ArticlePDF Available

Management of hyperglycaemia in type 2 diabetes, 2018. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD)

Authors:

Abstract and Figures

The American Diabetes Association and the European Association for the Study of Diabetes convened a panel to update the prior position statements, published in 2012 and 2015, on the management of type 2 diabetes in adults. A systematic evaluation of the literature since 2014 informed new recommendations. These include additional focus on lifestyle management and diabetes self-management education and support. For those with obesity, efforts targeting weight loss, including lifestyle, medication and surgical interventions, are recommended. With regards to medication management, for patients with clinical cardiovascular disease, a sodium–glucose cotransporter-2 (SGLT2) inhibitor or a glucagon-like peptide-1 (GLP-1) receptor agonist with proven cardiovascular benefit is recommended. For patients with chronic kidney disease or clinical heart failure and atherosclerotic cardiovascular disease, an SGLT2 inhibitor with proven benefit is recommended. GLP-1 receptor agonists are generally recommended as the first injectable medication.
This content is subject to copyright. Terms and conditions apply.
CONSENSUS REPORT
Management of hyperglycaemia in type 2 diabetes, 2018. A consensus
report by the American Diabetes Association (ADA) and the European
Association for the Study of Diabetes (EASD)
Melanie J. Davies
1,2
&David A. DAlessio
3
&Judith Fradkin
4
&Walter N. Kernan
5
&Chantal Mathieu
6
&
Geltrude Mingrone
7,8
&Peter Rossing
9,10
&Apostolos Tsapas
11
&Deborah J. Wexler
12,13
&John B. Buse
14
#European Association for the Study of Diabetes and American Diabetes Association 2018
Abstract
The American Diabetes Association and the European Association for the Study of Diabetes convened a panel to update the prior
position statements, published in 2012 and 2015, on the management of type 2 diabetes in adults. A systematic evaluation of the
literature since 2014 informed new recommendations. These include additional focus on lifestyle management and diabetes self-
management education and support. For those with obesity, efforts targeting weight loss, including lifestyle, medication and surgical
interventions, are recommended. With regards to medication management, for patients with clinical cardiovascular disease, a sodium
glucose cotransporter-2 (SGLT2) inhibitor or a glucagon-like peptide-1 (GLP-1) receptor agonist with proven cardiovascular benefit is
recommended. For patients with chronic kidney disease or clinical heart failure and atherosclerotic cardiovascular disease, an SGLT2
inhibitor with proven benefit is recommended. GLP-1 receptor agonists are generally recommended as the first injectable medication.
Keywords Cardiovascular disease .Chronic kidney disease .Costs .Glucose-lowering therapy .Guidelines .Heart failure .
Hypoglycaemia .Patient-centred care .Type 2 diabetes mellitus .Weight management
Abbreviations
ARR Absolute risk reduction
ASCVD Atherosclerotic cardiovascular
disease
CANVAS Canagliflozin Cardiovascular
Assessment Study
CKD Chronic kidney disease
CVD Cardiovascular disease
CVOT Cardiovascular outcomes trial
DKA Diabetic ketoacidosis
DPP-4 Dipeptidyl peptidase-4
DPP-4i Dipeptidyl peptidase-4 inhibitor
DSMES Diabetes self-management
education and support
EMPA-REG OUTCOME Empagliflozin, Cardiovascular
Outcome Event Trial in Type 2
Diabetes Mellitus Patients
ESRD End-stage renal disease
EXSCEL Exenatide Study of
Cardiovascular Event Lowering
GLP-1 Glucagon-like peptide-1
GLP-1 RA Glucagon-like peptide-1 receptor
agonist
HF Heart failure
LEADER Liraglutide Effect and Action in
Diabetes: Evaluation of of
Cardiovascular Outcomes Results
MACE Major adverse cardiac events
MI Myocardial infarction
MNT Medical nutrition therapy
RCT Randomised clinical trial
SGLT2 Sodiumglucose cotransporter-2
M. J. Davies and J. B. Buse were co-chairs for the Consensus Statement
Writing Group. D. A. DAlessio, J. Fradkin, W. N. Kernan and D. J.
Wexler were the writing group members for the ADA. C. Mathieu, G.
Mingrone, P. Rossing and A. Tsapas were writing group members for the
EASD.
This article is being simultaneously published in Diabetes Care and
Diabetologia by the American Diabetes Association and the European
Association for the Study of Diabetes.
*Melanie J. Davies
Melanie.davies@uhl-tr.nhs.uk
Extended author information available on the last page of the article
Diabetologia
https://doi.org/10.1007/s00125-018-4729-5
SGLT2i Sodiumglucose cotransporter-2
inhibitor
SMBG Self-monitoring of blood glucose
SU Sulfonylurea
SUSTAIN 6 Trial to Evaluate Cardiovascular
and Other Long-term Outcomes
with Semaglutide in Subjects
with Type 2 Diabetes
T2DM Type 2 diabetes mellitus
TZD Thiazolidinedione
UKPDS UK Prospective Diabetes Study
Introduction
The goals of treatment for type 2 diabetes are to prevent or
delay complications and maintain quality of life (Fig. 1). This
requires control of glycaemia and cardiovascular risk factor
management, regular follow-up and, importantly, a patient-
centred approach to enhance patient engagement in self-care
activities [1]. Careful consideration of patient factors and pre-
ferences must inform the process of individualising treatment
goals and strategies [2,3].
This consensus report addresses the approaches to manage-
ment of glycaemia in adults with type 2 diabetes, with the goal
of reducing complications and maintaining quality of life in
the context of comprehensive cardiovascular risk manage-
ment and patient-centred care. The principles of how this
can be achieved are summarised in Fig. 1and underpin the
approach to management and care. These recommendations
are not generally applicable to patients with monogenic dia-
betes, secondary diabetes or type 1 diabetes, or to children.
Data sources, searches and study selection
The writing group accepted the 2012 [4] and 2015 [5] editions
of this position statement as a starting point. To identify newer
evidence, a search was conducted on PubMed for randomised
clinical trials (RCTs), systematic reviews and meta-analyses
published in English between 1 January 2014 and 28
February 2018; eligible publications examined the effective-
ness or safety of pharmacological or non-pharmacological in-
terventions in adults with type 2 diabetes mellitus. Reference
lists were scanned in eligible reports to identify additional arti-
cles relevant to the subject. Details on the keywords and the
search strategy are available at https://doi.org/10.17632/
h5rcnxpk8w.1. Papers were grouped according to subject and
the authors reviewed this new evidence to inform the consensus
recommendations. The draft consensus recommendations were
peer reviewed (see Acknowledgements), and suggestions
incorporated as deemed appropriate by the authors.
Nevertheless, though evidence based, the recommendations
presented herein are the opinions of the authors.
The rationale, importance and context
of glucose-lowering treatment
Lifestyle management, including medical nutrition therapy
(MNT), physical activity, weight loss, counselling for
smoking cessation, and psychological support, often delivered
in the context of diabetes self-management education and sup-
port (DSMES), are fundamental aspects of diabetes care. The
expanding number of glucose-lowering treatmentsfrom be-
havioural interventions to medications and surgeryand
growing information about their benefits and risks provides
more options for people with diabetes and providers, but can
complicate decision making. In this consensus statement, we
attempt to provide an approach that summarises a large body
of recent evidence for practitioners in the USA and Europe.
Marked hyperglycaemia is associated with symptoms in-
cluding frequent urination, thirst, blurred vision, fatigue and
recurring infections. Beyond alleviating symptoms, the aim of
blood glucose lowering (hereafter, referred to as glycaemic
management) is to reduce long-term complications of diabe-
tes. Good glycaemic management yields substantial and en-
during reductions in onset and progression of microvascular
complications. This benefit has been demonstrated most clear-
ly early in the natural history of the disease in studies using
metformin,sulfonylureasand insulin but is supported by more
recent studies with other medication classes. The greatest ab-
solute risk reduction (ARR) comes from improving poor
glycaemic control, and a more modest reduction results from
near normalisation of glycaemia [6]. The impact of glucose
control on macrovascular complications is less certain.
Because the benefits of intensive glucose control emerge
slowly, while the harms can be immediate, people with longer
life expectancy have more to gain from intensive glucose con-
trol. A reasonable HbA
1c
target for most non-pregnant adults
with sufficient life expectancy to see microvascular benefits
(generally ~10 years) is around 53 mmol/mol (7%) or less [6].
Glycaemic treatment targets should be individualised based
on patient preferences and goals, risk of adverse effects of
therapy (e.g. hypoglycaemia and weight gain) and patient
characteristics, including frailty and comorbid conditions [2].
Atherosclerotic cardiovascular disease (ASCVD) is the
leading cause of death in people with type 2 diabetes [7].
Diabetes confers substantial independent ASCVD risk, and
most people with type 2 diabetes have additional risk factors
such as hypertension, dyslipidaemia, obesity, physical inactiv-
ity, chronic kidney disease (CKD) and smoking. Numerous
studies have demonstrated the benefits of controlling modifi-
able ASCVD risk factors in people with diabetes. Substantial
reductions in ASCVD events and death are seen when multi-
ple ASCVD risk factors are addressed simultaneously, with
long-standing benefits [8,9]. Comprehensive implementation
of evidence-based interventions has likely contributed to the
significant reductions in ASCVD events and mortality seen in
Diabetologia
Fig. 1 Decision cycle for patient-centred glycaemic management in type 2 diabetes
Diabetologia
people with diabetes in recent decades [10]. ASCVD risk
management in its many forms is an essential part of diabetes
management that is beyond the scope of this statement, but
physiciansshould be aware of the importance of multifactorial
treatment in type 2 diabetes [7].
Glucose management: monitoring
Glycaemic management is primarily assessed with the HbA
1c
test, which was the measure studied in trials demonstrating the
benefits of glucose lowering [2]. The performance of the test is
generally excellent for National Glycohemoglobin
Standardization Program (NGSP)-certified assays and laborato-
ries (www.ngsp.org)[11]. As with any laboratory test, HbA
1c
has limitations [2]. Because there is variability in the
measurement of HbA
1c
, clinicians should exercise judgement,
particularly when the result is close to the threshold that might
prompt a change in therapy. HbA
1c
results may be discrepant
from the patients true mean glycaemia in certain racial and
ethnic groups, and in conditions that alter red blood cell
turnover, such as anaemia, end-stage renal disease (ESRD; es-
pecially with erythropoietin therapy), and pregnancy, or if an
HbA
1c
assay sensitive to haemoglobin variants is used in some-
one with sickle cell trait or other haemoglobinopathy.
Discrepancies between measured HbA
1c
and measured or re-
ported glucose levels should prompt consideration that one of
these may not be reliable [12].
Regular self-monitoring of blood glucose (SMBG) may
help with self-management and medication adjustment, partic-
ularly in individuals taking insulin. SMBG plans should be
individualised. People with diabetes and the healthcare team
should use the data in an effective and timely manner. In peo-
ple with type 2 diabetes not using insulin, routine glucose
monitoring is of limited additional clinical benefit while adding
burden and cost [13,14]. However, for some individuals, glu-
cose monitoring can provide insight into the impact of lifestyle
and medication management on blood glucose and symptoms,
particularly when combined with education and support.
Novel technologies, such as continuous or flash glucose mon-
itoring, provide more information. However, in type 2 diabe-
tes, they have been associated with only modest benefits [15].
Principles of care
Providing patient-centred care that acknowledges multi-mor-
bidity, and is respectful of and responsive to individual patient
preferences and barriers, including the differential costs of
therapies, is essential to effective diabetes management [16].
Shared decision making, facilitated by decision aids that show
the absolutebenefit and riskof alternative treatment options, is
a useful strategy to arrive at the best treatment course for an
individual [1720]. Providers should evaluate the impact of
any suggested intervention, including self-care regimens, in
the context of cognitive impairment, limited literacy, distinct
cultural beliefs and individual fears or health concerns given
their impact on treatment efficacy.
DSMES
DSMES is a key intervention to enable people with diabetes to
make informed decisions and to assumeresponsibility for day-
to-day diabetes management. DSMES is central to establish-
ing and implementing the principles of care (Fig. 1). DSMES
programmes usually involve face-to-face contact in group or
individual sessions with trained educators, and key compo-
nents are shown in the Table 1[2125]. While DSMES should
be available on an ongoing basis, critical junctures when
DSMES should occur include at diagnosis, annually, when
complications arise and during transitions in life and care [22].
DSMES programmes delivered from diagnosis can pro-
mote medication adherence,healthy eating and physical activ-
ity, and increase self-efficacy. In type 2 diabetes, high-quality
evidence has consistently shown that DSMES is a cost-
effective intervention in the healthcare systems studied.
DSMES significantly improves clinical and psychological
outcomes, improves glycaemic control, reduces hospital ad-
missions, improves patient knowledge, and reduces the risk of
all-cause mortality [22,2631]. The best outcomes are
achieved in those programmes with a theory-based and struc-
tured curriculum, and with contact time of over 10 h. While
online programmes may reinforce learning, there is little evi-
dence they are effective when used alone [27].
Suboptimal adherence, including poor persistence, to ther-
apy affects almost half of people with diabetes, leading to
suboptimal glycaemic and cardiovascular disease (CVD) risk
factor control as well as increased risk of diabetes complica-
tions, mortality, hospital admissions and healthcare costs
[3236]. Though this consensus recommendation focuses on
medication adherence (including persistence), the principles
Consensus recommendation
Providers and healthcare systems should prioritise the
delivery of patient-centred care.
Consensus recommendation
All people with type 2 diabetes should be offered access
to ongoing DSMES programmes.
Consensus recommendation
Facilitating medication adherence should be specifi-
cally considered when selecting glucose-lowering
medications.
Diabetologia
are pertinent to all aspects of diabetes care. Multiple factors
contribute to inconsistent medication use and treatment dis-
continuation, including patient-perceived lack of medication
efficacy, fear of hypoglycaemia, lack of access to medication
and adverse effects of medication [37]. Medication adherence
(including persistence) varies across medication classes and
careful consideration of these differences may help improve
outcomes [38]. Ultimately, patient preference is a major
factor driving the choice of medication. Even in cases
where clinical characteristics suggest the use of a particular
medication based on the available evidence from clinical
trials, patient preferences regarding route of administra-
tion, injection devices, side effects or cost may prevent
their use by some individuals [39].
Therapeutic inertia, sometimes referred to as clinical iner-
tia, refers to failure to intensify therapy when treatment targets
are not met. The causes of therapeutic inertia are multifacto-
rial, occurring at the level of the practitioner, patient and/or
healthcare system [40]. Interventions targeting therapeutic in-
ertia have facilitated improved glycaemic control and timely
insulin intensification [41,42]. For example, multidisciplinary
teams that include nurse practitioners or pharmacists may help
reduce therapeutic inertia [43,44]. A fragmented healthcare
system may contribute to therapeutic inertia and impair deliv-
ery of patient-centred care. A coordinated chronic care model,
including self-management support, decision support, deliv-
ery system design, clinical information systems, and commu-
nity resources and policies, promotes interaction between
more empowered patients and better prepared and proactive
healthcare teams [45].
Recommended process for glucose-lowering
medication selection: where does new
evidence from cardiovascular outcomes trials
fit in?
In prior consensus statements, efficacy in reducing
hyperglycaemia, along with tolerability and safety were pri-
mary factors in glucose-lowering medication selection. Patient
preferences, glycaemic targets, comorbidities, polypharmacy,
side effects and cost were additional important considerations.
For every individual, the choice of glucose-lowering medica-
tion should be underpinned by lifestyle management, DSMES
and the patient-centred care principles outlined in Fig. 1.
Figure 2describes our new consensus approach to glucose
lowering with medications in type 2 diabetes. Because of the
new evidence for the benefit of specific medications to reduce
mortality, heart failure (HF) and progression of renal disease
in the setting of established CVD,their use was considered
Table 1 Key components of DSMES [21,2325]
Table 1. Key components of DSMES [21, 23–25]
Evidence-based
Individualised to the needs of the person, including language and culture
Has a structured theory-driven written curriculum with supporting materials
Delivered by trained and competent individuals (educators) who are quality assured
Delivered in group or individual settings
Aligns with the local population needs
Supports the person and their family in developing attitudes, beliefs, knowledge and skills to
self-manage diabetes
Includes core content, i.e. diabetes pathophysiology and treatment options; medication usage; moni-
toring, preventing, detecting and treating acute and chronic complications; healthy coping with
psychological issues and concerns; problem solving and dealing with special situations (i.e. travel,
fasting)
Available to patients at critical times (i.e. at diagnosis, annually, when complications arise and when
transitions in care occur)
Includes monitoring of patient progress, including health status, quality of life
Quality audited regularly
DSMES is a critical element of care for all people with diabetes and is the ongoing process of facilitating the knowledge, skills and ability
necessary for diabetes self-care as well as activities that assist a person implementing and sustaining behaviours needed to manage
their diabetes on an ongoing basis
National organisations in the USA and Europe have published standards to underpin DSMES. In the USA these are defined as DSMES
‘Services’ whereas in Europe they are often referred to as ‘programmes’. Nevertheless, the broad components are similar
Diabetologia
Fig. 2 Glucose-lowering medication in type 2 diabetes: overall approach
Diabetologia
compelling in this patient group. Thus, we recommend that
providers consider a history of CVD very early in the process
of treatment selection. Other factors affect the choice of
glucose-lowering medications, particularly in the setting of
patient-centred care. In addition to CVD, we recommend early
consideration of weight, hypoglycaemic risk, treatment cost
and other patient-related factors that may influence treatment
selection (Figs 2,3,4,5,6).
Implications of new evidence from cardiovascular
outcomes trials
The major change from prior consensus reports is based on
new evidence that specific sodiumglucose cotransporter-2
(SGLT2) inhibitors or glucagon-like peptide-1 (GLP-1) recep-
tor agonists improve cardiovascular outcomes, as well as sec-
ondary outcomes such as HF and progression of renal disease,
in patients with established CVD or CKD. Therefore, an im-
portant early step in this new approach (Fig. 3)istoconsider
the presence or absence of ASCVD, HF and CKD, conditions
in aggregate affecting 1525% of the population with type 2
diabetes. While the new evidence supporting the use of par-
ticular medications in patients who also have established CVD
or are at high risk of CVD is derived from large cardiovascular
outcomes trials (CVOTs) demonstrating substantial benefits
over 25 years, it is important to remember that each trial
constitutes a single experiment. Within each drug class, results
have been heterogeneous. It is not clear whether there are true
drug class effects with different findings for individual medi-
cations due to differences in trial design and conduct, or
whether there are real differences between medications within
a drug class due to properties of the individual compounds.
Where the current evidence is strongest for a specific medica-
tion within a class, it is noted. The ADAsStandards of med-
ical care in diabeteswill align with this document and will be
updated to reflect new evidence as it emerges from ongoing
clinical trials.
ASCVD is defined somewhat differently across trials, but all
trials enrolled individuals with established CVD (e.g. myocar-
dial infarction [MI], stroke, any revascularisation procedure)
while variably including related conditions compatible with
clinically significant atherosclerosis (e.g. transient ischaemic
attack, hospitalised unstable angina, amputation, congestive
heart failure New York Heart Association [NYHA] class II
III, >50% stenosis of any artery, symptomatic or asymptomatic
coronary artery disease documented by imaging, CKD with
estimated GFR [eGFR] <60 ml min
-1
[1.73 m]
-2
). Most trials
also included a risk factor onlygroup with entry criteria based
on age and usually the presence of two or more cardiac risk
factors [46]. Trials were designed to evaluate cardiovascular
safety (i.e. statistical non-inferiority compared with placebo),
but several showed ASCVD outcome benefit (i.e. statistical
superiority compared with placebo), including, in some cases,
mortality.
