adipocytokines and cardiovascular risk factors were assessed in 120 consecutive patients with SH and 120 healthy controls matched on age, gender and date of blood draw. Sixteen patients with SH were administered L-T4 and, after six months, circulating omentin-1 and other biomarkers were determined. Results: SH subjects presented significantly decreased circulating omentin-1 than control individuals (p<0.001). In all study participants, omentin-1 was negatively correlated with TSH, anti-thyroid antibodies, HOMA-IR, C-peptide, lipid and inflammatory biomarkers, adipokines and cardiovascular risk factors, including Framingham score and apolipoprotein B. Omentin-1 was positively associated with adiponectin and HDL-C. Circulating omentin-1 was independently associated with SH occurrence, above and beyond clinical and cardiometabolic factors (p=0.04). TSH was a negative independent predictor of serum omentin-1 levels (p<0.001). L-T4 treatment did not alter considerably the lower omentin-1 levels in treated SH patients (p=0.07). Conclusions: Omentin-1 may be a useful non-invasive biomarker reflecting cardiometabolic risk as well as a promising therapeutic target. More mechanistic and larger prospective studies shedding light on the pathogenetic role of omentin-1 in SH are required to confirm these findings.
Article
Background and objectives: Sodium-glucose cotransporter-2 (SGLT2) inhibitors have been associated with a higher risk of skeletal fractures in some randomized, placebo-controlled trials. Secondary hyperparathyroidism and increased bone turnover (also common in CKD) may contribute to the observed fracture risk. We aimed to determine if SGLT2 inhibitor use associates with a higher risk of fractures compared with dipeptidyl peptidase-4 (DPP-4) inhibitors, which have no known association with fracture risk. We hypothesized that this risk, if present, would be greatest in patients with lower eGFR. Design, setting, participants, & measurements: We conducted a population-based cohort study in Ontario, Canada between 2015 and 2019 using linked provincial administrative data to compare the incidence of fracture between new users of SGLT2 inhibitors and DPP-4 inhibitors. We used inverse probability of treatment weighting on the basis of propensity scores to balance the two groups of older adults (≥66 years of age) on indicators of baseline health. We compared the 180- and 365-day cumulative incidence rates of fracture between groups. Prespecified subgroup analyses were conducted by eGFR category (≥90, 60 to <90, 45 to <60, and 30 to <45 ml/min per 1.73 m2). Weighted hazard ratios were obtained using Cox proportional hazard regression. Results: After weighting, we identified a total of 38,994 new users of a SGLT2 inhibitor and 37,449 new users of a DPP-4 inhibitor and observed a total of 342 fractures at 180 days and 689 fractures at 365 days. The weighted 180- and 365-day risks of a fragility fracture did not significantly differ between new users of a SGLT2 inhibitor versus a DPP-4 inhibitor: weighted hazard ratio, 0.95 (95% confidence interval, 0.79 to 1.13) and weighted hazard ratio, 0.88 (95% confidence interval, 0.88 to 1.00), respectively. There was no observed interaction between fracture risk and eGFR category (P=0.53). Conclusions: In this cohort study of older adults, starting a SGLT2 inhibitor versus DPP-4 inhibitor was not associated with a higher risk of skeletal fracture, regardless of eGFR.
Article
Aim The “2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure” replaces the “2013 ACCF/AHA Guideline for the Management of Heart Failure” and the “2017 ACC/AHA/HFSA Focused Update of the 2013 ACCF/AHA Guideline for the Management of Heart Failure.” The 2022 guideline is intended to provide patient-centric recommendations for clinicians to prevent, diagnose, and manage patients with heart failure. Methods A comprehensive literature search was conducted from May 2020 to December 2020, encompassing studies, reviews, and other evidence conducted on human subjects that were published in English from MEDLINE (PubMed), EMBASE, the Cochrane Collaboration, the Agency for Healthcare Research and Quality, and other relevant databases. Additional relevant clinical trials and research studies, published through September 2021, were also considered. This guideline was harmonized with other American Heart Association/American College of Cardiology guidelines published through December 2021. Structure Heart failure remains a leading cause of morbidity and mortality globally. The 2022 heart failure guideline provides recommendations based on contemporary evidence for the treatment of these patients. The recommendations present an evidence-based approach to managing patients with heart failure, with the intent to improve quality of care and align with patients’ interests. Many recommendations from the earlier heart failure guidelines have been updated with new evidence, and new recommendations have been created when supported by published data. Value statements are provided for certain treatments with high-quality published economic analyses.
Article
Most physicians do not know, or do not remember, the name of phlorizin. Hence this molecule has a major historical importance because it was the precursor of gliflozins, a new class of oral antidiabetic drugs with recent therapeutic perspectives beyond diabetes. This article recalls the history of phlorizin: its discovery in the 19th century by De Koninck and Stas, the demonstration of its ability to induce glucosuria and reduce hyperglycaemia by von Mering, its use to demonstrate the concept of glucose toxicity by the team of DeFronzo and finally the development of selective (phlorizin being not selective) sodium-glucose cotransporter type 2 inhibitors (gliflozins) which block glucose reabsorption in renal tubules. Gliflozins have increasing therapeutic indications, not only in type 2 diabetes, but also in cardiology and nephrology among non-diabetic people with heart failure or renal insufficiency.