Among GLP-1 receptor agonists, liraglutide, studied in the
Liraglutide Effect and Action in Diabetes: Evaluation of
Cardiovascular Outcomes Results (LEADER) trial (n= 9340)
demonstrated an ARR of 1.9% with an HR of 0.87 (95% CI
0.78, 0.97; p= 0.01 for superiority) for the primary composite
outcome of cardiovascular death, non-fatal MI and non-fatal
stroke (major adverse cardiac events [MACE]) compared with
placebo over 3.8 years. Each component of the composite con-
tributed to the benefit, and the HR for cardiovascular death was
0.78 (95% CI 0.66, 0.93; p= 0.007; ARR 1.7%). The LEADER
trial also demonstrated an HR of 0.85 (95% CI, 0.74 to 0.97;
p= 0.02; ARR 1.4%) for all-cause mortality [47]. In the Trial to
Evaluate Cardiovascular and Other Long-term Outcomes with
Semaglutide in Subjects with Type 2 Diabetes (SUSTAIN 6)
(n= 3297), semaglutide compared with placebo demonstrated
an ARR of 2.3% with HR 0.74 for MACE (95% CI 0.58, 0.95;
p= 0.02 for superiority) over 2.1 years, but the reduction in
events appeared to be driven by the rate of stroke, rather than
CVD death [48]. The Exenatide Study of Cardiovascular Event
Lowering (EXSCEL) compared exenatide extended-release
with placebo over 3.2 years in 14,752 participants with type 2
diabetes. While the medication was safe (non-inferior), the HR
for MACE in the entire trial was 0.91 (95% CI 0.83, 1.0; p=
0.06) not reaching the threshold for demonstrated superiority vs
placebo; ARR was 0.8% [49]. All-cause death was lower in the
exenatide arm (ARR 1%, HR 0.86 [95% CI 0.77, 0.97]), but it
was not considered to be statistically significant in the hierar-
chical testing procedure applied. Lixisenatide, a short-acting
GLP-1 receptor agonist, did not demonstrate CVD benefit or
harm in a trial of patients recruited within 180 days of an acute
coronary syndrome admission [50]. Taken together, it appears
that among patients with established CVD, some GLP-1 recep-
tor agonists may provide cardiovascular benefit, with the evi-
dence of benefit strongest for liraglutide, favourable for
semaglutide, and less certain for exenatide. There is no evi-
dence of cardiovascular benefit with lixisenatide. Adverse ef-
fects for the class are discussed in the section The full range of
therapeutic options: lifestyle management, medication and obe-
sity management.
Among the SGLT2 inhibitors, empagliflozin compared with
placebo was studied in the Empagliflozin, Cardiovascular
Outcome Event Trial in Type 2 Diabetes Mellitus Patients
Consensus recommendation
Among patients with type 2 diabetes who have
established ASCVD, SGLT2 inhibitors or GLP-1
receptor agonists with proven cardiovascular benefit
are recommended as part of glycaemic management
(Figs 2 and 3).
Diabetologia
Fig. 3 Choosing glucose-lowering medication in those with established atherosclerotic cardiovascular disease (ASCVD) or chronic kidney disease
(CKD)
Diabetologia
Fig. 4 Choosing glucose-lowering medication if compelling need to minimise weight gain or promote weight loss
Diabetologia
Fig. 5 Choosing glucose-lowering medication if compelling need to minimise hypoglycaemia
Diabetologia
(EMPA-REG OUTCOME) in 7020 participants with type 2
diabetes and CVD. With a median follow-up of 3.1 years, the
ARR was 1.6% and the HR was 0.86 (95% CI 0.74, 0.99; p=
0.04 for superiority) for the primary composite endpoint of non-
Fig. 6 Choosing glucose-lowering medication if cost is a major issue
Diabetologia
fatal MI, non-fatal stroke and cardiovascular death. The ARR
was 2.2% and the HR was 0.62 (95% CI 0.49, 0.77; p<0.001)
for cardiovascular death [51]. The ARR was 2.6% and the HR
was 0.68 (95% CI, 0.57, 0.82; p< 0.001) for death from any
cause.Canagliflozincomparedwithplacebowasstudiedinthe
Canagliflozin Cardiovascular Assessment Study (CANVAS)
Program (comprised of two similar trials, CANVAS and
CANVAS-Renal; n= 10,142) in participants with type 2 diabe-
tes, 66% of whom had a history of CVD. Participants were
followed for a median of 3.6 years. In the combined analysis
of the two trials, the primary composite endpoint of MI, stroke
or cardiovascular death was reduced with canagliflozin (26.9 vs
31.5 participants per patient-year with placebo; HR 0.86, 95%
CI 0.75, 0.97; p= 0.02) for superiority in the pooled analysis,
with consistent findings in the component studies. Though
there was a trend towards benefit for cardiovascular death, the
difference from placebo was not statistically significant in the
CANVAS Program [52]. For the SGLT2 inhibitors studied to
date, it appears that among patients with established CVD,there
is likely cardiovascular benefit, with the evidence of benefit
modestly stronger for empagliflozin than canagliflozin.
Adverse effects for the class are discussed in the section The
full range of therapeutic options: lifestyle management, medi-
cation and obesity management.
While the evidence of an ASCVD outcomes benefit for
GLP-1 receptor agonists and SGLT2 inhibitors has been dem-
onstrated for people with established ASCVD, the evidence of
benefit beyond glucose lowering has not been demonstrated in
those without ASCVD. Indeed, in subgroup analyses of these
trials, lower risk individuals have not been observed to have an
ASCVD benefit. While this may be due to the short time frame
of the studies and the low event rate in those without ASCVD,
the finding is consistent across the reported trials. Overall,
CVOTs of dipeptidyl peptidase-4 (DPP-4) inhibitors have dem-
onstrated safety, i.e. non-inferiority relative to placebo, for the
primary MACE endpoint, but not cardiovascular benefit.
The available evidence for cardiovascular event reduction
in patients with type 2 diabetes and clinical CVD is derived
from trials in which the participants were not meeting
glycaemic targets (HbA
1c
53 mmol/mol [7%] at baseline).
Furthermore, most (~70% across trials) participants were
treated with metformin at baseline. Thus, we recommend that
patients with clinical CVD not meeting individualised
glycaemic targets while treated with metformin (or in whom
metformin is contraindicated or not tolerated) should have an
SGLT2 inhibitor or GLP-1 receptor agonist with proven ben-
efit for cardiovascular risk reduction added to their treatment
programme. There are no clinical trial data that support pre-
scribing an SGLT2 inhibitor or GLP-1 receptor agonist with
theintentofreducingcardiovascularriskinpatientswithan
HbA
1c
<53 mmol/mol (<7%). Limited data suggest that there
is no heterogeneity in the cardiovascular benefits of SGLT2
inhibitors or GLP-1 receptor agonists as a function of
background glucose-lowering therapy. Thus, background
glucose-lowering therapy in patients with clinical CVD argu-
ably is not pertinent in clinical decision making. However,
dose adjustment or discontinuation of background medica-
tions may be required to avoid hypoglycaemia when adding
a new agent to a regimen containing insulin, sulfonylurea or
glinide therapy, particularly in patients at or near glycaemic
goals. Full efforts to achieve glycaemic and blood pressure
targets and to adhere to lipid, antiplatelet, antithrombotic and
tobacco cessation guidelines [7] should continue after an
SGLT2 inhibitor or GLP-1 receptor agonist is added, as such
efforts were integral to all studies that have demonstrated car-
diovascular benefit of these agents.
Patients with type 2 diabetes are at increased risk of HF
[53]. In the EMPA-REG OUTCOME and CANVAS CVOT
studies testing SGLT2 inhibitors, which enrolled participants
with ASCVD, >85% of participants did not have symptomatic
HF at baseline. Yet, in both trials there was a clinically and
statistically significant reduction in hospitalisation for HF for
the SGLT2 inhibitor as compared with placebo. In the EMPA-
REG OUTCOME study with empagliflozin [54], the ARR
was 1.4%, and the HR 0.65 (95% CI 0.50, 0.85) and in the
CANVAS Program with canagliflozin the HR was 0.67 (95%
CI 0.52, 0.87), with a rate of hospitalised HF of 5.5 vs 8.7
events per 1000 patient-years [55]. Because HF was neither
well characterised at baseline nor as carefully adjudicated as it
wouldhavebeeninatrialspecificallydesignedtoevaluateHF
outcomes, and because HF was a secondary endpoint in the
trials, further ongoing studies are required to conclusively ad-
dress the issue. That said, the significant reduction in
hospitalisation for HF demonstrated in the two study popula-
tions and the consistency across two independent trial
programmes suggest to us that treatment with SGLT2 inhibi-
tors in the setting of clinical HF may provide substantial ben-
efit and should be specifically considered in people with type
2 diabetes and ASCVD and HF.
In the GLP-1 receptor agonist studies LEADER,
SUSTAIN 6 and EXSCEL, there was no significant effect
on hospitalization for HF with HR 0.86 (95% CI 0.71, 1.06),
1.11 (95% CI 0.77, 1.61) and 0.94 (95% CI 0.78, 1.13), re-
spectively [4749]. Two short-term studies of liraglutide in
patients with reduced ejection fraction suggested a lack of
benefit in this setting [56,57].
Among the recent cardiovascular safety outcomes trials
testing DPP-4 inhibitors, the Saxagliptin Assessment of
Vascular Outcomes Recorded in Patients with Diabetes
Consensus recommendation
Among patients with ASCVD in whom HF coexists or is
of special concern, SGLT2 inhibitors are recommended
(Figs 2 and 3).
Diabetologia
Mellitus Thrombolysis in Myocardial Infarction 53
(SAVOR-TIMI 53) study evaluating saxagliptin demonstrated
a significant increased risk of HF, with 3.5% risk of
hospitalisation for HF vs 2.8% for placebo (HR 1.27; 95%
CI 1.07, 1.51; p= 0.007) [58]. In the subsequent
Examination of Cardiovascular Outcomes with Alogliptin versus
Standard of Care (EXAMINE) study of alogliptin there was no
statistically significant difference in HF hospitalisation (3.9% vs
3.3% with placebo) [59],andintheTrialEvaluating
Cardiovascular Outcomes with Sitagliptin (TECOS), the rate of
hospitalisation for HF was 3.1% in both sitagliptin- and placebo-
treated patients [60].
Patients with type 2 diabetes and kidney disease are at an
increased risk for cardiovascular events. A substantial number
of participants with an eGFR of 3060 ml min
1
[1.73 m]
2
were
included in EMPA-REG OUTCOME, CANVAS, LEADER
and SUSTAIN 6. An important finding in the studies was reduc-
tion of the primary ASCVD outcome even among participants
with stage 3 CKD (eGFR 3060 ml min
1
[1.73 m]
2
). For
SGLT2 inhibitors, this contrasts with the glucose-lowering effect,
which diminishes with declining eGFR.
In addition to the primary cardiovascular endpoints, most
of the SGLT2 inhibitor and GLP-1 receptor agonist CVOTs
reported benefit in renal endpoints, albeit as secondary out-
comes. The renal outcome benefit has been most pronounced
and consistent for SGLT2 inhibitors. EMPA-REG
OUTCOME (empagliflozin) demonstrated an ARR 6.1%,
HR of 0.61 (95% CI 0.53, 0.70) for the composite outcome
of new or worsening nephropathy (progression to urine
albumin/creatinine ratio >33.9 mg/mmol (>300 mg/g), dou-
bling of serum creatinine and ESRD, or death by ESRD). The
most prevalent outcome component was the development of
sustained albuminuria, but the other components were each
significantly reduced relative to placebo [61]. CANVAS
(canagliflozin) reported an HR of 1.7 (95% CI 1.51, 1.91)
for regression of albuminuria and a 40% reduction in risk in
the composite outcome of eGFR, ESRD or renal death (5.5 vs
9.0 participants per 1000 patient-years; HR 0.60; 95% CI
0.47, 0.77) [52]. Additional trials with primary renal endpoints
are ongoing in high-risk renal populations. The Canagliflozin
and Renal Endpoints in Diabetes with Established
Nephropathy Clinical Evaluation (CREDENCE) trial examin-
ing canagliflozin in CKD withproteinuria has been stopped at
a planned interim analysis for achieving the primary efficacy
endpoint [62].
In LEADER and SUSTAIN 6, the GLP-1 receptor agonist
liraglutide was associated with an ARR of 1.5% and an HR of
0.78 (95% CI 0.67, 0.92) for new or worsening nephropathy
[63], and semaglutide demonstrated an ARR of 2.3% and an
HR of 0.64 (95% CI 0.46, 0.88) for new or worsening ne-
phropathy [48]. Progression of albuminuria was the most
prevalent component of the composite renal endpoint, where-
as the other components (doubling of serum creatinine, ESRD
or renal death) did not contribute substantially to the benefit.
In the DPP-4 inhibitor CVOTs, the DPP-4 inhibitors have
been shown to be safe from a renal perspective, with modest
reduction in albuminuria [64].
The full range of therapeutic options: lifestyle
management, medication and obesity
management
This section summarises the lifestyle, medication and obe-
sity management therapies that lower glucose or improve
other outcomes in patients with type 2 diabetes. A more
comprehensive discussion of these issues is available else-
where [3,21,65]. For more details on weight loss medi-
cations and metabolic surgery, see the section Obesity
management beyond lifestyle intervention. Basic infor-
mation about specific options in each category of therapy
is summarised in Table 2.
Lifestyle interventions, including MNT and physical activ-
ity, are effective and safe for improving glucose control in type
2 diabetes. For these reasons, they are recommended as first-
line therapies from the time of diagnosis and as co-therapy for
patients who also require glucose-lowering medications or
metabolic surgery. Lifestyle management should be part of
the ongoing discussion with individuals with type 2 diabetes
at each visit.
Lifestyle management
Medical nutrition therapy
MNT comprises education and support to help patients
adopt healthy eating patterns. The goal of MNT is to
manage blood glucose and cardiovascular risk factors to
reduce risk for diabetes-related complications while pre-
serving the pleasure of eating [21]. Two basic dimen-
sions of MNT include dietary quality and energy restric-
tion. Strategies directed at each dimension can improve
glycaemic control.
Consensus recommendation
An individualised programme of MNT should be offered
to all patients.
Consensus recommendation
For patients with type 2 diabetes and CKD, with or
without CVD, consider the use of an SGLT2 inhibitor
shown to reduce CKD progression or, if contraindicated
or not preferred, a GLP-1 receptor agonist shown to
reduce CKD progression (Figs 2 and 3).
Diabetologia
Table 2 Glucose-lowering medications and therapies available in the USA or Europe and specific characteristics that may guide individualised treatment choices in non-pregnant adults with type 2
diabetes
Class Medications/therapies in
class
Primary physiological
action(s)
Advantages Disadvantages/adverse effects Efficacy
Lifestyle
Diet quality Mediterranean type
DASH
Low carbohydrate
Vegetarian
Others
Depends on diet Inexpensive
No side effects
Requires instruction
Requires motivation
Requires lifelong behavioural change
Social barriers may exist
Intermediate
Physical activity Running, walking
Bicycling (including
stationary)
Swimming
Resistance training
Yog a
Tai chi
Many others
Energy expenditure
Weight management
Insulin sensitivity
Inexpensive
Fall risk by increasing
balance/strength
? Improves mental health
Bone density
Blood pressure
Weight
Improves ASCVD risk factors
Risk of musculoskeletal injury
Requires motivation
Risk of foot trauma in patients with
neuropathy
Requires lifelong behavioural change
Intermediate
Energy restriction Individual energy
restriction with or without
energy tracking
Programmes with
counselling
Food substitution
programmes
Energy restriction
Weight management
Hepatic and pancreatic fat
Insulin sensitivity
Lowers glycaemia
Reduces need for diabetes and
other medications
No serious side effects
Improves ASCVD risk factors
Requires motivation
Requires lifelong behavioural change
Variable, with potential
for very high efficacy;
often intermediate
Oral medications
Biguanides Metformin Hepatic glucose production
Multiple other
non-insulin-mediated
mechanisms
Extensive experience
No hypoglycaemia
Inexpensive
GI symptoms
Vitamin B
12
deficiency
Use with caution or dose adjustment
for CKD stage 3B (eGFR
3044 ml min
1
[1.73 m]
2
)
Lactic acidosis (rare)
High
SGLT2 inhibitors Canagliflozin
Dapagliflozin
Empagliflozin
Ertugliflozin
Blocks glucose reabsorption
by the kidney, increasing
glucosuria
? Other tubulo-glomerular
effects
No hypoglycaemia
Weight
Blood pressure
Effective at all stages of T2DM
with preserved glomerular function
MACE, HF, CKD with some
agents (see text)
Genital infections
UTI
Polyuria
Volume depletion/
hypotension/dizziness
LDL-C
Creatinine (transient)
Dose adjustment/avoidance for renal
disease
Risk for amputation (canagliflozin)
Risk for fracture (canagliflozin)
Risk for DKA (rare)
Fourniers gangrene (rare)
Expensive
Intermediatehigh
(dependent on GFR)
Diabetologia
Tab l e 2 (continued)
Class Medications/therapies in
class
Primary physiological
action(s)
Advantages Disadvantages/adverse effects Efficacy
DPP-4 inhibitors Sitagliptin
Vildagliptin
a
Saxagliptin
Linagliptin
Alogliptin
Glucose dependent:
Insulin secretion
Glucagon secretion
No hypoglycaemia
Weight neutral
Well tolerated
Rare urticaria/angioedema
HF hospitalisation (saxagliptin)
Dose adjustment/avoidance for renal
disease depending on agent
? Pancreatitis
? Arthralgia
? Bullous pemphigoid
Expensive (USA); variable in Europe
Intermediate
Sulfonylureas Glibenclamide/glyburide
Glipizide
Gliclazide
a
Glimepiride
Insulin secretion Extensive experience
Microvascular risk (UKPDS)
Inexpensive
Hypoglycaemia
Weight
Uncertain cardiovascular safety
Dose adjustment/avoidance for renal
disease
High rate of secondary failure
High
TZDs Pioglitazone
Rosiglitazone
b
Insulin sensitivity Low risk for hypoglycaemia
Durability
HDL-C
Triacylglycerols (pioglitazone)
ASCVD events (pioglitazone: in
a post-stroke insulin-resistant
population and as secondary
endpoint in a high-CVD-risk
diabetes population)
Lower cost
Weight
Oedema/heart failure
Bone loss
Bone fractures
LDL-C (rosiglitazone)
? Bladder cancer
? Macular oedema
High
Meglitinides (Glinides) Repaglinide
Nateglinide
Insulin secretion Postprandial glucose excursions
Dosing flexibility
Safe in advanced renal disease with
cautious dosing (especially
repaglinide)
Lower cost
Hypoglycaemia
Weight
Uncertain cardiovascular safety
Frequent dosing schedule
Intermediatehigh
α-Glucosidase inhibitors Acarbose
Miglitol
Slows carbohydrate
digestion/absorption
Low risk for hypoglycaemia
Postprandial glucose excursions
Non-systemic mechanism of action
Cardiovascular safety
Lower cost
Frequent GI side effects
Frequent dosing schedule
Dose adjustment/avoidance for renal
disease
Lowintermediate
Bile acid sequestrants Colesevelam
b
?Hepatic glucose
production
?Incretin levels
No hypoglycaemia
LDL-C
Constipation
Triacylglycerols
May absorption of other medications
Intermediate expense
Lowintermediate
Dopamine-2 agonists Quick-release
bromocriptine
b
Modulates hypothalamic
regulation of metabolism
Insulin sensitivity
No hypoglycaemia
?ASCVD events
Headache/dizziness/syncope
Nausea
Fatigue
Rhinitis
High cost
Lowintermediate
Diabetologia
Tab l e 2 (continued)
Class Medications/therapies in
class
Primary physiological
action(s)
Advantages Disadvantages/adverse effects Efficacy
Injectable medications
Insulins
Long acting (basal) Degludec (U100, U200)
Detemir
Glargine (U100, U300)
Activates insulin receptor
Glucose disposal
Glucose production
Nearly universal response
Theoretically unlimited efficacy
Once daily injection
Hypoglycaemia
Weight gain
Training requirements
Frequent dose adjustment for optimal
efficacy
High cost
Very high
Intermediate acting
(basal)
Human NPH Activates insulin receptor
Glucose disposal
Glucose production
Nearly universal response
Theoretically unlimited efficacy
Less expensive than analogues
Hypoglycaemia
Weight gain
Training requirements
Often given twice daily
Frequent dose adjustment for optimal
efficacy
Very high
Rapid acting Aspart (conventional and
fast-acting)
Lispro (U100, U200)
Glulisine
Activates insulin receptor
Glucose disposal
Glucose production
Nearly universal response
Theoretically unlimited efficacy
Postprandial glucose
Hypoglycaemia
Weight gain
Training requirements
May require multiple daily injections
Frequent dose adjustment for optimal
efficacy
High cost
Very high
Inhaled rapid acting Human insulin inhalation
powder
b
Activates insulin receptor
Glucose disposal
Glucose production
Nearly universal response
Postprandial glucose
More rapid onset and shorter
duration than rapid-acting
analogues
Spirometry (FEV
1
) required before
initiating, after 6 months and annually
Contraindicated in chronic lung
disease
Not recommended in smokers
Hypoglycaemia
Weight gain
Training requirements
May require multiple inhalations daily
Frequent dose adjustment for optimal
efficacy; limited options in dosing
interval
High cost
Respiratory side effects (e.g.
bronchospasm, cough, decline in
FEV
1
)
High
Short acting Human regular (U100,
U500)
Activates insulin receptor
Glucose disposal
Glucose production
Nearly universal response
Theoretically unlimited efficacy
Postprandial glucose
Less expensive than analogues
Hypoglycaemia
Weight gain
Training requirements
Frequent dose adjustment for optimal
efficacy
May require multiple daily injections
Very high
Diabetologia
Tab l e 2 (continued)
Class Medications/therapies in
class
Primary physiological
action(s)
Advantages Disadvantages/adverse effects Efficacy
Premixed Many Activates insulin receptor
Glucose disposal
Glucose production
Nearly universal response
Theoretically unlimited efficacy
Fewer injections than basal/bolus
before every meal
Recombinant human analogues are
less expensive
Hypoglycaemia
Weight gain
Training requirements
Frequent dose adjustment for optimal
efficacy
High cost (except human insulin
premix)
Canleadtoobligateeating
Very high
GLP-1 RA
Shorter acting Exenatide
Lixisenatide
Glucose dependent:
Insulin secretion
Glucagon secretion
Slows gastric emptying
Satiety
No hypoglycaemia as
monotherapy
Weight
Excellent postprandial glucose
efficacy for meals after injections
Improves cardiovascular risk
factors
Frequent GI side effects that may be
transient
Modestly heart rate
Training requirements
Dose adjustment/avoidance in renal
disease
Acute pancreatitis (rare/uncertain)
Very high c o s t
Intermediatehigh
Longer acting Dulaglutide
Exenatide
extended-release
Liraglutide
Semaglutide
Glucose dependent:
Insulin secretion
Glucagon secretion
Satiety
No hypoglycaemia as
monotherapy
Weight
Postprandial glucose excursions
Improves cardiovascular risk
factors
MACE with some agents (see
text)
Albuminuria with some agents
(see text)
Greater lowering of fasting glucose
vs short-acting preparations
Once weekly dosing (except
liraglutide, which is daily)
GI side effects, including gall bladder
disease
Greater heart rate
Training requirements
Dose adjustment/avoidance for some
agents in renal disease
Acute pancreatitis (rare/uncertain)
C cell hyperplasia/medullary thyroid
tumours (rare/uncertain; observed in
animals only)
Very high c o s t
Highvery high
Other injectables
Amylin mimetics Pramlintide
b
Glucagon secretion
Slows gastric emptying
Satiety
Postprandial glucose excursions
Weight
Hypoglycaemia
Frequent dosing schedule
Training requirements
Frequent GI side effects
Very high c o s t
Intermediate
Fixed-dose
combination of GLP-1
RA and basal insulin
analogues
Liraglutide/degludec
Lixisenatide/glargine
Combined activities of
components
Enhanced glycaemic efficacy vs
components
Reduced adverse effects (e.g. GI,
hypoglycaemia) vs components
Less weight loss than GLP-1 receptor
agonist alone
Very high c o s t
Very high
Diabetologia
Tab l e 2 (continued)
Class Medications/therapies in
class
Primary physiological
action(s)
Advantages Disadvantages/adverse effects Efficacy
Weight loss medications Lorcaserin
b
Naltrexone/bupropion
Orlistat
Phentermine/topiramate
b
Liraglutide 3 mg
Reduced appetite
Fat malabsorption (orlistat)
Mean 39 kg weight loss vs
placebo
High discontinuation rates from side
effects
<50% achieve 5% weight loss
Drug-specific side effects
Limited durability
High cost
Intermediate
Metabolic surgery VSG
RYGB
Adjustable gastric band
BPD
Restriction of food intake
(all)
Malabsorption (RYGB,
BPD)
Changes in hormonal and
possibly neuronal signalling
(VSG, RYGB, BPD)
Sustained weight reduction
Rate of remission of diabetes
Number of diabetes drugs
Blood pressure
Improved lipid metabolism
High initial cost
Risk for early and late surgical
complications
Risk for reoperation
Risk for dumping syndrome
Nutrient and vitamin malabsorption
Risk for new-onset depression
Risk for new-onset opioid use
Risk for gastroduodenal ulcer
Risk for hypoglycaemia
Risk for alcohol use disorder
Very high
More details available in ADAsStandards of Medical Care in Diabetes2018[3]
Glucose-lowering efficacy of drugs by change in HbA
1c
: >22 mmol/mol (2%) very high, 1122 mmol/mol (12%) high, 611 mmol/mol (0.51.5%) intermediate, <6 mmol/mol (0.5%) low
a
Not licensed in the USA for type 2 diabetes
b
Not licensed in Europe for type 2 diabetes
BPD, biliopancreatic diversion; DASH, Dietary Approaches to Stop Hypertension; DKA, diabetic ketoacidosis; FEV
1
, forced expiratory volume in 1 s on pulmonary function testing; GI, gastrointestinal;
HDL-C, HDL-cholesterol; LDL-C, LDL-cholesterol; RYGB, Roux-en-Y gastric bypass; VSG, vertical sleeve gastroplasty; UTI, urinary tract infection
Diabetologia
Dietary quality and eating patterns There is no single ratio of
carbohydrate, proteins and fat intake that is optimal for every
person with type 2 diabetes. Instead, there are many good
options and professional guidelines usually recommend indi-
vidually selected eating patterns that emphasise foods of dem-
onstrated health benefit, that minimise foods of demonstrated
harm and that accommodate patient preference and metabolic
needs, with the goal of identifying healthy dietary habits that
are feasible and sustainable. Three trials of a Mediterranean
eating pattern reported modest weight loss and improved
glycaemic control [6668]. In one of these, people with
new-onset diabetes assigned to a low-carbohydrate
Mediterranean eating pattern were 37% less likely to require
glucose-lowering medications over 4 years compared with
patients assigned to a low-fat diet (HR 0.63; 95% CI 0.51,
0.86). A meta-analysis of RCTs in patients with type 2 diabe-
tes showed that the Mediterranean eating pattern reduced
HbA
1c
more than control diets (mean difference 3.3 mmol/
mol, 95% CI 5.1, 1.5 mmol/mol [0.30%, 95% CI 0.46%,
0.14%]) [69]. Low-carbohydrate, low-glycaemic index and
high-protein diets, and the Dietary Approaches to Stop
Hypertension (DASH) diet all improve glycaemic control,
but the effect of the Mediterranean eating pattern appears to
be the greatest [7072]. Low-carbohydrate diets (<26% of
total energy) produce substantial reductions in HbA
1c
at
3 months (5.2 mmol/mol, 95% CI 7.8, 2.5 mmol/mol
[0.47%, 95% CI 0.71%, 0.23%]) and 6 months
(4.0 mmol/mol, 95% CI 6.8, 1.0 mmol/mol [0.36%,
95% CI 0.62%, 0.09%]), with diminishing effects at 12
and 24 months; no benefit of moderate carbohydrate restric-
tion (2645%) was observed [73]. Vegetarian eating patterns
have been shown to lower HbA
1c
, but not fasting glucose,
compared with non-vegetarian ones [74]. Very recent trials
of different eating patterns in type 2 diabetes have typically
also included weight reduction, hindering firm conclusions
regarding the distinct contribution of dietary quality.
Non-surgical energy restriction for weight loss If a patient
wishes to aim for remission of type 2 diabetes, particularly
within 6 years of diagnosis, evidence-based weight manage-
ment programmes are often successful.
The most effective non-surgical strategies for weight reduc-
tion involve food substitution and intensive, sustained counsel-
ling (e.g. 1226 individual counselling sessions over 6
12 months). Among adults with type 2 diabetes, meal replace-
ment (825853 kcal/day [34503570 kj/day] formula-diet for
35 months) followed by gradual reintroduction of food and
intensive counselling resulted in 9 kg placebo-adjusted weight
loss at 1 year and high rates of diabetes remission (46% vs 4%;
OR 19.7; 95% CI 7.8, 49.8) compared with best usual practice
[75]. In terms of intensive behavioural interventions, the Action
for Health in Diabetes (Look AHEAD) trial [76]randomised
5145 overweight or obese patients with type 2 diabetes to an
intensive lifestyle programme that promoted energy restriction,
incorporating meal replacements to induce and sustain weight
loss, along with increased physical activity compared with stan-
dard diabetes education and support in the control group. After
9.6 years, weight loss was greater in the intervention group
(8.6%vs0.7%at1year;6.0%vs3.5%atstudyend;bothp
<0.05). HbA
1c
also fell in the intervention group despite less
use of glucose-lowering medications. Cardiovascular event
rates were not reduced but there were numerous other benefits.
In a 12 month trial, 563 adults with type 2 diabetes who were
randomised to Weight Watchers compared with standard care
had a 2.1% net weight loss (4.0% vs 1.9%; p< 0.001), a
5.3 mmol/mol (3.5 vs +1.8 mmol/mol; p= 0.020) net absolute
improvement in HbA
1c
(0.48% [0.32% vs +0.16%]), and a
greater reduction in use of glucose-lowering medications
(26% vs +12%; p<0.001) [77]. Similar programmes have
resulted in a net 3 kg weight loss over 1218 months [7880].
Physical activity
Aerobic exercise, resistance training, and the combination of
the two are effective in reducing HbA
1c
by about 6.6 mmol/
mol (0.6%) [8184]. Of these modalities, some evidence
suggests that aerobic exercise and the combination of aerobic
exercise and resistance training may be more effective than
resistance training alone [85], but this remains controversial.
When considering exercise interventions, special consider-
ations are required for individuals with CVD, uncontrolled
retinopathy or nephropathy and severe neuropathy. A wide
range of physical activity, including leisure time activities
(e.g. walking, swimming, gardening, jogging, tai chi and
yoga) can significantly reduce HbA
1c
[8690]. In general,
supervision of exercise and motivational strategies, such as
monitoring using a step counter, can improve the effect of
exercise on HbA
1c
compared with advice alone [84,91].
The combination of dietary change for weight reduction and
physical exercise improves hyperglycaemia and reduces
cardiovascular risk factors more than dietary interventions or
physical activity alone [92].
Consensus recommendation
All overweight and obese patients with diabetes should
be advised of the health benefits of weight loss and
encouraged to engage in a programme of intensive
lifestyle management, which may include food
substitution.
Consensus recommendation
Increasing physical activity improves glycaemic control
and should be encouraged in all people with type 2
diabetes.
Diabetologia
Medications for lowering glucose
Metformin
Metformin is an oral medication that reduces plasma glucose
via multiple mechanisms. It is available as an immediate-
release formulation that is typically administered twice a day
and as extended-release formulations for once-daily or twice-
daily administration. The formulations are equally effective
with no consistent differences in side effect profile [93].
Dosages of immediate-release metformin start at 500 mg once
or twice a day with meals, andshould be increased as tolerated
to a target dosage of 1000 mg twice a day. The maximum daily
dose is 2550 mg in the USA and 3000 mg in the European
Union, though doses above 2000 mg are generally associated
with little additional efficacy and poorer tolerability [94].
Gastrointestinal symptoms are common and dose-dependent,
and may improve over time or with dose reduction. Metformin
should not be used in patients with an eGFR <30 ml min
1
[1.73 m]
2
and dose reduction should be considered when the
eGFR is <45 ml min
1
[1.73 m]
2
[9597]. Caution should be
taken when conditions are present that may reduce eGFR.
Advantages of metformin include its high efficacy, low cost,
minimal hypoglycaemia risk when used as monotherapy, and
the potential for some weight loss. Some studies have sug-
gested a benefit for preventing CVD [98], but this has not
been supported by the results of a recent meta-analysis [99].
However, metformin may lower risk for cardiovascular mor-
tality compared with sulfonylurea therapy [100]. Rare cases of
lactic acidosis have been reported, usually in the setting of
severe illness or acute kidney injury. Therefore, metformin
should be omitted in the setting of severe illness, vomiting
or dehydration. Metformin may result in lower serum vitamin
B
12
concentration; therefore, periodic monitoring and supple-
mentation is generally recommended if levels are deficient,
particularly in those with anaemia or neuropathy [101].
Because of its high efficacy in lowering HbA
1c
,goodsafety
profile and low cost, metformin remains the first-line medica-
tion for management of type 2 diabetes.
SGLT2 inhibitors
SGLT2 inhibitors are oral medications that reduce plasma glu-
cose by enhancing urinary excretion of glucose [102]. The
glucose-lowering efficacy of these medications is dependent
on renal function. Initiation and continuation of SGLT2 inhib-
itors are restricted by eGFR and require intermittent monitor-
ing of renal function (refer to European Medicines Agency
and US Food and Drug Administration prescribing informa-
tion for current recommendations). These medications are of
high efficacy in lowering glucose in the setting of normal renal
function [51,52,103]. All SGLT2 inhibitors are associated
with a reduction in weight and blood pressure. Alone or with
metformin, they do not increase the risk for hypoglycaemia.
Empagliflozin and canagliflozin have cardiac and renal bene-
fits in patients with established or at high risk of ASCVD.
Cardiac and renal benefits have been demonstrated down to
an eGFR of 30 ml min
1
[1.73 m]
2
, though currently none of
the SGLT2 inhibitors have been approved for use by regula-
tors at an eGFR below 45 ml min
1
[1.73 m]
2
(see the section
Recommended process for glucose-lowering medication se-
lection: where does new evidence from cardiovascular out-
comes trials fit in?)[51,52,61]. The class is associated with
increased risk for mycotic genital infections (mostly vaginitis
in women, balanitis in men) [51,52,104,105]. Case reports of
diabetic ketoacidosis with SGLT2 inhibitors in type 2 diabetes
continue to raise concern, though increased rates have not
been confirmed in large trials [102,106]. Therefore, the
SGLT2 inhibitors should be used with caution and appropriate
patient education should be provided for those with insulin
deficiency. SGLT2 inhibitors have been associated with an
increased risk of acute kidney injury, dehydration and ortho-
static hypotension; caution should be taken when SGLT2 in-
hibitors are used in combination with diuretics and/or ACE
inhibitors and angiotensin receptor blockers. Canagliflozin
has been associated with increased risk for lower limb ampu-
tation (6.3 canagliflozin vs 3.4 per 1000 patient-years with
placebo after 3.1 years; HR 1.97; 95% CI 1.41, 2.75;) [52].
Similarly, fracture risk has been reported with canagliflozin
(15.4 vs 11.9 participants with fracture per 1000 patient-
years; HR 1.26; 95% CI 1.04, 1.52) [52]. It is uncertain wheth-
er amputation and fractures are class effects.
GLP-1 receptor agonists
GLP-1 receptor agonists are currently delivered by subcutane-
ous injection. These medications stimulate insulin secretion
and reduce glucagon secretion in a glucose-dependent manner,
improve satiety and promote weight loss [107,108]. Structural
differences among GLP-1 receptor agonists affect duration of
action, and their formulation and dosing may affect efficacy
for glucose-lowering and weight reduction as well as side
effect profile and cardiovascular effects [109]. Dulaglutide,
exenatide extended-release and semaglutide are administered
once weekly [108,109]. Liraglutide and lixisenatide are ad-
ministered once daily, and exenatide is available in a twice-
daily formulation. GLP-1 receptor agonists have high glucose-
lowering efficacy, but with variation within the drug class
[110,111]. Evidence suggests that the effect may be greatest
for semaglutide once weekly, followed by dulaglutide and
liraglutide, closely followed by exenatide once weekly, and
then exenatide twice daily and lixisenatide [110,112116].
The short-acting medications exenatide twice daily and
lixisenatide have greater postprandial effects, at least after
the meals with which they are administered. All GLP-1 recep-
tor agonists reduce weight [110]; the reduction ranges from
Diabetologia
about 1.5 kg to 6.0 kg over about 30 weeks of therapy [110,
117]. Liraglutide and semaglutide have been shown to im-
prove cardiovascular outcomes [47,48] (see the section
Recommended process for glucose-lowering medication se-
lection: where does new evidence from cardiovascular out-
comes trials fit in?). The most common side effects of GLP-
1 receptor agonists are nausea, vomiting and diarrhoea, though
these tend to diminish over time. GLP-1 receptor agonists
have minimal risk for hypoglycaemia, but may increase the
hypoglycaemic potential of insulin and sulfonylureas when
combined with those medications [118]. Contrary to early sig-
nals, GLP-1 receptor agonists do not seem to substantially
increase risk for pancreatitis, pancreatic cancer or bone disease
[119]. They are associated with increased risk of gallbladder
events [120]. Semaglutide was associated with increased reti-
nopathy complications in the SUSTAIN 6 trial (HR 1.76, 95%
CI 1.11, 2.78), largely among those with baseline retinopathy
who had rapid improvement of glycaemic control [48]. While
this observation remains unexplained, this is also a recognised
effect of intensification of glycaemic control with insulin.
DPP-4 inhibitors
DPP-4 inhibitors are oral medications that increase insulin se-
cretion and reduce glucagon secretion in a glucose-dependent
manner. They have moderate glucose-lowering efficacy [121,
122]. DPP-4 inhibitors are well tolerated, have a neutral effect
on weight and have minimal risk of hypoglycaemia when used
as monotherapy [123]. When added to sulfonylurea therapy,
however, the risk for hypoglycaemia is increased 50% com-
pared with sulfonylurea therapy alone [124]. The recommend-
ed dose foreach DPP-4 inhibitor is determined and needs to be
adjusted based on renal function; linagliptin is the exception as
it has minimal renal excretion. Rare but increased rates of
pancreatitis [125] and musculoskeletal side effects have been
reported [126]. CVOTs demonstrated the cardiovascular safety
but no cardiovascular benefit of three DPP-4 inhibitors
(saxagliptin, alogliptin and sitagliptin) as well as imbalances
regarding HF for saxagliptin and alogliptin [127,128](seethe
section Recommended process for glucose-lowering medica-
tion selection: where does new evidence from cardiovascular
outcomes trials fit in?.
Thiazolidinediones
Thiazolidinediones (TZDs) (pioglitazone and rosiglitazone)
are oral medications that increase insulin sensitivity and are
of high glucose-lowering efficacy [129131]. TZDs increase
HDL-cholesterol [132,133], and pioglitazone has been shown
to reduce cardiovascular endpoints [132,134138] and hepat-
ic steatohepatitis [139], but without conclusive evidence for
benefit. TZDs are associated with the best evidence among
glucose-lowering medications for glycaemic durability
[140]. However, these notable benefits must be balanced with
safety concerns regarding fluid retention and congestive heart
failure [136,140,141], weight gain [132,136,140142],
bone fracture [143,144] and, possibly, bladder cancer [145].
Lower dose therapy (e.g. pioglitazone 1530 mg) mitigates
weight gain and oedema, but the broader benefits and harms
of low-dose TZD therapy have not been evaluated.
Sulfonylureas
Sulfonylureas are oral medications that lower glucose by stim-
ulating insulin secretion from pancreatic beta cells. They are
inexpensive, widely available, and have high glucose-
lowering efficacy [146]. Sulfonylureas were used as part of
the glucose-lowering regimen in the UK Prospective Diabetes
Study (UKPDS) [147] and Action in Diabetes and Vascular
Disease: Preterax and Diamicron MR Controlled Evaluation
(ADVANCE) [148] trials, which both demonstrated reduc-
tions in microvascular complications. Sulfonylureas are asso-
ciated with weight gain and risk for hypoglycaemia and down
titration of dose to reduce the risk of hypoglycaemia results in
higher HbA
1c
[146,149,150]. Sulfonylureas are known to be
associated with a lack of durable effect on glucose lowering
[144,151].Theweightgainassociatedwithsulfonylureasis
relatively modest in large cohort studies and the incidence of
severe hypoglycaemia is lower than with insulin [152].
Important differences among sulfonylureas affect both safety
and efficacy. Glibenclamide (known as glyburide in the USA
and Canada) has a higher risk of hypoglycaemia compared
with other sulfonylureas [153]. Glipizide, glimepiride and
gliclazide may have a lower risk for hypoglycaemia compared
with other sulfonylureas [152,154]. Adverse cardiovascular
outcomes with sulfonylureas in some observational studies
have raised concerns, although findings from recent system-
atic reviews have found no increase in all-cause mortality
compared with other active treatments [152]. As newer-
generation sulfonylureas appear to confer a lower risk of
hypoglycaemia and have favourable cost, efficacy and safety
profiles, sulfonylureas remain a reasonable choice among
glucose-lowering medications, particularly when cost is an
important consideration. Patient education and use of low or
variable dosing with later-generation sulfonylureas may be
used to mitigate the risk of hypoglycaemia. Greatest caution
in this regard is warranted for people at high risk of
hypoglycaemia, such as older patients and those with CKD.
Insulin
Numerous formulations of insulin are available with differing
durations of action. Humaninsulins (NPH, regular [R], and
premixed combinations of NPH and R) are recombinant
DNA-derived human insulin, while insulin analogues have
been designed to change the onset or duration of action. The
Diabetologia
main advantage of insulin over other glucose-lowering medi-
cations is that insulin lowers glucose in a dose-dependent
manner over a wide range, to almost any glycaemic target as
limited by hypoglycaemia. Older formulations of insulin have
also demonstrated reduction in microvascular complications
and with long-term follow-up, all-cause mortality and
diabetes-related death [147,155]. Beyond hypoglycaemia,
the disadvantages of insulin include weight gain and the need
for injection, frequent titration for optimal efficacy and glu-
cose monitoring [156].
The effectiveness of insulin is highly dependent on its ap-
propriate use; patient selection and training; adjustment of
dose for changes in diet, activity or weight; and titration to
acceptable, safe glucose targets. Formulations of intermediate-
and long-acting insulin have different timings of onset, dura-
tions of action and risks of hypoglycaemia. However, the way
in which insulin is administered, including the dose, timing of
injection and glycaemic targets, has a greater impact on the
adverse effects of insulin than differences among insulin
formulations.
Basal insulin Basal insulin refers to longer-acting insulin that
is meant to cover the bodys basal metabolic insulin
requirement (regulating hepatic glucose production), in
contrast to bolus or prandial insulin, which is meant to reduce
glycaemic excursions after meals. Basal insulin is the
preferred initial insulin formulation in patients with type 2
diabetes. Options include once- or twice-daily administration
of intermediate-acting NPH or detemir insulin and the once-
daily administration of glargine (U100 or U300) or degludec
(U100 or U200). Long-acting insulin analogues (degludec
[U100 or U200], glargine [U100 and U300], detemir) have a
modestly lower absolute risk for hypoglycaemia compared
with NPH insulin, but cost more [157160]. However, in
real-world settings where patients are treated to conventional
treatment targets, initiation of NPH compared with determir or
glargine U100 did not increase hypoglycaemia-related
emergency department visits or hospital admissions [161].
When comparing human and analogue insulins, cost
differences can be large while differences in hypoglycaemia
risk are modest and differences in glycaemic efficacy
minimal.
Degludec is associated with a lower risk of severe
hypoglycaemia compared with glargine U100 insulin when
targeting intensive glycaemic control in patients with long-
standing type 2 diabetes at high risk of CVD; absolute inci-
dence difference of 1.7% over 2 years (rate ratio 0.60; p<
0.001 for superiority; OR 0.73; p< 0.001 for superiority)
[162]. Biosimilar formulations are now available for glargine
with similar efficacy profile and lower cost [163]. No insulin
has been shown to reduce risk for CVD [156], but data suggest
that glargine U100 and degludec do not increase risk for
MACE [162,164].
Concentrated formulationsof degludec (U200) and glargine
(U300) are available that allow injection of a reduced volume,
a convenience for patients on higher doses. Glargine U300 is
associated with a lower risk of nocturnal hypoglycaemia com-
pared with glargine U100 but requires a 1014% higher dose
of glargine for equivalent efficacy [165167].
Not all patients have their blood glucose adequately con-
trolled with basal insulin. In particular, patients with higher
pre-treatment HbA
1c
, higher BMI, longer duration of disease,
and a greater number of oral glucose-lowering medications are
more likely to require intensified therapy [168].
Other insulin formulations Short- and rapid-acting insulin
formulations administered at mealtime are generally used
to intensify basal insulin therapy in patients not meeting
glycaemic targets. Options include human regular insulin,
various analogues (aspart, glulisine and lispro), formula-
tions (faster insulin aspart, lispro U200), biosimilars
(lispro), and insulins with different routes of administration
(inhaled). Rapid-acting insulin analogues have a modestly
lower risk for hypoglycaemia compared with human regular
insulin but at a higher cost. Various premixed formulations
of human and analogue insulins are available and continue
to be widely used in some regions, though they tend to have
an increased risk of hypoglycemia as compared with basal
insulin alone (Table 2and Fig. 7).
Other glucose-lowering medications
Other oral glucose-lowering medications (i.e. meglitinides,
α-glucosidase inhibitors, colesevelam, quick-release bro-
mocriptine, pramlintide) are not used commonly in the
USA and some are not licensed at all in Europe. No major
new scientific information on these medications has
emerged in recent years. Their basic characteristics are
listed in Table 2.
Obesity management beyond lifestyle intervention
Medications for weight loss
Several clinical practice guidelines recommend weight loss
medications as an optional adjunct to intensive lifestyle man-
agement for patients with obesity, particularly if they have dia-
betes [169171]. Others do not [172]. Several medications and
medication combinations approved in the USA or Europe for
weight loss have been found to improve glucose control in
people with diabetes [173,174]. One glucose-lowering medica-
tion, liraglutide, is also approved for the treatment of obesity at a
higher dose [175]. Cost, side effects and modest efficacy limit
the role of pharmacotherapy in long-term weight management.
Diabetologia
Fig. 7 Intensifying to injectable therapies
Diabetologia
Metabolic surgery
Metabolic surgery is highly effective in improving glucose con-
trol [176178] and often produces disease remission [179182].
The effects can be sustained for at least 5 years [177,182].
Benefits include a reduction in the number of glucose-lowering
medications needed to achieve glycaemic targets [178,179].
Several clinical practice guidelines and position statements
recommend consideration of metabolic surgery as a treatment
option for adults with type 2 diabetes and (1) a BMI 40.0 kg/
m
2
(BMI 37.5 kg/m
2
in people of Asian ancestry) or (2) a
BMI of 35.039.9 kg/m
2
(32.537.4 kg/m
2
in people of Asian
ancestry) who do not achieve durable weight loss and im-
provement in comorbidities with reasonable non-surgical
methods [65,183]. Because baseline BMI does not predict
surgical benefits on glycaemia or hard outcomes and the im-
provement in glyacemic control occurs early through weight-
independent mechanisms [183], metabolic surgery may be
considered for those with a BMI of 30.034.9 kg/m
2
(27.5
32.4 in people of Asian ancestry) who do not achieve durable
weight loss and improvement in comorbidities with reason-
able non-surgical methods.
Adverse effects of bariatric surgery which vary by proce-
dure include surgical complications (e.g. anastomotic or staple
line leaks, gastrointestinal bleeding,intestinal obstruction, the
need for re-operation), late metabolic complications (e.g. pro-
tein malnutrition, mineral deficiency, vitamin deficiency,
anaemia, hypoglycaemia) and gastroesophageal reflux [184,
185]. Patients who undergo metabolic surgery may be at risk
for substance use, including drug and alcohol use and cigarette
smoking [186]. People with diabetes presenting for metabolic
surgery also have increased rates of depression and other ma-
jor psychiatric disorders [187]. These factors should be
assessed pre-operatively and during follow-up. Metabolic sur-
gery should be performed in high-volume centres with multi-
disciplinary teams that are experienced in the management of
diabetes and gastrointestinal surgery. Long-term lifestyle sup-
port and routine monitoring of micronutrient and nutritional
status must be provided to patients after surgery [188,189].
Putting it all together: strategies for implementation
For an increasing number of patients, presence of specific co-
morbidities (e.g. ASCVD, HF, CKD, obesity), safety concerns
(e.g. risk of hypoglycaemia) or healthcare environment (e.g.
cost of medications) mandate a specific approach to the choice
of glucose-lowering medication. These are considered in Figs
2,3,4,5,6. For patients not reaching their target HbA
1c
,itis
important to re-emphasise lifestyle measures, assess adherence
and arrange timely follow-up (e.g. within 36 months) (Fig. 1).
Initial monotherapy
Metformin remains the preferred option for initiating glucose-
lowering medication in type 2 diabetes and should be added to
lifestyle measures in newly diagnosed patients. This recom-
mendation is based on the efficacy, safety, tolerability, low
cost and extensive clinical experience with this medication.
Results from a substudy of the UKPDS (n= 342) showed
benefits of initial treatment with metformin on clinical out-
comes related to diabetes, with less hypoglycaemia and
weight gain than with insulin or sulfonylureas [98].
Initial combination therapy compared with stepwise
addition of glucose-lowering medication
In most patients, type 2 diabetes is a progressive disease, a
consequence generally attributed to a steady decline of insulin
secretory capacity. The practical impact of gradual loss of beta
cell function is that achieving a glycaemic target with mono-
therapy is typically limited to several years. Stepwise therapy
(i.e. adding medications to metformin to maintain HbA
1c
at
target) is supported by clinical trials [3]. While there is some
support for initial combination therapy due to the greater initial
reduction of HbA
1c
than can be provided by metformin alone
[190,191], there is little evidence that this approach is superior
to sequential addition of medications for maintaining
glycaemic control, or slowing the progression of diabetes.
However, since the absolute effectiveness of most oral medi-
cations rarely exceeds an 11 mmol/mol (1%) reduction in
HbA
1c
, initial combination therapy may be considered in pa-
tients presenting with HbA
1c
levels more than 17 mmol/mol
(1.5%) above their target. Fixed-dose formulations can im-
prove medication adherence when combination therapy is used
[192], and may help achieve glycaemic targets more rapidly
[100]. Potential benefits of combination therapy need to be
weighed against the exposure of patients to multiple
Consensus recommendation
Metformin is the preferred initial glucose-lowering
medication for most people with type 2 diabetes.
Consensus recommendation
The stepwise addition of glucose-lowering medication is
generally preferred to initial combination therapy.
Consensus recommendation
Metabolic surgery is a recommended treatment option
for adults with type 2 diabetes and (1) a BMI 40.0
kg/m2 (BMI 37.5 kg/m2 in people of Asian ancestry) or
(2) a BMI of 35.0–39.9 kg/m2 (32.5–37.4 kg/m2 in people
of Asian ancestry) who do not achieve durable weight
loss and improvement in comorbidities with reasonable
non-surgical methods.
Diabetologia
medications and potential side effects, increased cost and, in
the case of fixed combination medications, less flexibility in
dosing.
Choice of glucose-lowering medication
after metformin
As detailed in the Medications for lowering glucosesection,
the glucose-lowering medications that can be added to met-
formin have distinct profiles of action, efficacy and adverse
effects [100,193]. The early introduction of basal insulin is
well established, in particular when HbA
1c
levels are very
high (>97 mmol/mol [>11%]), symptoms of hyperglycaemia
are present or there is evidence of ongoing catabolism (e.g.
weight loss). This constellation of symptoms can occur in type
2 diabetes but suggest insulin deficiency and raise the possi-
bility of autoimmune (type 1) or pancreatogenic diabetes in
which insulin would be the preferred therapy. While this re-
mains the usual strategy for patients when HbA
1c
levels are
very high, SGLT2 inhibitors [194] and GLP-1 receptor ago-
nists [195] have demonstrated efficacy in patients with HbA
1c
levels exceeding 75 mmol/mol (9%), with the additional ben-
efits of weight reduction and reduced risk of hypoglycaemia.
Evidence from clinical trials supports the use of several of
the SGLT2 inhibitors and GLP-1 receptor agonists as add-on
therapy for people with type 2 diabetes with an HbA
1c
>53 mmol/mol (>7%) and established CVD [48,51,52].
However, since only 1520% of patients with type 2 diabetes
conform to the characteristics of patients in these trials, other
clinical features need to be considered in the majority when
selecting second medications to add to metformin (Figs 2,3,
4,5,6)[149,196204].
Sulfonylureas and insulin are associated with an increased
risk for causing hypoglycaemia and would not be preferred for
patients in whom this is a concern. Furthermore,
hypoglycaemia is distressing and so may reduce treatment ad-
herence (Fig. 5). For patients prioritizing weight loss or weight
maintenance (Fig. 4), important considerations include the
weight reduction associated with SGLT-2 inhibitors and
GLP-1 receptor agonists, the weight neutrality ofDPP-4 inhib-
itors, and the weight gain associated with sulfonylureas, basal
insulin and TZDs. An important consideration for society in
general and for many patients in particular is the cost of med-
ications; sulfonylureas, pioglitazone and recombinant human
insulins are relatively inexpensive, although their cost may
vary across regions. Short-term acquisition costs, longer-term
treatment cost and cost-effectiveness should be considered in
clinical decision making when data are available (Fig. 6).
Intensification beyond two medications
The lack of a substantial response to one or more non-insulin
therapies should raise the issue of adherence and, in those with
weight loss, the possibility that the patient has autoimmune
(type 1) or pancreatogenic diabetes. However, it is common in
people with long-standing diabetes to require more than two
glucose-lowering agents, often including insulin. Compared
with the knowledge base guiding dual therapy of type 2 dia-
betes, there is less evidence guiding these choices [205]. In
general, intensification of treatment beyond two medications
follows the same general principles as the addition of a second
medication, with the assumption that the efficacy of third and
fourth medications will be generally less than expected. No
specific combination has demonstrated superiority except for
those that include insulin and GLP-1 receptor agonists that
have broad ranges of glycaemic efficacy. As more medica-
tions are added, there is an increased risk of adverse effects.
It is important to consider medication interactions and whether
regimen complexity may become an obstacle to adherence.
Finally, with each additional medication comes increased
costs, which can affect patient burden, medication-taking be-
haviour and medication effectiveness. [193,205211].
While most patients require intensification of glucose-
lowering medications, some require medication reduction
or discontinuation of medication, particularly if the therapy
is ineffective or is exposing patients to a higher risk of side
effects such as hypoglycaemia, or when glycaemic goals
have changed due to a change in clinical circumstances
(e.g. development of comorbidities or even healthy age-
ing). A guiding principle is that for all therapies the re-
sponse should be reviewed at regular intervals, including
the impact on efficacy (HbA
1c
, weight) and safety; the
therapy should be stopped, or the dose reduced if there
are minimal benefits or if harm outweighs any benefit. In
particular, ceasing or reducing the dose of medications that
have an increased risk of hypoglycaemia is important when
any new glucose-lowering treatment (lifestyle or medica-
tion) is started (Fig. 7)[40]. HbA
1c
levels below 48 mmol/
mol (6.5%) or substantially below the individualised
glycaemic target should prompt consideration of stopping
Consensus recommendation
Intensification of treatment beyond dual therapy to
maintain glycaemic targets requires consideration of the
impact of medication side effects on comorbidities, as
well as the burden of treatment and cost.
Consensus recommendation
The selection of medication added to metformin is
based on patient preference and clinical characteristics.
Important clinical characteristics include the presence of
established ASCVD and other comorbidities such as HF
or CKD; the risk for specific adverse medication effects,
particularly hypoglycaemia and weight gain; as well as
safety, tolerability and cost (Figs 2–6).
Diabetologia
or reducing the dose of medications with risk of
hypoglycaemia or weight gain.
Addition of injectable medications
See the Insulinand Basal insulinsections in Medications
for lowering glucosefor more medication details.
Patients often prefer combinations of oral medications
to injectable medications. The range of combinations avail-
able with current oral medications allows many people to
reach glycaemic targets safely. However, there is currently
no evidence that any single medication or combination has
durable effects and, for many patients, injectable medica-
tions become necessary within 510 years of diabetes
diagnosis.
Evidence from trials comparing GLP-1 receptor agonists
and insulin (basal, premixed or basal-bolus) shows similar or
even better efficacy in HbA
1c
reduction [212,213]. GLP-1
receptor agonists have a lower risk of hypoglycaemia and
are associated with reductions in body weight compared with
weight gain with insulin [212,214]. Some GLP-1 receptor
agonists allow for once weekly injections, as opposed to daily
or more often for insulin. Based on these considerations, a
GLP-1 receptor agonist is the preferred option in a patient with
a definite diagnosis of type 2 diabetes who needs injectable
therapy. However, the tolerability and high cost of GLP-1
receptor agonists are important limitations to their use. If ad-
ditional glucose lowering is needed despite therapy with a
long-acting GLP-1 receptor agonist, the addition of basal in-
sulin is a reasonable option [215,216].
Alternatively, the addition of insulin to oral medica-
tion regimens is well established. In particular, using
basal insulin in combination with oral medications is
effective, and has less hypoglycaemia and weight gain
than combinations using premixed insulin formulations
or prandial insulin [217]. A standard approach for
optimising basal insulin regimens is to titrate the dose
based on a target fasting glucose concentration, which
is a simple index of effectiveness. Either NPH insulin
or long-acting insulin analogues are efficacious for con-
trolling fasting glucose, although basal analogue formu-
lations show reduced risks of hypoglycaemia, particularly
overnight, when titrated to the same fasting glucose tar-
get as NPH insulin [157,218].
Beyond basal insulin
It has become common practice to approach insulin use in
people with type 2 diabetes by following the established par-
adigms developed for those with type 1 diabetes. This in-
cludes multiple daily injections with doses of insulin ana-
logues before meals that are adjusted based on ambient blood
glucose and meal constituents. While this is reasonable for
people with type 2 diabetes who are lean, insulinopenic and
sensitive to exogenous insulin, it ignores the substantial dif-
ferences in pathophysiology between most people with type 2
diabetes and type 1 diabetes. Most people withtype 2 diabetes
are obese and insulin resistant, requiring much larger doses of
insulin and experiencing lower rates of hypoglycaemia than
those with type 1 diabetes. In patients with type 2 diabetes,
weight gain is a particularly problematic side effect of insulin
use. Recent evidence supports the effectiveness of combina-
tions of insulin with glucose-lowering medications that do not
increase body weight. For example, SGLT2 inhibitors can be
added to insulin regimens to lower blood glucose levels with-
out increasing insulin doses, weight gain or hypoglycaemia
[219221]. In a meta-analysis that studied the combination of
either SGLT2 inhibitors or DPP-4 inhibitors with insulin, the
SGLT2 inhibitorinsulin combination was associated with a
greater reduction in HbA
1c
, an advantage in terms of body
weight and no increase in the rates of hypoglycaemia [222,
223]. Depending on baseline HbA
1c
, glycaemic profile and
individual response, the insulin dose may need to be reduced
to prevent hypoglycaemia when adding an SGLT2 inhibitor.
The combination of basal insulin and a GLP-1 receptor
agonist has high efficacy, with recent evidence from clinical
trials demonstrating the benefits of this combination to lower
HbA
1c
and limit weight gain and hypoglycaemia compared
with intensified insulin regimens [224,225]. Most data come
from studies in which a GLP-1 receptor agonist is added to
basal insulin. However, there is evidence that insulin added to
a GLP-1 receptor agonist can also effectively lower HbA
1c
,
although some weight gain results [215]. Fixed-ratio combi-
nations of insulin and GLP-1 receptor agonists are available
and can decrease the number of injections compared with
administering the medications separately [226228].
A final approach to glycaemic management when basal
insulin plus oral medications is insufficient to achieve
HbA
1c
targets is intensified insulin regimens (Figs 7and 8).
DSMES focused on insulin therapy is particularly helpful
when intensified insulin therapy is considered. Referral to a
Consensus recommendation
Patients who are unable to maintain glycaemic targets
on basal insulin in combination with oral medications
can have treatment intensified with GLP-1 receptor
agonists, SGLT2 inhibitors or prandial insulin (Figs 7
and 8).
Consensus recommendation
In patients who need the greater glucose-lowering effect
of an injectable medication, GLP-1 receptor agonists
are the preferred choice to insulin. For patients with
extreme and symptomatic hyperglycaemia, insulin is
recommended (Fig. 7).
Diabetologia
diabetes specialist team should be considered in cases where
the provider is uncomfortable or unfamiliar with intensifica-
tion, poor outcomes continue despite intensification, or pa-
tients have other issues that complicate intensification.
Intensified insulin regimens include (1) one or more daily in-
jections of rapid- or short-acting insulin before meals (prandial
insulin) or (2) switching to one to three daily administrations of
a fixed combination of short- and long-acting insulin
(premixed or biphasic insulins) [229,230]. When adding pran-
dial insulin, giving one injection with the largest meal of the
day is a simple and safe approach [231]. Over time, if
glycaemic targets are not met with one dose of prandial insulin
daily, additional prandial injections can be added to other
meals [232]. Results of meta-analyses suggest a modestly
Fig. 8 Considering oral therapy in combination with injectable therapies
Diabetologia
greater reduction in HbA
1c
with basal-prandial regimens com-
pared with biphasic insulin regimens, but at the expense of
greater weight gain [233235]. While still commonly used,
we do not generally advocate premixed insulin regimens, par-
ticularly those administered three times daily, for routine use
when intensifying insulin regimens (Fig. 7).
Continuous insulin infusion using insulin pumps may have
a role in a small minority of people with type 2 diabetes [236].
Access and cost
The availability of glucose-lowering medications, patient sup-
port systems and blood glucose-monitoring devices can differ
worldwide, depending on a regions economy, culture and
healthcare system. Cost of and access to newer medications
and insulin remain important issues throughout the world.
Although the economics of diabetes care is complex and
broadly includes the costs to society of diabetic complications
and long-term outcomes, the cost of drugs and the affordability
of treatment are often the primary basis for decision making.
Within healthcare systems, variance in medication coverage is
based on different assessments of cost-effectiveness. This re-
sults in huge disparities in the cost of new and old glucose-
lowering medications in some countries, limiting access to the
full range of diabetes therapies in large segments of the popu-
lation and creating a two-tiered system of treatment. Since
glycaemic management remains a cornerstone of the preven-
tion of diabetes complications, these disparities raise questions
of fairness, equity and overall public health. Nonetheless, the
use of less expensive agents, such as metformin, sulfonylureas
and human insulin, remain effective options (Figs 2and 6).
Redoubling lifestyle management efforts can also have great
impact, but behavioural intervention and support can also be
costly, and socioeconomic barriers to improving lifestyle are
well-described [237].
Emerging technology
There is an increasing call for the use of technology and tele-
medicine to improve patientshealth [238]. Many types of in-
puts can be digitalised, such as blood glucose levels, time spent
exercising, steps walked, energy ingested, medication doses
administered, blood pressure and weight. Patterns in these var-
iables can be identified by software, leading to specific treat-
ment recommendations supported by real-time algorithms.
Telemedicine incorporates multiple types of communication
services, such as two-way video, email, texting, smartphones,
tablets, wireless monitors, decision support tools and other
forms of telecommunication technologies. Results overall sug-
gest a modest improvement in glycaemic control [239,240].
Key knowledge gaps
Despite over 200 years of research on lifestyle management of
diabetes and more than 50 years of comparative-effectiveness
research in diabetes, innumerable unanswered questions re-
garding the management of type 2 diabetes remain. In the
context of our current consensus recommendations, the fol-
lowing is an incomplete discussion of vexing issues that must
be addressed.
Evolving areas of current investigation will provide im-
provements in diabetes care and hold great hope for new
treatments.
&Implementation science. The tools available to prevent
and treat diabetes are vastly improved. However, imple-
mentation of effective innovation has lagged behind.
&Basic science. Our understanding of the basic mechanisms
of diabetes, the development of complications, and the
treatment of both, though continuously advancing, has
highlighted how much we do not know.
&Personalised/precision medicine. Though promising,
these -omics and big data approaches addressing both per-
sonal and environmental factors and their interaction are
largely unrealised in diabetes care and will require large
investments and coordination to have impact.
&Informatics. The benefits and role of enhanced monitoring
of glucose and other variables leveraged with real-time in-
formatics-based approaches to adapt treatment on an indi-
vidual basis has great potential but has not been elucidated.
&Overweight/obesity. Current therapy is clearly inadequate.
Innovation in methods and implementation would trans-
form diabetes prevention and care. Understanding the bi-
ology, psychology and sociology of obesity to identify
pharmacological, behavioural and political approaches to
preventing and treating this principal cause of type 2 dia-
betes is essential.
&Lifestyle management and DSMES. Though the benefits
of these approaches are clear, better paradigms on how to
target, individualise and sustain the effects are needed.
&Beta-cell function. Preserving and enhancing beta cell
function is perceived as the holy grail of diabetes and yet
effective techniques are inadequately developed.
&Translational research. There is a huge gap between the
knowledge gained from clinical trials and application of
that information in clinical practice. This gap should be
filled with pragmatic studies and other designs that include
costs, measures of patient preference and other patient-
recorded outcome measures. Patients and other
Consensus recommendation
Access, treatment cost and insurance coverage should
all be considered when selecting glucose-lowering
medications.
Diabetologia
stakeholders should have more input into trial designs and
outcomes. Pragmatic designs will enhance generalisability
of resultsand reduce cost. Better application ofreal-world
evidencewill complement randomised trial evidence.
&Drug development. New medications will require demon-
stration of broad efficacy for glucose, comorbidities and/
or complications as well as safety and tolerability to com-
pete in the marketplace.
&Complications. Steatohepatitis, HF, non-albuminuric
CKD, chronic mental illness and other emerging issues
are complications in diabetes that may supplant classical
microvascular and macrovascular disease in importance
and impact. Understanding optimal diagnostic, screen-
ing and treatment strategies is urgently needed.
Other areas of importance include better segmentation of
type 2 diabetes, as well as appropriate diagnosis of secondary
diabetes, which should allow more informed individualisation of
care. Better data on optimal approaches to diabetes management
in frail and older adult patients is urgently required considering
the controversy around glycaemic targets and the benefits and
harms of specific treatments from lifestyle management to med-
ications. Current approaches to the management of type 2 diabe-
tes in adolescents and young adults do not seem to alter the loss
of beta cell function and most individuals in this age group
quickly transition to insulin therapy. Studies to guide optimal
therapyinthisemergingpopulationwithaterrifyinglyhighrisk
of early disability is an immediate need.
There are enduring questions that continue to challenge
guideline development. For example, does metformin provide
cardiovascular benefit in patients with type 2 diabetes early in
the natural history of diabetes, as suggested by the UKPDS
study? Is metformins role as first-line medication manage-
ment truly evidence-based or a quirk of history? Though the
rationale for early combinationtherapy targeting normal levels
of glycaemia in early diabetes is seductive, clinical trial evi-
dence to support specific combinations and targets is essen-
tially non-existent. As the cost implications for these ap-
proaches is enormous, evidence is desperately needed.
Different models of care are being implemented globally.
Defining optimal cost-effective approaches to care, particular-
ly in the management of patients (multi-morbidity), is
essential.
New questions arise from the recent cardiovascular out-
comes studies. Do the cardiovascular and renal benefits of
SGLT2 inhibitors and GLP-1 receptor agonists demonstrated
in patients with established CVD extend to lower-risk pa-
tients? Is there additive benefit of use of GLP-1 receptor ago-
nists and SGLT2 inhibitors for prevention of cardiovascular
and renal events? If so, in what populations?
Addressing these and other vital clinical questions will re-
quire additional investment in basic, translational, clinical and
implementation research. More time- and cost-efficient research
paradigms to address patient-centred endpoints will need to be
developed through regulatory reform and leveraging informat-
ics and coordinated learning healthcare systems. The increasing
burden of cardiorenal metabolic disease in terms of incidence,
prevalence and cost is an existential threat to society. Urgent
attention to improve prevention and treatment is of the essence.
The management of hyperglycaemia in type 2 diabetes has
become extraordinarily complex with the number of glucose-
lowering medications now available. Patient-centred decision
making and support and consistent efforts to improve diet and
exercise remain the foundation of all glycaemic management.
Initial use of metformin, followed by addition of glucose-
lowering medications based on patient comorbidities and con-
cerns is recommended as we await answers to the many ques-
tions that remain.
Acknowledgements The authors would like to acknowledge Mindy
Saraco (Associate Director, Scientific & Medical Communication),
Gedeon Topacio (Finance & Project Manager, Research & Scientific
Programs) and Erika Berg (PhD, Director, Scientific and Medical
Affairs) from the American Diabetes Association as well as Mary Hata
(Executive Assistant) and Petra Niemann (Executive Assistant) from
EASD for their help with the development of the consensus report and
related meetings/presentations.
The authors would like to also acknowledge Mike Bonar (Creative
Director) and Charlie Franklin (Design Assistant) from the Leicester
Diabetes Centre, Leicester, UK, who provided considerable support in
drafting and amending the figures. The authors also acknowledge
Francesco Zaccardi (PhD, Clinical Research Fellow, University of
Leicester, Leicester, UK) and David Kloecker (Medical Student, University
of Leicester) who assisted with extracting PubMed articles and identifying
relevant records by title and abstract; Francesco Zaccardi helped to define the
initial search strategy and prepare the Excel file.
The authors acknowledge the invited peer reviewers who provided com-
ments on an earlier draft of this report: Amanda Adler (Addenbrookes
Hospital, Cambridge, UK), Kåre I. Birkeland (University of Oslo, Oslo,
Norway), James J. Chamberlain (St. Marks Hospital, Salt Lake City, UT,
USA), Jill P. Crandall (Albert Einstein College of Medicine, New York City,
NY, USA), Ian H. de Boer (University of Washington, Seattle, WA, USA),
Stefano Del Prato (University of Pisa, Pisa, Italy), George Dimitriadis (Athens
University, Athens, Greece), Sean Dinneen (National University of Ireland,
Galway, Ireland), Vivian A. Fonseca (Tulane University, New Orleans, LA,
USA), Simon R. Heller (University of Sheffield, Sheffield, UK), Richard I. G.
Holt (University of Southampton, Southampton, UK), Silvio E. Inzucchi
(Yale University, New Haven, CT, USA), Eric L. Johnson (University of
North Dakota, Grand Forks, ND, USA), Joshua J. Neumiller (Washington
State University, Spokane, WA, USA), Kamlesh Khunti (University of
Leicester, Leicester, UK), Harald H. Klein (Ruhr University of Bochum,
Bochum, Germany), Line Kleinebreil (Hôpital national de Saint Maurice,
Saint-Maurice, France), José Manuel Fernández-Real (Universitat de
Girona, Girona, Spain), Sally M. Marshall (Newcastle University,
Newcastle upon Tyne, UK), Manel Mata-Cases (Institut Universitari
dInvestigació en Atenció Primària Jordi Gol [IDIAP Jordi Gol], Barcelona,
Spain), David R. Matthews (University of Oxford, Oxford, UK), David M.
Nathan (Massachusetts General Hospital, Boston, MA, USA), Michael A.
Nauck (Diabetes Center Bochum-Hattingen, St Josef-Hospital, Ruhr-
University, Bochum, Germany), Frank Nobels (OLV-Hospital, Aalst,
Belgium), Richard E. Pratley (Florida Hospital Diabetes Institute, Orlando,
FL, USA), Maria Jose Redondo (Baylor College of Medicine, Houston, TX,
USA), Michael R. Rickels (University of Pennsylvania, Philadelphia, PA,
USA), Matthew C. Riddle (Oregon Health & Science University, Portland,
OR, USA), Julio Rosenstock (Diabetes and Endocrine Center, Dallas, TX,
Diabetologia
USA), Giorgio Sesti (Magna Graecia University of Catanzaro, Catanzaro,
Italy), Neil Skolnik (Abington Family Medicine, Jenkintown, PA, USA),
Krzysztof Strojek (Silesian Medical University, Zabrze, Poland), Jennifer
Trujillo (University of Colorado, Denver, CO, USA), Guillermo E.
Umpierrez (Emory University, Atlanta, GA, USA) and Jennifer Wyckoff
(University of Michigan, Ann Arbor, MI, USA).
Data availability The details of the search strategy, the results and the
classification for the included articles are available at https://doi.org/10.
17632/h5rcnxpk8w.1.
Funding This activity was funded by the American Diabetes Association
and the European Association for the Study of Diabetes.
Duality of interest M. J. Davies reports personal fees and grants from
Boehringer Ingelheim, Janssen, Novo Nordisk and Sanofi and personal
fees from AstraZeneca, Eli Lilly, Gilead Sciences Ltd., Intarcia/Servier,
Merck Sharp & Dohme, Mitsubishi Tanabe Pharma Corporation and
Takeda Pharmaceuticals International Inc.
D. DAlessio reports personal fees from Eli Lilly, Merck, Novo
Nordisk, and Intarcia, and grants from Merck, and Ligand during the con-
duct of the study; personal fees from Eli Lilly, Merck, Novo Nordisk, and
Intarcia, and grants from Merck, and Ligand outside the submitted work.
J. Fradkin has nothing to disclose. J. Fradkins input into this consen-
sus report is from her own perspective and the Report does not reflect the
view of the National Institutes of Health, Department of Health and
Human Services or the US Government.
W. N. Kernan has nothing to disclose.
C. Mathieu reports grants and personal fees from Novo Nordisk,
grants and personal fees from Sanofi, grants and personal fees from
Merck Sharp & Dohme Ltd., grants and personal fees from Eli Lilly
and Company, grants and personal fees from Novartis, personal fees from
Bristol-Myers Squibb, personal fees from AstraZeneca, grants and per-
sonal fees from Boehringer Ingelheim, personal fees from Hanmi
Pharmaceuticals, grants and personal fees from Roche Diagnostics, grants
and personal fees from Medtronic, grants and personal fees from
Intrexon, grants and personal fees from Abbott, and personal fees from
UCB, outside the submitted work.
G Mingrone reports grants and personal fees from Novo Nordisk,
personal fees from Johnson & Johnson, personal fees from Fractyl Inc.,
during the conduct of the study.
P. Rossing reports grants, non-financial support and other from Novo
Nordisk, grants and other from AstraZeneca, other from Bayer, other
from Boehringer Ingelheim, other from MSD, other from Eli Lilly, during
the conduct of the study.
A. Tsapas reports non-financial support from the European Association
for the Study of Diabetes, during the conduct of the study; grants and other
from Boehringer Ingelheim, grants and other from Novo Nordisk, other
from Novartis, grants and other from Sanofi, grants and other from
AstraZeneca, grants from GSK, grants and other from European
Foundation for the Study of Diabetes (EFSD), outside the submitted work.
D. J. Wexler has nothing to disclose.
J. B. Buse has provided consultation to Adocia, AstraZeneca, Eli
Lilly, GI Dynamics, Intarcia, MannKind, NovaTarg, Novo Nordisk,
Senseonics, and vTv Therapeutics with fees paid to the University of
North Carolina. He has received grant support from AstraZeneca,
Johnson & Johnson, Novo Nordisk, Sanofi, and vTv Therapeutics. He
is a consultant to Neurimmune AG. He holds stock options in Mellitus
Health, PhaseBio and Stability Health. He is supported by a grant from
the National Institutes of Health (UL1TR002489).
Contribution statement All authors were responsible for drafting the
article and revising it critically for important intellectual content. All
authors approved the version to be published.
References
1. Rodriguez-Gutierrez R, Gionfriddo MR, Ospina NS et al (2016)
Shared decision making in endocrinology: present and future di-
rections. Lancet Diabetes Endocrinol 4:706716. https://doi.org/
10.1016/S2213-8587(15)00468-4
2. American Diabetes Association (2018) 6. Glycemic targets: stan-
dards of medical care in diabetes2018. Diabetes Care 41:S55
S64. https://doi.org/10.2337/dc18-S006
3. American Diabetes Association (2018) 8. Pharmacologic ap-
proaches to glycemic treatment: standards of medical care in dia-
betes2018. Diabetes Care 41:S73S85. https://doi.org/10.2337/
dc18-S008
4. Inzucchi SE, Bergenstal RM, Buse JB et al (2012) Management of
hyperglycaemia in type 2 diabetes: a patient-centered approach.
Position statement of the American Diabetes Association (ADA)
and the European Association for the Study of Diabetes (EASD).
Diabetologia 55:15771596
5. Inzucchi SE, Bergenstal RM, Buse JB et al (2015) Management of
hyperglycaemia in type 2 diabetes, 2015: a patient-centered ap-
proach. Update to a position statement of the American Diabetes
Association and the European Association for the Study of
Diabetes. Diabetologia 58:429442. https://doi.org/10.2337/
dc14-2441
6. Riddle MC, Gerstein HC, Holman RR et al (2018) A1C targets
should be personalized to maximize benefits while limiting risks.
Diabetes Care 41:11211124. https://doi.org/10.2337/dci18-0018
7. American Diabetes Association (2018) 9. Cardiovascular dis-
ease and risk management: standards of medical care in dia-
betes-2018. Diabetes Care 41:S86S104. https://doi.org/10.
2337/dc18-S009
8. Gæde P, Oellgaard J, Carstensen B et al (2016) Years of life gained
by multifactorial intervention in patients with type 2 diabetes
mellitus and microalbuminuria: 21 years follow-up on the Steno-
2 randomised trial. Diabetologia 59:22982307. https://doi.org/
10.1007/s00125-016-4065-6
9. Khunti K, Kosiborod M, Ray KK (2018) Legacy benefits of blood
glucose, blood pressure and lipid control in individuals with dia-
betes and cardiovascular disease: time to overcome multifactorial
therapeutic inertia? Diabetes Obes Metab 20:13371341. https://
doi.org/10.1111/dom.13243
10. Gregg EW, Sattar N,Ali MK (2016) The changing face ofdiabetes
complications. Lancet Diabetes Endocrinol 4:537547. https://
doi.org/10.1016/S2213-8587(16)30010-9
11. Little RR, Rohlfing CL, Sacks DB (2011) Status of HbA
1c
mea-
surement and goals for improvement: from chaos to order for
improving diabetes care. Clin Chem 57:204214
12. American Diabetes Association (2018) 2. Classification and diag-
nosis of diabetes: standards of medical care in diabetes2018.
Diabetes Care 41:S13S27. https://doi.org/10.2337/dc18-S002
13. MannucciE, Antenore A, Giorgino F, Scavini M (2018) Effects of
structured versus unstructured self-monitoring of blood glucose on
glucose control in patients with non-insulin-treated type 2 diabe-
tes: a meta-analysis of randomized controlled trials. J Diabetes Sci
Technol 12:183189. https://doi.org/10.1177/1932296817719290
14. Young LA, Buse JB, Weaver MA et al (2017) Glucose self-
monitoring in noninsulin-treated patients with type 2 diabetes
in primary care settings: a randomized trial. JAMA Intern Med
177:920929. https://doi.org/10.1001/jamainternmed.2017.1233
15. Anjana RM, Kesavadev J, Neeta D et al (2017) A multicenterreal-
life study on the effect of flash glucose monitoring on glycemic
control in patients with type 1 and type 2 diabetes. Diabetes
Technol Ther 19:533540
Diabetologia
16. American Diabetes Association (2018) 3. Comprehensive medical
evaluation and assessment of comorbidities: standards of medical
care in diabetes2018. Diabetes Care 41:S28S37. https://doi.
org/10.2337/dc18-S003
17. Kunneman M, Montori VM, Castaneda-Guarderas A, Hess EP
(2016) What is shared decision making? (and what it is not).
Acad Emerg Med 23:13201324. https://doi.org/10.1111/acem.
13065
18. Breslin M, Mullan RJ, Montori VM (2008) The design of a deci-
sion aid about diabetes medications for use during the consultation
with patients with type 2 diabetes. Patient Educ Couns 73:465
472. https://doi.org/10.1016/j.pec.2008.07.024
19. Mullan RJ, Montori VM, Shah ND et al (2009) The diabetes
mellitus medication choice decision aid: a randomized trial.
Arch Intern Med 169:15601568. https://doi.org/10.1001/
archinternmed.2009.293
20. Stacey D, Légaré F, Lewis K et al (2017) Decision aids for people
facing health treatmentor screening decisions.Cochrane Database
Syst Rev 4:CD001431. https://doi.org/10.1002/14651858.
CD001431.pub5
21. American Diabetes Association (2018) 4. Lifestyle management:
standards of medical care in diabetes2018. Diabetes Care 41:
S38S50. https://doi.org/10.2337/dc18-S004
22. Powers MA, Bardsley J, Cypress M et al (2015) Diabetes self-
management education and support in type 2 diabetes: a joint
position statement of the American Diabetes Association, the
American Association of Diabetes Educators, and the Academy
of Nutrition and Dietetics. Diabetes Care 38:13721382. https://
doi.org/10.2337/dc15-0730
23. Department of Health; Diabetes UK (2005) Structured patient ed-
ucation in diabetes: report from the Patient Education Working
Group. Available from http://webarchive.nationalarchives.gov.
uk/20130105204013/http://www.dh.gov.uk/en/
Publicationsandstatistics/Publications/PublicationsPolicy
AndGuidance/DH_4113195. Accessed 29 Aug 2018
24. National Institute for Health and Clinical Excellence (2011)
Quality standard for diabetes in adults, statements 2 and 3.
Available from https://www.nice.org.uk/guidance/qs6.Accessed
29 Aug 2018
25. Beck J, Greenwood DA, Blanton L et al (2017) 2017 national
standards for diabetes self-management education and support.
Diabetes Educ 43:449464
26. Chrvala CA, Sherr D, Lipman RD (2016) Diabetes self-
management education for adults with type 2 diabetes
mellitus: a systematic review of the effect on glycemic con-
trol. Patient Educ Couns 99:926943. https://doi.org/10.
1016/j.pec.2015.11.003
27. Pillay J, Armstrong MJ, Butalia S et al (2015) Behavioral pro-
grams for type 2 diabetes mellitus: a systematic review and net-
work meta-analysis. Ann Intern Med 163:848. https://doi.org/10.
7326/M15-1400
28. Zhao F-F, Suhonen R, Koskinen S,Leino-Kilpi H (2017) Theory-
based self-management educational interventions on patients with
type 2 diabetes: a systematic review and meta-analysis of random-
ized controlled trials. J Adv Nurs 73:812833. https://doi.org/10.
1111/jan.13163
29. Odgers-Jewell K, Ball LE, Kelly JT et al (2017) Effectiveness of
group-based self-management education for individuals with type
2 diabetes: a systematic review with meta-analyses and meta-re-
gression. Diabet Med 34:10271039. https://doi.org/10.1111/dme.
13340
30. He X, Li J, Wang B et al (2017) Diabetes self-management edu-
cation reduces risk of all-cause mortality in type 2 diabetes pa-
tients: a systematic review and meta-analysis. Endocrine 55:712
731. https://doi.org/10.1007/s12020-016-1168-2
31. Chatterjee S, Davies MJ, Heller S et al (2018) Diabetes structured
self-management education programmes: a narrative review and
current innovations. Lancet Diabetes Endocrinol 6:130142.
https://doi.org/10.1016/S2213-8587(17)30239-5
32. Egede LE, Gebregziabher M, Echols C, Lynch CP (2014)
Longitudinal effects of medication nonadherence on glycemic
control. Ann Pharmacother 48:562570. https://doi.org/10.1177/
1060028014526362
33. Huber CA, Reich O (2016) Medication adherence in patients with
diabetes mellitus: does physician drug dispensing enhance quality
of care? Evidence from a large health claims database in
Switzerland. Patient Prefer Adherence 10:18031809. https://doi.
org/10.2147/PPA.S115425
34. Iglay K, Cartier SE, Rosen VM et al (2015) Meta-analysis of
studies examining medication adherence, persistence, and discon-
tinuation of oral antihyperglycemic agents in type 2 diabetes. Curr
Med Res Opin 31:12831296. https://doi.org/10.1185/03007995.
2015.1053048
35. McGovern A, Tippu Z, Hinton W et al (2016) Systematic review
of adherence rates by medication class in type 2 diabetes: a study
protocol. BMJ Open 6:e010469. https://doi.org/10.1136/bmjopen-
2015-010469
36. Khunti K, Seidu S, Kunutsor S, Davies M (2017) Association
between adherence to pharmacotherapy and outcomes in type 2
diabetes: a meta-analysis. Diabetes Care 40:15881596. https://
doi.org/10.2337/dc16-1925
37. Polonsky WH, Henry RR (2016) Poor medication adherence in
type 2 diabetes: recognizing the scope of the problem and its key
contributors. Patient Prefer Adherence 10:12991307. https://doi.
org/10.2147/PPA.S106821
38. McGovern A, Tippu Z, Hinton W et al (2018) Comparison of
medication adherence and persistence in type 2 diabetes: a sys-
tematic review and meta-analysis. Diabetes Obes Metab 20:1040
1043. https://doi.org/10.1111/dom.13160
39. Lasalvia P, Barahona-Correa JE, Romero-Alvernia DM et al
(2016) Pen devices for insulin self-administration compared with
needle and vial: systematic review of the literature and meta-anal-
ysis. J Diabetes Sci Technol 10:959966. https://doi.org/10.1177/
1932296816633721
40. Khunti K, Davies MJ (2017) Clinical inertiatime to reappraise
the terminology? Prim Care Diabetes 11:105106. https://doi.org/
10.1016/j.pcd.2017.01.007
41. Furler J, ONeal D, Speight J et al (2017) Supporting insulin
initiation in type 2 diabetes in primary care: results of the
Stepping Up pragmatic cluster randomised controlled clinical trial.
BMJ:j783. https://doi.org/10.1136/bmj.j783
42. Manski-Nankervis J-A, Furler J, ONealDetal(2017)
Overcoming clinical inertia in insulin initiation in primary care
for patients with type 2 diabetes: 24-month follow-up of the
Stepping Up cluster randomised controlled trial. Prim Care
Diabetes 11:474481. https://doi.org/10.1016/j.pcd.2017.06.005
43. Tabesh M, Magliano DJ, Koye DN, ShawJE (2018) The effect of
nurse prescribers on glycaemic control in type 2 diabetes: a sys-
tematic review and meta-analysis. Int J Nurs Stud 78:3743.
https://doi.org/10.1016/j.ijnurstu.2017.08.018
44. Murphy ME, Byrne M, Galvin R et al (2017) Improving risk
factor management for patients with poorly controlled type 2 dia-
betes: a systematic review of healthcare interventions in primary
care and community settings. BMJ Open 7:e015135. https://doi.
org/10.1136/bmjopen-2016-015135
45. American Diabetes Association (2018) 1. Improving care and pro-
moting health in populations: standards of medical care in diabe-
tes2018. Diabetes Care 41:S7S12. https://doi.org/10.2337/
dc18-S001
46. Cefalu WT, Kaul S, Gerstein HC et al (2018) Cardiovascular out-
comestrialsintype2diabetes:wheredowegofromhere?
Diabetologia
Reflections from a Diabetes Care EditorsExpert Forum.
Diabetes Care 41:1431. https://doi.org/10.2337/dci17-0057
47. Marso SP, Daniels GH, Brown-Frandsen K et al (2016) Liraglutide
and cardiovascular outcomes in type 2 diabetes. N Engl J Med
375:311322. https://doi.org/10.1056/NEJMoa1603827
48. Marso SP, Bain SC, Consoli A et al (2016) Semaglutide and cardio-
vascular outcomes in patients with type 2 diabetes. N Engl J Med
375:18341844. https://doi.org/10.1056/NEJMoa1607141
49. Holman RR, Bethel MA, Mentz RJ et al (2017) Effects of once-
weekly exenatide on cardiovascular outcomes in type 2 diabetes.
N Engl J Med 377:12281239. https://doi.org/10.1056/
NEJMoa1612917
50. Pfeffer MA, Claggett B, Diaz R et al (2015) Lixisenatide in patients
with type 2 diabetes and acute coronary syndrome. N Engl J Med
373:22472257. https://doi.org/10.1056/NEJMoa1509225
51. Zinman B, Wanner C, Lachin JM et al (2015) Empagliflozin, cardio-
vascular outcomes, and mortality in type 2 diabetes. N Engl J Med
373:21172128. https://doi.org/10.1056/NEJMoa1504720
52. Neal B, Perkovic V, Mahaffey KW et al (2017) Canagliflozin and
cardiovascular and renal events in type 2 diabetes. N Engl J Med
377:644657. https://doi.org/10.1056/NEJMoa1611925
53. Lehrke M, Marx N (2017) Diabetes mellitus and heart failure. Am
J Med 130:S40S50. https://doi.org/10.1016/j.amjmed.2017.04.
010
54. Fitchett D, Zinman B, Wanner C et al (2016) Heart failure out-
comes with empagliflozin in patients with type 2 diabetes at high
cardiovascular risk: results of the EMPA-REG OUTCOME® trial.
Eur Heart J 37:15261534. https://doi.org/10.1093/eurheartj/
ehv728
55. Rådholm K, Figtree G, Perkovic Vet al (2018) Canagliflozin and
heart failure in type 2 diabetes mellitus: results from the CANVAS
Program (Canagliflozin Cardiovascular Assessment Study).
Circulation 138:458468. https://doi.org/10.1161/
CIRCULATIONAHA.118.034222
56. Margulies KB, Hernandez AF, Redfield MM et al (2016) Effects
of liraglutide on clinical stability among patients with advanced
heart failure and reduced ejection fraction: a randomized clinical
trial. JAMA 316:500508. https://doi.org/10.1001/jama.2016.
10260
57. Jorsal A, Kistorp C, Holmager P et al (2017) Effect of liraglutide, a
glucagon-like peptide-1 analogue, on left ventricular function in
stable chronic heart failure patients with and without diabetes
(LIVE)-a multicentre, double-blind, randomised, placebo-
controlled trial. Eur J Heart Fail 19:6977. https://doi.org/10.
1002/ejhf.657
58. Scirica BM, Bhatt DL, Braunwald E et al (2013) Saxagliptin and
cardiovascular outcomes in patients with type 2 diabetes mellitus.
N Engl J Med 369:13171326. https://doi.org/10.1056/
NEJMoa1307684
59. White WB, Cannon CP, Heller SR et al (2013) Alogliptin after acute
coronary syndrome in patients with type 2 diabetes. N Engl J Med
369:13271335. https://doi.org/10.1056/NEJMoa1305889
60. Green JB, Bethel MA, Armstrong PW et al (2015) Effect of
sitagliptin on cardiovascular outcomes in type 2 diabetes. N Engl
J Med 373:232242. https://doi.org/10.1056/NEJMoa1501352
61. Wanner C, Inzucchi SE, Lachin JM et al (2016)Empagliflozin and
progression of kidney disease in type 2 diabetes. N Engl J Med
375:323334. https://doi.org/10.1056/NEJMoa1515920
62. Jardine MJ, Mahaffey KW, Neal B et al (2017) The Canagliflozin
and Renal Endpoints in Diabetes with Established Nephropathy
Clinical Evaluation (CREDENCE) study rationale, design, and
baseline characteristics. Am J Nephrol 46:462472. https://doi.
org/10.1159/000484633
63. Mann JFE, Ørsted DD, Brown-Frandsen K et al (2017) Liraglutide
and renal outcomes in type 2 diabetes. N Engl J Med 377:839848.
https://doi.org/10.1056/NEJMoa1616011
64. Deacon CF (2018) A review of dipeptidyl peptidase-4 inhibitors.
Hot topics from randomized controlled trials. Diabetes Obes
Metab 20(Suppl 1):3446. https://doi.org/10.1111/dom.13135
65. American Diabetes Association (2018) 7. Obesity management
for the treatment of type 2 diabetes: standards of medical care in
diabetes2018. Diabetes Care 41:S65S72. https://doi.org/10.
2337/dc18-S007
66. Shai I, Schwarzfuchs D, Henkin Y et al (2008) Weight loss with a
low-carbohydrate, Mediterranean, or low-fat diet. N Engl J Med
359:229241. https://doi.org/10.1056/NEJMoa0708681
67. Esposito K, Maiorino MI, Ciotola M et al (2009) Effects of a
Mediterranean-style diet on the need for antihyperglycemic drug
therapy in patients with newly diagnosed type 2 diabetes: a ran-
domized trial. Ann Intern Med 151:306314
68. Esposito K, Maiorino MI, Petrizzo M et al (2014) The effects of a
Mediterranean diet on the need for diabetes drugs and remission of
newly diagnosed type 2 diabetes: follow-up of a randomized trial.
Diabetes Care 37:18241830. https://doi.org/10.2337/dc13-2899
69. Huo R, Du T, Xu Yet al (2015) Effects of Mediterranean-style diet
on glycemic control, weight loss and cardiovascular risk factors
among type 2 diabetes individuals: a meta-analysis. Eur J Clin
Nutr 69:12001208. https://doi.org/10.1038/ejcn.2014.243
70. Snorgaard O, Poulsen GM, Andersen HK, Astrup A (2017)
Systematic review and meta-analysis of dietary carbohydrate re-
strictionin patients withtype 2 diabetes.BMJ Open Diabetes Res
Care 5:e000354. https://doi.org/10.1136/bmjdrc-2016-000354
71. Ajala O, English P, Pinkney J (2013) Systematic review and meta-
analysis of different dietary approaches to the management of type
2 diabetes. Am J Clin Nutr 97:505516. https://doi.org/10.3945/
ajcn.112.042457
72. Azadbakht L, Fard NRP, Karimi M et al (2011) Effects of the
Dietary Approaches to Stop Hypertension (DASH) eating plan
on cardiovascular risks among type 2 diabetic patients: a random-
ized crossover clinical trial. Diabetes Care 34:5557. https://doi.
org/10.2337/dc10-0676
73. Sainsbury E, Kizirian NV, Partridge SR et al (2018) Effect of
dietary carbohydrate restriction on glycemic control in adults with
diabetes: a systematic review and meta-analysis. Diabetes Res
Clin Pract 139:239252. https://doi.org/10.1016/j.diabres.2018.
02.026
74. Yokoyama Y, Barnard ND, Levin SM, Watanabe M (2014)
Vegetarian diets and glycemic control in diabetes: a systematic
review and meta-analysis. Cardiovasc Diagn Ther 4:373382.
https://doi.org/10.3978/j.issn.2223-3652.2014.10.04
75. Lean ME, Leslie WS, Barnes AC et al (2018) Primary care-led
weight management for remission of type 2 diabetes (DiRECT):
an open-label, cluster-randomised trial. Lancet 391:541551.
https://doi.org/10.1016/S0140-6736(17)33102-1
76. Look AHEAD Research Group, Wing RR, Bolin P et al (2013)
Cardiovascular effects of intensive lifestyle intervention in type 2
diabetes. N Engl J Med 369:145154. https://doi.org/10.1056/
NEJMoa1212914
77. ONeil PM, Miller-Kovach K, Tuerk PW et al (2016)
Randomized controlled trial of a nationally available weight
control program tailored for adults with type 2 diabetes. Obes
Silver Spring Md 24:22692277. https://doi.org/10.1002/oby.
21616
78. Jebb SA, Ahern AL, Olson AD et al (2011) Primary care referral
to a commercial providerfor weight loss treatment versus standard
care: a randomised controlled trial. Lancet 378:14851492.
https://doi.org/10.1016/S0140-6736(11)61344-5
79. Leblanc ES, OConnor E, Whitlock EP et al (2011) Effectiveness
of primary care-relevant treatments for obesity in adults: a system-
atic evidence review for the U.S. Preventive Services Task Force.
AnnInternMed155:434447. https://doi.org/10.7326/0003-
4819-155-7-201110040-00006
Diabetologia
80. Delahanty LM, Dalton KM, Porneala B et al (2015) Improving
diabetes outcomes through lifestyle change a randomized con-
trolled trial. Obes Silver Spring Md 23:17921799. https://doi.org/
10.1002/oby.21172
81. Boulé NG, Haddad E, Kenny GP et al (2001) Effects of
exercise on glycemic control and body mass in type 2 diabe-
tes mellitus: a meta-analysis of controlled clinical trials.
JAMA 286:12181227
82. Chudyk A, Petrella RJ (2011) Effects of exercise on cardiovascu-
lar risk factors in type 2 diabetes: a meta-analysis. Diabetes Care
34:12281237. https://doi.org/10.2337/dc10-1881
83. Yang Z, Scott CA, Mao C et al (2014) Resistance exercise versus
aerobic exercise for type 2 diabetes: a systematic review and meta-
analysis. Sports Med Auckl NZ 44:487499. https://doi.org/10.
1007/s40279-013-0128-8
84. Balducci S, Zanuso S, Nicolucci A et al (2010) Effect of an inten-
sive exercise intervention strategy on modifiable cardiovascular
risk factors in subjects with type 2 diabetes mellitus: a randomized
controlled trial: the Italian Diabetes and Exercise Study (IDES).
Arch Intern Med 170:17941803. https://doi.org/10.1001/
archinternmed.2010.380
85. Schwingshackl L, Missbach B, Dias S et al (2014) Impact of
different training modalities on glycaemic control and blood lipids
in patients with type 2 diabetes: a systematic review and network
meta-analysis. Diabetologia 57:17891797. https://doi.org/10.
1007/s00125-014-3303-z
86. Qiu S, Cai X, Schumann U et al (2014) Impact of walking on
glycemic control and other cardiovascular risk factors in type 2
diabetes: a meta-analysis. PLoS One 9:e109767. https://doi.org/
10.1371/journal.pone.0109767
87. Rees JL, Johnson ST, Boulé NG (2017) Aquatic exercise for
adults with type 2 diabetes: a meta-analysis. Acta Diabetol 54:
895904. https://doi.org/10.1007/s00592-017-1023-9
88. Pai L-W, Li T-C, Hwu Y-J et al (2016) The effectiveness of regular
leisure-time physical activities on long-term glycemic control in
people with type 2 diabetes: a systematic review and meta-analy-
sis. Diabetes Res Clin Pract 113:7785. https://doi.org/10.1016/j.
diabres.2016.01.011
89. Lee MS, Jun JH, Lim H-J, Lim H-S (2015) A systematic review
and meta-analysis of tai chi for treating type 2 diabetes. Maturitas
80:1423. https://doi.org/10.1016/j.maturitas.2014.09.008
90. Cui J, Yan J-H,Yan L-M et al (2017) Effects of yoga in adults with
type 2 diabetes mellitus: a meta-analysis. J Diabetes Investig 8:
201209. https://doi.org/10.1111/jdi.12548
91. Qiu S, Cai X, Chen X et al (2014) Step counter use in type 2
diabetes: a meta-analysis of randomized controlled trials. BMC
Med 12:36. https://doi.org/10.1186/1741-7015-12-36
92. Franz MJ, Boucher JL, Rutten-Ramos S, VanWormer JJ (2015)
Lifestyle weight-loss intervention outcomes in overweight and
obese adults with type 2 diabetes: a systematic review and meta-
analysis of randomized clinical trials. J Acad Nutr Diet 115:1447
1463. https://doi.org/10.1016/j.jand.2015.02.031
93. Aggarwal N, Singla A, Mathieu C et al (2018) Metformin
extended-release versus immediate-release: an international, ran-
domized, double-blind, head-to-head trial in pharmacotherapy-
naïve patients with type 2 diabetes. Diabetes Obes Metab 20:
463467. https://doi.org/10.1111/dom.13104
94. Garber AJ, Duncan TG, Goodman AM et al (1997) Efficacy of
metformin in type II diabetes: results of a double-blind, placebo-
controlled, dose-response trial. Am J Med 103:491497
95. Inzucchi SE, Lipska KJ, Mayo H et al (2014) Metformin in pa-
tients with type 2 diabetes and kidney disease: a systematic review.
JAMA 312:26682675. https://doi.org/10.1001/jama.2014.15298
96. Lalau J-D,Kajbaf F, Bennis Yet al (2018) Metformin treatment in
patients with type 2 diabetes and chronic kidney disease stages
3A, 3B, or 4. Diabetes Care 41:547553. https://doi.org/10.2337/
dc17-2231
97. Imam TH (2017) Changes in metformin use in chronic kidney
disease. Clin Kidney J 10:301304. https://doi.org/10.1093/ckj/
sfx017
98. UK Prospective Diabetes Study (UKPDS) Group (1998) Effect of
intensive blood-glucose control with metformin on complications
in overweight patients with type 2 diabetes (UKPDS 34). Lancet
352:854865
99. Griffin SJ, Leaver JK, Irving GJ (2017) Impact of metformin on
cardiovascular disease: a meta-analysis of randomised trials
among people with type 2 diabetes. Diabetologia 60:16201629.
https://doi.org/10.1007/s00125-017-4337-9
100. Maruthur NM, Tseng E, Hutfless S et al (2016) Diabetes medica-
tions as monotherapy or metformin-based combination therapy for
type 2 diabetes: a systematic review and meta-analysis. Ann Intern
Med 164:740751. https://doi.org/10.7326/M15-2650
101. Aroda VR, Edelstein SL, Goldberg RB et al (2016) Long-term
metforminuseandvitaminB12deficiencyintheDiabetes
Prevention Program Outcomes Study. J Clin Endocrinol Metab
101:17541761. https://doi.org/10.1210/jc.2015-3754
102. Zhang X-L, Zhu Q-Q, Chen Y-H et al (2018) Cardiovascular safe-
ty, long-term noncardiovascular safety, and efficacy of sodium-
glucose cotransporter 2 inhibitors in patients with type 2 diabetes
mellitus:a systemic review and meta-analysis with trial sequential
analysis. J Am Heart Assoc 7:e007165. https://doi.org/10.1161/
JAHA.117.007165
103. Storgaard H, Gluud LL, Bennett C et al(2016) Benefits and harms
of sodium-glucose co-transporter 2 inhibitors in patients with type
2 diabetes: a systematic review and meta-analysis. PLoS One 11:
e0166125. https://doi.org/10.1371/journal.pone.0166125
104. Li D, Wang T, Shen S et al (2017) Urinary tract and genital infec-
tions in patients with type 2 diabetes treated with sodium-glucose
co-transporter 2 inhibitors: a meta-analysis of randomized con-
trolled trials. Diabetes Obes Metab 19:348355. https://doi.org/
10.1111/dom.12825
105. Jabbour S, Seufert J, Scheen A et al (2018) Dapagliflozin in pa-
tients with type 2 diabetes mellitus: a pooled analysis of safety data
from phase IIb/III clinical trials. Diabetes Obes Metab 20:620
628. https://doi.org/10.1111/dom.13124
106. Tang H, Li D, Wang T et al (2016) Effect of sodium-glucose
cotransporter 2 inhibitors on diabetic ketoacidosis among patients
with type 2 diabetes: a meta-analysis of randomized controlled
trials. Diabetes Care 39:e123e124. https://doi.org/10.2337/
dc16-0885
107. Thrasher J (2017) Pharmacologic management of type 2 diabetes
mellitus: available therapies. Am J Med 130:S4S17. https://doi.
org/10.1016/j.amjmed.2017.04.004
108. Karagiannis T, Liakos A, Bekiari E et al (2015) Efficacy and safety
of once-weekly glucagon-like peptide 1 receptor agonists for the
management of type 2 diabetes: a systematic review and meta-
analysis of randomized controlled trials. Diabetes Obes Metab
17:10651074. https://doi.org/10.1111/dom.12541
109. Zaccardi F, Htike ZZ, Webb DR et al (2016) Benefits and harms of
once-weekly glucagon-like peptide-1 receptor agonist treatments:
a systematic review and network meta-analysis. Ann Intern Med
164:102113. https://doi.org/10.7326/M15-1432
110. Htike ZZ, Zaccardi F, Papamargaritis D et al (2017) Efficacy and
safety of glucagon-like peptide-1 receptor agonists in type 2 dia-
betes: a systematic review and mixed-treatment comparison anal-
ysis. Diabetes Obes Metab 19:524536. https://doi.org/10.1111/
dom.12849
111. Sorli C, Harashima S-I, Tsoukas GM et al (2017) Efficacy and
safety of once-weekly semaglutide monotherapy versus placebo
in patients with type 2 diabetes (SUSTAIN 1): a double-blind,
randomised, placebo-controlled, parallel-group, multinational,
Diabetologia
multicentre phase 3a trial. Lancet Diabetes Endocrinol 5:251260.
https://doi.org/10.1016/S2213-8587(17)30013-X
112. Pratley RE, Aroda VR, Lingvay I et al (2018) Semaglutide versus
dulaglutide once weekly in patients with type 2 diabetes
(SUSTAIN 7): a randomised, open-label, phase 3b trial. Lancet
Diabetes Endocrinol 6:275286. https://doi.org/10.1016/S2213-
8587(18)30024-X
113. Dungan KM, Povedano ST, Forst T et al (2014) Once-weekly
dulaglutide versus once-daily liraglutide in metformin-treated pa-
tients with type 2 diabetes (AWARD-6): a randomised, open-label,
phase 3, non-inferiority trial. Lancet 384:13491357. https://doi.
org/10.1016/S0140-6736(14)60976-4
114. Buse JB, Nauck M, Forst T et al (2013) Exenatide once weekly
versus liraglutide once daily in patients with type 2 diabetes
(DURATION-6): a randomised, open-label study. Lancet 381:
117124. https://doi.org/10.1016/S0140-6736(12)61267-7
115. Buse JB, Rosenstock J, Sesti G et al (2009) Liraglutide once a day
versus exenatide twice a day for type 2 diabetes: a 26-week
randomised, parallel-group, multinational, open-label trial
(LEAD-6). Lancet 374:3947. https://doi.org/10.1016/S0140-
6736(09)60659-0
116. Drucker DJ, Buse JB, Taylor K et al (2008) Exenatide once week-
ly versus twice daily for the treatment of type 2 diabetes: a
randomised, open-label, non-inferiority study. Lancet 372:1240
1250. https://doi.org/10.1016/S0140-6736(08)61206-4
117. Andreadis P, Karagiannis T, Malandris K et al (2018) Semaglutide
for type 2 diabetes mellitus: a systematic review and meta-analy-
sis. Diabetes Obes Metab 20:22552263. https://doi.org/10.1111/
dom.13361
118. Li Z, Zhang Y, Quan X et al (2016) Efficacy and acceptability of
glycemic control of glucagon-like peptide-1 receptor agonists
among type 2 diabetes: a systematic review and network meta-
analysis. PLoS One 11:e0154206. https://doi.org/10.1371/
journal.pone.0154206
119. Storgaard H, Cold F, Gluud LL et al (2017) Glucagon-like peptide-
1 receptor agonists and risk of acute pancreatitis in patients with
type 2 diabetes. Diabetes Obes Metab 19:906908. https://doi.org/
10.1111/dom.12885
120. Monami M, Nreu B, Scatena A et al (2017) Safety issues with
glucagon-like peptide-1 receptor agonists (pancreatitis, pancreatic
cancer and cholelithiasis): data from randomized controlled trials.
Diabetes Obes Metab 19:12331241. https://doi.org/10.1111/
dom.12926
121. Esposito K, Chiodini P, Maiorino MI et al (2014) Glycaemic du-
rability with dipeptidyl peptidase-4 inhibitors in type 2 diabetes: a
systematic review and meta-analysis of long-term randomised
controlled trials. BMJ Open 4:e005442. https://doi.org/10.1136/
bmjopen-2014-005442
122. Aroda VR, Henry RR, Han J et al (2012) Efficacy of GLP-1
receptor agonists and DPP-4 inhibitors: meta-analysis and system-
atic review. Clin Ther 34:12471258.e22. https://doi.org/10.1016/
j.clinthera.2012.04.013
123. Wu S, Chai S, Yang J et al (2017) Gastrointestinal adverse events
of dipeptidyl peptidase 4 inhibitors in type 2 diabetes: a systematic
review and network meta-analysis. Clin Ther 39:17801789.e33.
https://doi.org/10.1016/j.clinthera.2017.07.036
124. Salvo F, Moore N, Arnaud M et al (2016) Addition of dipeptidyl
peptidase-4 inhibitors to sulphonylureas and risk of
hypoglycaemia: systematic review and meta-analysis. BMJ 353:
i2231. https://doi.org/10.1136/bmj.i2231
125. TkáčI, Raz I (2017) Combined analysis of three large interven-
tional trials with gliptins indicates increased incidence of acute
pancreatitis in patients with type 2 diabetes. Diabetes Care 40:
284286. https://doi.org/10.2337/dc15-1707
126. Mascolo A, Rafaniello C, Sportiello L et al (2016) Dipeptidyl
peptidase (DPP)-4 inhibitor-induced arthritis/arthralgia: a review
of clinical cases. Drug Saf 39:401407. https://doi.org/10.1007/
s40264-016-0399-8
127. Nauck MA, Meier JJ, Cavender MA et al (2017) Cardiovascular
actions and clinical outcomes with glucagon-like peptide-1 recep-
tor agonists and dipeptidyl peptidase-4 inhibitors. Circulation 136:
849870. https://doi.org/10.1161/CIRCULATIONAHA.117.
028136
128. Li L, Li S, Deng K et al (2016) Dipeptidyl peptidase-4 inhibitors
and risk of heart failure in type 2 diabetes: systematic review and
meta-analysis of randomised and observational studies. BMJ 352:
i610
129. Aronoff S, Rosenblatt S, Braithwaite S et al (2000) Pioglitazone
hydrochloride monotherapy improves glycemic control in the
treatment of patients with type 2 diabetes: a 6-month randomized
placebo-controlled dose-response study. The Pioglitazone 001
Study Group. Diabetes Care 23:16051611
130. Einhorn D, Rendell M, Rosenzweig J et al (2000) Pioglitazone
hydrochloride in combination with metformin in the treatment of
type 2 diabetes mellitus: a randomized, placebo-controlled study.
The Pioglitazone 027 Study Group. Clin Ther 22:13951409
131. Yki-Järvinen H (2004) Thiazolidinediones. N Engl J Med 351:
11061118. https://doi.org/10.1056/NEJMra041001
132. Kernan WN, Viscoli CM, Furie KL et al (2016) Pioglitazone after
ischemic stroke or transient ischemic attack. N Engl J Med 374:
13211331. https://doi.org/10.1056/NEJMoa1506930
133. Hanefeld M, Marx N, Pfützner A et al (2007) Anti-inflammatory
effects of pioglitazone and/or simvastatin in high cardiovascular
risk patients with elevated high sensitivity C-reactive protein: the
PIOSTAT Study. J Am Coll Cardiol 49:290297. https://doi.org/
10.1016/j.jacc.2006.08.054
134. Saremi A, Schwenke DC, Buchanan TA et al (2013) Pioglitazone
slows progression of atherosclerosis in prediabetes independent of
changes in cardiovascular risk factors. Arterioscler Thromb Vasc
Biol 33:393399. https://doi.org/10.1161/ATVBAHA.112.
300346
135. Nissen SE, Nicholls SJ, Wolski K et al (2008) Comparison of
pioglitazone vs glimepiride on progression of coronary atheroscle-
rosis in patients with type 2 diabetes: the PERISCOPE random-
ized controlled trial. JAMA 299:15611573. https://doi.org/10.
1001/jama.299.13.1561
136. Dormandy JA, Charbonnel B, Eckland DJA et al (2005)
Secondary prevention of macrovascular events in patients with
type 2 diabetes in the PROactive Study (PROspective
pioglitAzone Clinical Trial In macroVascular Events): a
randomised controlled trial. Lancet 366:12791289. https://doi.
org/10.1016/S0140-6736(05)67528-9
137. Bach RG, Brooks MM, Lombardero M et al (2013) Rosiglitazone
and outcomes for patients with diabetes mellitus and coronary
artery disease in the Bypass Angioplasty Revascularization
Investigation 2 Diabetes (BARI 2D) trial. Circulation 128:785
794. https://doi.org/10.1161/CIRCULATIONAHA.112.000678
138. Mahaffey KW, Hafley G, Dickerson S et al (2013) Results of a
reevaluation of cardiovascular outcomes in the RECORD trial.
Am Heart J 166:240249.e1. https://doi.org/10.1016/j.ahj.2013.
05.004
139. Cusi K, Orsak B, Bril F et al (2016) Long-term pioglitazone treat-
ment for patients with nonalcoholic steatohepatitis and prediabetes
or type 2 diabetes mellitus: a randomized trial. Ann Intern Med
165:305315. https://doi.org/10.7326/M15-1774
140. Kahn SE, Haffner SM, Heise MA et al (2006) Glycemic durability of
rosiglitazone, metformin, or glyburide monotherapy. N Engl J Med
355:24272443. https://doi.org/10.1056/NEJMoa066224
141. Home PD, Pocock SJ, Beck-Nielsen H et al (2009) Rosiglitazone
evaluated for cardiovascular outcomes in oral agent combination
therapy for type 2 diabetes (RECORD): a multicentre,
Diabetologia
randomised, open-label trial. Lancet Lond Engl 373:21252135.
https://doi.org/10.1016/S0140-6736(09)60953-3
142. Hanefeld M, Brunetti P, Schernthaner GH et al (2004) One-year
glycemic control with a sulfonylurea plus pioglitazone versus a
sulfonylurea plus metformin in patients with type 2 diabetes.
Diabetes Care 27:141147. https://doi.org/10.2337/diacare.27.1.
141
143. Viscoli CM, Inzucchi SE, Young LH et al (2017) Pioglitazone and
risk for bone fracture: safety data from a randomized clinical trial.
J Clin Endocrinol Metab 102:914922. https://doi.org/10.1210/jc.
2016-3237
144. Kahn SE, Zinman B, Lachin JM et al (2008) Rosiglitazone-
associated fractures intype 2 diabetes: an analysis from a diabetes
outcome progression trial (ADOPT). Diabetes Care 31:845851.
https://doi.org/10.2337/dc07-2270
145. Lewis JD, Habel LA, Quesenberry CP et al (2015) Pioglitazone
use and risk of bladder cancer and other common cancers in per-
sons with diabetes. JAMA 314:265277. https://doi.org/10.1001/
jama.2015.7996
146. Hirst JA, Farmer AJ, Dyar A et al (2013) Estimating the effect of
sulfonylurea on HbA
1c
in diabetes: a systematic review and meta-
analysis. Diabetologia 56:973984. https://doi.org/10.1007/
s00125-013-2856-6
147. UK Prospective Diabetes Study (UKPDS) Group (1998) Intensive
blood-glucose control with sulphonylureas or insulin compared
with conventional treatment and risk of complications in patients
with type 2 diabetes (UKPDS 33). Lancet 352:837853
148. ADVANCE Collaborative Group, Patel A, MacMahon S et al
(2008) Intensive blood glucose control and vascular outcomes in
patients with type 2 diabetes. N Engl J Med 358:25602572.
https://doi.org/10.1056/NEJMoa0802987
149. Monami M, Dicembrini I, Kundisova L et al (2014) A meta-
analysis of the hypoglycaemic risk in randomized controlled trials
with sulphonylureas in patients with type 2 diabetes. Diabetes
Obes Metab 16:833840. https://doi.org/10.1111/dom.12287
150. Del Prato S, Camisasca R, Wilson C, Fleck P (2014) Durabilityof
the efficacy and safety of alogliptin compared with glipizide in
type 2 diabetes mellitus: a 2-year study. Diabetes Obes Metab
16:12391246. https://doi.org/10.1111/dom.12377
151. Mishriky BM, CummingsDM, Tanenberg RJ (2015) The efficacy
and safety of DPP4 inhibitors compared to sulfonylureas as add-
on therapy to metformin in patients with type 2 diabetes: a sys-
tematic review and meta-analysis. Diabetes Res Clin Pract 109:
378388. https://doi.org/10.1016/j.diabres.2015.05.025
152. Khunti K, Chatterjee S, Gerstein HC et al (2018) Do
sulphonylureas still have a place in clinical practice? Lancet
Diabetes Endocrinol. https://doi.org/10.1016/S2213-8587(18)
30025-1
153. Gangji AS, Cukierman T, Gerstein HC et al (2007) A systematic
review and meta-analysis of hypoglycemia and cardiovascular
events: a comparison of glyburide with other secretagogues and
with insulin. Diabetes Care 30:389394. https://doi.org/10.2337/
dc06-1789
154. Chan SP, Colagiuri S (2015) Systematic review and meta-analysis
of the efficacy and hypoglycemic safety of gliclazide versus other
insulinotropic agents. Diabetes Res Clin Pract 110:7581. https://
doi.org/10.1016/j.diabres.2015.07.002
155. Holman RR, Paul SK, Bethel MA et al (2008) 10-year follow-up
of intensive glucose control in type 2 diabetes. N Engl J Med 359:
15771589. https://doi.org/10.1056/NEJMoa0806470
156. Erpeldinger S, Rehman MB, Berkhout C et al (2016) Efficacy and
safety of insulin in type 2 diabetes: meta-analysis of randomised
controlled trials. BMC Endocr Disord 16:39. https://doi.org/10.
1186/s12902-016-0120-z
157. Owens DR, Traylor L, Mullins P, Landgraf W (2017) Patient-level
meta-analysis of efficacy and hypoglycaemia in people with type 2
diabetes initiating insulin glargine 100U/mL or neutral protamine
Hagedorn insulin analysed according to concomitant oral
antidiabetes therapy. Diabetes Res Clin Pract 124:5765. https://
doi.org/10.1016/j.diabres.2016.10.022
158. Goldman J, Kapitza C, Pettus J, Heise T (2017) Understanding
how pharmacokinetic and pharmacodynamic differences of basal
analog insulins influence clinical practice. Curr Med Res Opin 33:
18211831. https://doi.org/10.1080/03007995.2017.1335192
159. Freemantle N, Chou E, Frois C et al (2016) Safety and efficacy of
insulin glargine 300 u/mL compared with other basal insulin ther-
apies in patients with type 2 diabetes mellitus: a network meta-
analysis. BMJ Open 6:e009421. https://doi.org/10.1136/bmjopen-
2015-009421
160. Russell-Jones D, Gall M-A, Niemeyer M et al (2015) Insulin
degludec results in lower rates of nocturnal hypoglycaemia and
fasting plasma glucose vs. insulin glargine: a meta-analysis of
seven clinical trials. Nutr Metab Cardiovasc Dis 25:898905.
https://doi.org/10.1016/j.numecd.2015.06.005
161. Lipska KJ, Parker MM, Moffet HH et al (2018) Association of
initiation of basal insulin analogs vs neutral protamine hagedorn
insulin with hypoglycemia-related emergency department visits or
hospital admissions and with glycemic control in patients with
type 2 diabetes. JAMA 320:5362. https://doi.org/10.1001/jama.
2018.7993
162. Marso SP, McGuire DK, Zinman B et al (2017) Efficacy and
safety of degludec versus glargine in type 2 diabetes. N Engl J
Med 377:723732. https://doi.org/10.1056/NEJMoa1615692
163. Rosenstock J, Hollander P, Bhargava A et al (2015) Similar effi-
cacy and safety of LY2963016 insulin glargine and insulin
glargine (Lantus®) in patients with type 2 diabetes who were
insulin-naïve or previously treated with insulin glargine: a ran-
domized, double-blind controlled trial (the ELEMENT 2 study).
Diabetes Obes Metab 17:734741. https://doi.org/10.1111/dom.
12482
164. ORIGIN Trial Investigators, Gerstein HC, Bosch J et al (2012)
Basal insulin and cardiovascular and other outcomes in
dysglycemia. N Engl J Med 367:319328. https://doi.org/10.
1056/NEJMoa1203858
165. Riddle MC, Yki-Järvinen H, Bolli GB et al (2015) One-year
sustained glycaemic control and less hypoglycaemia with new
insulin glargine 300 U/mlcompared with 100 U/ml in people with
type 2 diabetes using basal plus meal-timeinsulin: the EDITION 1
12-month randomized trial, including 6-month extension.
Diabetes Obes Metab 17:835842. https://doi.org/10.1111/dom.
12472
166. Yki-Järvinen H, Bergenstal R, Ziemen M et al (2014) New insulin
glargine 300 units/mL versus glargine 100 units/mL in people with
type 2 diabetes using oral agents and basal insulin: glucose control
and hypoglycemia in a 6-month randomized controlled trial
(EDITION 2). Diabetes Care 37:32353243. https://doi.org/10.
2337/dc14-0990
167. Riddle MC, Bolli GB, Ziemen M et al (2014)New insulin glargine
300 units/mL versus glargine 100 units/mL in people with type 2
diabetes using basal and mealtime insulin: glucose control and
hypoglycemia in a 6-month randomized controlled trial
(EDITION 1). Diabetes Care 37:27552762. https://doi.org/10.
2337/dc14-0991
168. Khunti K, Damci T, Husemoen LL et al (2017) Exploring the
characteristics of suboptimally controlled patients after 24 weeks
of basal insulin treatment: an individualized approach to intensifi-
cation. Diabetes Res Clin Pract 123:209217. https://doi.org/10.
1016/j.diabres.2016.11.028
169. Jensen MD, Ryan DH, Apovian CM et al (2014) 2013 AHA/
ACC/TOS guideline for the management of overweight and obe-
sity in adults: a report of the American College of Cardiology/
American Heart Association Task Force on practice guidelines
Diabetologia
and The Obesity Society. J Am Coll Cardiol 63:29853023.
https://doi.org/10.1016/j.jacc.2013.11.004
170. Garvey WT, Mechanick JI, Brett EM et al (2016) American
Association of Clinical Endocrinologists and American College
of endocrinology comprehensive clinical practice guidelines for
medical care of patients with obesity. Endocr Pract 22(Suppl 3):1
203. https://doi.org/10.4158/EP161365.GL
171. Apovian CM, Aronne LJ, Bessesen DH et al (2015)
Pharmacological management of obesity: an Endocrine Society
clinical practice guideline. J Clin Endocrinol Metab 100:342
362. https://doi.org/10.1210/jc.2014-3415
172. Moyer VA, U.S. Preventive Services Task Force (2012) Screening
for and management of obesity in adults: U.S. Preventive Services
Task Force recommendation statement. Ann Intern Med 157:373
378. https://doi.org/10.7326/0003-4819-157-5-201209040-00475
173. Gaal LV, Dirinck E (2016) Pharmacological approaches in the
treatment and maintenance of weight loss. Diabetes Care 39:
S260S267. https://doi.org/10.2337/dcS15-3016
174. Khera R, Pandey A, Chandar AK et al (2018) Effects of weight-
loss medications on cardiometabolic risk profiles: a systematic
review and network meta-analysis. Gastroenterology 154:1309
1319.e7. https://doi.org/10.1053/j.gastro.2017.12.024
175. Davies MJ, Bergenstal R, Bode B et al (2015) Efficacy of
liraglutide for weight loss among patients with type 2 diabetes:
the SCALE diabetes randomized clinical trial. JAMA 314:687
699. https://doi.org/10.1001/jama.2015.9676
176. Schauer PR, Bhatt DL, Kirwan JP et al (2014) Bariatric surgery
versus intensive medical therapy for diabetes3-year outcomes.
N Engl J Med 370:20022013. https://doi.org/10.1056/
NEJMoa1401329
177. Schauer PR, Bhatt DL, Kirwan JP et al (2017) Bariatric surgery
versus intensive medical therapy for diabetes5-year outcomes.
N Engl J Med 376:641651. https://doi.org/10.1056/
NEJMoa1600869
178. Ikramuddin S, Korner J, Lee W-J et al (2018) Lifestyle interven-
tion and medical management with vs without roux-en-y gastric
bypass and control of hemoglobin A1c, LDL cholesterol, and
systolic blood pressure at 5 years in the diabetes surgery study.
JAMA 319:266278. https://doi.org/10.1001/jama.2017.20813
179. Dixon JB, OBrien PE, Playfair J et al (2008) Adjustable gastric
banding and conventional therapy for type 2 diabetes: a random-
ized controlled trial. JAMA 299:316323.https://doi.org/10.1001/
jama.299.3.316
180. Mingrone G, Panunzi S, De Gaetano A et al (2012) Bariatric
surgery versus conventional medical therapy for type 2 diabetes.
N Engl J Med 366:15771585. https://doi.org/10.1056/
NEJMoa1200111
181. Sjöström L, Peltonen M, Jacobson P et al (2014) Association of
bariatric surgery with long-term remission of type 2 diabetes and
with microvascular and macrovascular complications. JAMA 311:
22972304. https://doi.org/10.1001/jama.2014.5988
182. Mingrone G, Panunzi S, De Gaetano A et al (2015) Bariatric-
metabolic surgery versus conventional medical treatment in obese
patients with type 2 diabetes: 5 year follow-up of an open-label,
single-centre, randomised controlled trial. Lancet 386:964973.
https://doi.org/10.1016/S0140-6736(15)00075-6
183. Rubino F, Nathan DM, Eckel RH etal (2016) Metabolic surgery in
the treatment algorithm for type 2 diabetes: a joint statement by
international diabetes organizations. Diabetes Care 39:861877.
https://doi.org/10.2337/dc16-0236
184. Maggard-Gibbons M, Maglione M, Livhits M et al (2013)
Bariatric surgery for weight loss and glycemic control in
nonmorbidly obese adults with diabetes: a systematic review.
JAMA 309:22502261. https://doi.org/10.1001/jama.2013.4851
185. Mingrone G, Bornstein S, Le Roux CW (2018) Optimisation of
follow-up after metabolic surgery. Lancet Diabetes Endocrinol 6:
487499. https://doi.org/10.1016/S2213-8587(17)30434-5
186. Conason A, Teixeira J, Hsu C-H et al (2013) Substance use fol-
lowing bariatric weight loss surgery. JAMA Surg 148:145150.
https://doi.org/10.1001/2013.jamasurg.265
187. Dawes AJ, Maggard-Gibbons M, Maher AR et al (2016) Mental
health conditions among patients seeking and undergoing bariatric
surgery: a meta-analysis. JAMA 315:150163. https://doi.org/10.
1001/jama.2015.18118
188. Mechanick JI, Kushner RF, Sugerman HJ, et al (2009) American
Association of Clinical Endocrinologists, The Obesity Society,
and American Society for Metabolic & Bariatric Surgery medical
guidelines for clinical practice for the perioperative nutritional,
metabolic, and nonsurgical support of the bariatric surgery patient.
Obesity (Silver Spring) 17 Suppl 1:S1-S70, v. https://doi.org/10.
1038/oby.2009.28
189. Mechanick JI, Youdim A, Jones DB et al (2013) Clinical practice
guidelines for the perioperative nutritional, metabolic, and nonsur-
gical support of the bariatric surgery patient2013 update:
cosponsored by American Association of Clinical
Endocrinologists, The Obesity Society, and American Society
for Metabolic & Bariatric Surgery. Obesity (Silver Spring)
21(Suppl 1):S1S27. https://doi.org/10.1002/oby.20461
190. Phung OJ, Sobieraj DM, Engel SS, Rajpathak SN (2014) Early
combination therapy for the treatment of type 2 diabetes mellitus:
systematic review and meta-analysis. Diabetes Obes Metab 16:
410417. https://doi.org/10.1111/dom.12233
191. Abdul-Ghani MA, Puckett C, Triplitt C et al (2015) Initial combi-
nation therapy with metformin, pioglitazone and exenatide is more
effective than sequential add-on therapy in subjects with new-
onset diabetes. Results from the Efficacy and Durability of Initial
Combination Therapy for type 2 diabetes (EDICT): a randomized
trial. Diabetes Obes Metab 17:268275. https://doi.org/10.1111/
dom.12417
192. Vijayakumar TM, Jayram J, Meghana Cheekireddy Vet al (2017)
Safety, efficacy, and bioavailability of fixed-dose combinations in
type 2 diabetes mellitus: a systematic updated review. Curr Ther
Res Clin Exp 84:49. https://doi.org/10.1016/j.curtheres.2017.01.
005
193. Mearns ES, Saulsberry WJ, White CM et al (2015) Efficacy and
safety of antihyperglycaemic drug regimens added to metformin
and sulphonylurea therapy in type 2 diabetes: a network meta-
analysis. Diabet Med 32:15301540. https://doi.org/10.1111/
dme.12837
194. Henry RR, Murray AV, Marmolejo MH et al (2012)Dapagliflozin,
metformin XR, or both: initial pharmacotherapy for type 2 diabe-
tes, a randomised controlled trial. Int J Clin Pract 66:446456.
https://doi.org/10.1111/j.1742-1241.2012.02911.x
195. Buse JB, Peters A, Russell-Jones D et al (2015) Is insulin the most
effective injectable antihyperglycaemic therapy? Diabetes Obes
Metab 17:145151. https://doi.org/10.1111/dom.12402
196. Vaccaro O, Masulli M, Nicolucci A et al (2017) Effects on the
incidence of cardiovascular events of the addition of pioglitazone
versus sulfonylureas in patients with type 2 diabetes inadequately
controlled with metformin (TOSCA.IT): a randomised,
multicentre trial. Lancet Diabetes Endocrinol 5:887897. https://
doi.org/10.1016/S2213-8587(17)30317-0
197. Wang Z, Sun J, Han R et al (2018) Efficacy and safety of sodium-
glucose cotransporter-2 inhibitors versus dipeptidyl peptidase-4
inhibitors as monotherapy or add-on to metformin in patients with
type 2 diabetes mellitus: a systematic review and meta-analysis.
Diabetes Obes Metab 20:113120. https://doi.org/10.1111/dom.
13047
198. Kawalec P, Mikrut A, Łopuch S (2014) The safety of dipeptidyl
peptidase-4 (DPP-4) inhibitors or sodium-glucose cotransporter 2
Diabetologia
(SGLT-2) inhibitors added to metformin background therapy in
patients with type 2 diabetes mellitus: a systematic review and
meta-analysis. Diabetes Metab Res Rev 30:269283
199. Leiter LA, Yoon K-H, Arias P et al (2015) Canagliflozin provides
durable glycemic improvements and body weight reduction over
104 weeks versus glimepiride in patients with type 2 diabetes on
metformin: a randomized, double-blind, phase 3 study. Diabetes
Care 38:355364. https://doi.org/10.2337/dc13-2762
200. Del Prato S, Nauck M, Durán-Garcia S et al (2015) Long-term
glycaemic response and tolerability of dapagliflozin versus a
sulphonylurea as add-on therapy to metformin in patients with
type 2 diabetes: 4-year data. Diabetes Obes Metab 17:581590.
https://doi.org/10.1111/dom.12459
201. Ridderstråle M, Andersen KR, Zeller C et al (2014) Comparison
of empagliflozin and glimepiride as add-on to metformin in pa-
tients with type 2 diabetes: a 104-week randomised, active-con-
trolled, double-blind, phase 3 trial. Lancet Diabetes Endocrinol 2:
691700. https://doi.org/10.1016/S2213-8587(14)70120-2
202. Zhang Y, Hong J, Chi J et al (2014) Head-to-head comparison of
dipeptidyl peptidase-IV inhibitors and sulfonylureasa meta-
analysis from randomized clinical trials. Diabetes Metab Res
Rev 30:241256. https://doi.org/10.1002/dmrr.2482
203. Foroutan N, Muratov S, Levine M (2016) Safety and efficacy of
dipeptidyl peptidase-4 inhibitors vs sulfonylurea in metformin-
based combination therapy for type 2 diabetes mellitus: systematic
review and meta-analysis. Clin Invest Med Med Clin Exp 39:E48
E62
204. Chen K, Kang D, Yu M et al (2018) Direct head-to-head compar-
ison of glycaemic durability of dipeptidyl peptidase-4 inhibitors
and sulphonylureas in patients with type 2 diabetes mellitus: a
meta-analysis of long-term randomized controlled trials.
Diabetes Obes Metab 20:10291033. https://doi.org/10.1111/
dom.13147
205. Zaccardi F, Dhalwani NN, Dales J et al (2018) Comparison of
glucose-lowering agents after dual therapy failure in type 2 diabe-
tes: a systematic review and network meta-analysis of randomized
controlled trials. Diabetes Obes Metab 20:985997. https://doi.
org/10.1111/dom.13185
206. Downes MJ, Bettington EK, Gunton JE, Turkstra E (2015) Triple
therapy in type 2 diabetes; a systematic reviewand network meta-
analysis. PeerJ 3:e1461. https://doi.org/10.7717/peerj.1461
207. Lee CMY, Woodward M, Colagiuri S (2016) Triple therapy com-
binations for the treatment of type 2 diabetesa network meta-
analysis.Diabetes Res Clin Pract 116:149158. https://doi.org/10.
1016/j.diabres.2016.04.037
208. Lukashevich V, Del Prato S, Araga M, Kothny W (2014) Efficacy
and safety of vildagliptin in patients with type 2 diabetes mellitus
inadequately controlled with dual combination of metformin and
sulphonylurea. Diabetes Obes Metab 16:403409. https://doi.org/
10.1111/dom.12229
209. Hong AR, Lee J, Ku EJ et al (2015) Comparison of vildagliptin as
an add-on therapy and sulfonylurea dose-increasing therapy in
patients with inadequately controlled type 2 diabetes using met-
formin and sulfonylurea (VISUAL study): a randomized trial.
Diabetes Res Clin Pract 109:141148. https://doi.org/10.1016/j.
diabres.2015.04.019
210. Moses RG, Kalra S, Brook D et al (2014) A randomized con-
trolled trial of the efficacy and safety of saxagliptin as add-on
therapy in patients with type 2 diabetes and inadequate glycaemic
control on metformin plus a sulphonylurea. Diabetes Obes Metab
16:443450. https://doi.org/10.1111/dom.12234
211. Moses RG, Round E, Shentu Yet al (2016) A randomized clinical
trial evaluating the safety and efficacy of sitagliptin added to the
combination of sulfonylurea and metformin in patients with type2
diabetes mellitus and inadequate glycemic control. J Diabetes 8:
701711. https://doi.org/10.1111/1753-0407.12351
212. Singh S, Wright EE, Kwan AYM et al (2017) Glucagon-like pep-
tide-1 receptor agonists compared with basal insulins for the treat-
ment of type 2 diabetes mellitus: a systematic review and meta-
analysis. Diabetes Obes Metab 19:228238. https://doi.org/10.
1111/dom.12805
213. Levin PA, Nguyen H, Wittbrodt ET, Kim SC (2017) Glucagon-
like peptide-1 receptor agonists: a systematic review of compara-
tive effectiveness research. Diabetes Metab Syndr Obes Targets
Ther 10:123139. https://doi.org/10.2147/DMSO.S130834
214. Abd El Aziz MS, Kahle M, Meier JJ, Nauck MA (2017) A meta-
analysis comparing clinical effects of short- or long-acting GLP-1
receptor agonists versus insulin treatment from head-to-head stud-
ies in type 2 diabetic patients. Diabetes Obes Metab 19:216227.
https://doi.org/10.1111/dom.12804
215. Eng C, Kramer CK, Zinman B, Retnakaran R (2014) Glucagon-
like peptide-1 receptor agonist and basal insulin combination treat-
ment for the management of type 2 diabetes: a systematic review
and meta-analysis. Lancet 384:22282234. https://doi.org/10.
1016/S0140-6736(14)61335-0
216. Aroda VR, Bailey TS, Cariou B et al (2016) Effect of adding
insulin degludec to treatment in patients with type 2 diabetes in-
adequately controlled with metformin and liraglutide: a double-
blind randomized controlled trial (BEGIN: ADD TO GLP-1
Study). Diabetes Obes Metab 18:663670. https://doi.org/10.
1111/dom.12661
217. Aschner P, Sethi B, Gomez-Peralta F et al (2015) Insulin glargine
compared with premixed insulin for management of insulin-naïve
type 2 diabetes patients uncontrolled onoral antidiabetic drugs: the
open-label, randomized GALAPAGOS study. J Diabetes
Complicat 29:838845. https://doi.org/10.1016/j.jdiacomp.2015.
04.003
218. Rosenstock J, Fonseca V, Schinzel S et al (2014) Reduced risk of
hypoglycemia with once-daily glargine versus twice-daily NPH
and number needed to harm with NPH to demonstrate the risk of
one additional hypoglycemic event in type 2 diabetes: evidence
from a long-term controlled trial. J Diabetes Complicat 28:742
749. https://doi.org/10.1016/j.jdiacomp.2014.04.003
219. Tang H, Cui W, Li D et al (2017) Sodium-glucose co-transporter 2
inhibitors in addition to insulin therapy for management of type 2
diabetes mellitus: a meta-analysis of randomized controlled trials.
Diabetes Obes Metab 19:142147. https://doi.org/10.1111/dom.
12785
220. Rosenstock J, Jelaska A, Zeller C et al (2015) Impact of
empagliflozin added on to basal insulin in type 2 diabetes inade-
quately controlled on basal insulin: a 78-week randomized, dou-
ble-blind, placebo-controlled trial. Diabetes Obes Metab 17:936
948. https://doi.org/10.1111/dom.12503
221. Rosenstock J, Jelaska A, Frappin G et al (2014) Improved glucose
control with weight loss, lower insulin doses, and no increased
hypoglycemia with empagliflozin added to titrated multiple daily
injections of insulin in obese inadequately controlled type 2 dia-
betes. Diabetes Care 37:18151823. https://doi.org/10.2337/dc13-
3055
222. Yang W, Cai X, Gao X et al (2017) Addition of dipeptidyl
peptidase-4 inhibitors to insulin treatment in type 2 diabetes pa-
tients: a meta-analysis. J Diabetes Investig 9:813821. https://doi.
org/10.1111/jdi.12764
223. Min SH, Yoon J-H, Hahn S, Cho YM (2017) Comparison between
SGLT2 inhibitors and DPP4 inhibitors added to insulin therapy in
type 2 diabetes: a systematic review with indirect comparison
meta-analysis: SGLT2 or DPP4 inhibitor with insulin. Diabetes
Metab Res Rev 33:e2818. https://doi.org/10.1002/dmrr.2818
224. Wysham CH, Lin J, Kuritzky L (2017) Safety and efficacy of a
glucagon-like peptide-1 receptor agonist added to basal insulin
therapy versus basal insulin with or without a rapid-acting insulin
in patients with type 2 diabetes: results of a meta-analysis.
Diabetologia
Postgrad Med 129:436445. https://doi.org/10.1080/00325481.
2017.1297669
225. Maiorino MI, Chiodini P, Bellastella G et al (2017) Insulin and
glucagon-like peptide 1 receptor agonist combination therapy in
type 2 diabetes: a systematic review and meta-analysis of random-
ized controlled trials. Diabetes Care 40:614624. https://doi.org/
10.2337/dc16-1957
226. Gough SCL, Bode BW, Woo VC et al (2015) One-year efficacy
and safety of a fixed combination of insulin degludec and
liraglutide in patients with type 2 diabetes: results of a 26-week
extension to a 26-week main trial. Diabetes Obes Metab 17:965
973. https://doi.org/10.1111/dom.12498
227. Rosenstock J, Aronson R, Grunberger G et al (2016) Benefits of
LixiLan, a titratable fixed-ratio combination of insulin glargine
plus lixisenatide, versus insulin glargine and lixisenatide
monocomponents in type 2 diabetes inadequately controlled on
oral agents: the LixiLan-O randomized trial. Diabetes Care 39:
20262035. https://doi.org/10.2337/dc16-0917
228. Aroda VR, Rosenstock J, Wysham C et al (2016) Efficacy and
safety of LixiLan, a titratable fixed-ratio combination of insulin
glargine plus lixisenatide in type 2 diabetes inadequately con-
trolled on basal insulin and metformin: the LixiLan-L randomized
trial. Diabetes Care 39:19721980. https://doi.org/10.2337/dc16-
1495
229. Garber AJ, Wahlen J, Wahl T et al (2006) Attainment of
glycaemic goals in type 2 diabetes with once-, twice-, or
thrice-daily dosing with biphasic insulin aspart 70/30 (the 1-
2-3 study). Diabetes Obes Metab 8:5866. https://doi.org/10.
1111/j.1463-1326.2005.00563.x
230. Linjawi S, Lee B-W, Tabak Ö et al (2018) A 32-week randomized
comparison of stepwise insulin intensification of biphasic insulin
aspart (BIAsp 30) versus basal-bolus therapy in insulin-naïve pa-
tients with type 2 diabetes. Diabetes Ther 9:111. https://doi.org/
10.1007/s13300-017-0334-8
231. Raccah D, Huet D, Dib A et al (2017) Review of basal-plus insulin
regimen options for simpler insulin intensification in people with
type 2 diabetes mellitus. Diabet Med J Br Diabet Assoc 34:1193
1204. https://doi.org/10.1111/dme.13390
232. Rodbard HW, Visco VE, Andersen H et al (2014) Treatment in-
tensification with stepwise addition of prandial insulin aspart
boluses compared with full basal-bolus therapy (FullSTEP
Study): a randomised, treat-to-target clinical trial. Lancet
Diabetes Endocrinol 2:3037. https://doi.org/10.1016/S2213-
8587(13)70090-1
233. Giugliano D, Chiodini P, Maiorino MI et al (2016) Intensification
of insulin therapy with basal-bolus or premixed insulin regimens
in type 2 diabetes: a systematic review and meta-analysis of ran-
domized controlled trials. Endocrine 51:417428. https://doi.org/
10.1007/s12020-015-0718-3
234. Wang C, Mamza J, Idris I (2015) Biphasic vs basal bolus insulin
regimen in type 2 diabetes: a systematic review and meta-analysis
of randomized controlled trials. Diabet Med 32:585594. https://
doi.org/10.1111/dme.12694
235. Anyanwagu U, Mamza J, Gordon J et al (2017) Premixed vs
basal-bolus insulin regimen in type 2 diabetes: comparison of
clinical outcomes from randomized controlled trials and real-
world data. Diabet Med 34:17281736. https://doi.org/10.1111/
dme.13518
236. Aronson R, Reznik Y, Conget I et al (2016) Sustained efficacy of
insulin pump therapy compared with multiple daily injections in
type 2 diabetes: 12-month data from the OpT2mise randomized
trial. Diabetes Obes Metab 18:500507. https://doi.org/10.1111/
dom.12642
237. Kelly S, Martin S, Kuhn I et al (2016) Barriers and facilitators to
the uptake and maintenance of healthy behaviours by people at
mid-life: a rapid systematic review. PLoS One 11:e0145074.
https://doi.org/10.1371/journal.pone.0145074
238. Gee PM, Greenwood DA, Paterniti DA et al (2015) The eHealth
enhanced chronic care model: a theory derivation approach. J Med
Internet Res 17:e86. https://doi.org/10.2196/jmir.4067
239. Faruque LI, Wiebe N, Ehteshami-Afshar A et al (2017) Effect of
telemedicine on glycated hemoglobin in diabetes: a systematic
review and meta-analysis of randomized trials. CMAJ 189:
E341E364. https://doi.org/10.1503/cmaj.150885
240. Lee SWH, Chan CKY, Chua SS, Chaiyakunapruk N (2017)
Comparative effectiveness of telemedicine strategies on type 2
diabetes management: a systematic review and network meta-
analysis. Sci Rep 7:12680. https://doi.org/10.1038/s41598-017-
12987-z
Affiliations
Melanie J. Davies
1,2
&David A. DAlessio
3
&Judith Fradkin
4
&Walter N. Kernan
5
&Chantal Mathieu
6
&
Geltrude Mingrone
7,8
&Peter Rossing
9,10
&Apostolos Tsapas
11
&Deborah J. Wexler
12,13
&John B. Buse
14
1
Diabetes Research Centre, University of Leicester, Leicester, UK
2
Leicester Diabetes Centre, Leicester General Hospital,
Leicester, LE5 4PW, UK
3
Department of Medicine, Duke University School of Medicine,
Durham, NC, USA
4
National Institute of Diabetes and Digestive and Kidney Diseases,
National Institutes of Health, Bethesda, MD, USA
5
Departmentof Medicine, Yale School of Medicine, NewHaven, CT,
USA
6
Clinical and Experimental Endocrinology, UZ Gasthuisberg, KU
Leuven, Leuven, Belgium
7
Department of Internal Medicine, Catholic University, Rome, Italy
8
Diabetes and Nutritional Sciences, Kings College London,
London, UK
9
Steno Diabetes Center Copenhagen, Gentofte, Denmark
10
University of Copenhagen, Copenhagen, Denmark
11
Second Medical Department, Aristotle University Thessaloniki,
Thessaloniki, Greece
12
Department of Medicine and Diabetes Unit, Massachusetts General
Hospital, Boston, MA, USA
13
Harvard Medical School, Boston, MA, USA
14
Department of Medicine, University of North Carolina School of
Medicine, Chapel Hill, NC, USA
Diabetologia
... Lagana [11], no entanto, observaram que 44,9% dos participantes da pesquisa realizada em Curitiba/PR implementaram modificações nas atividades físicas após o diagnóstico do DM. A proposição é preocupante, uma vez que a realização de atividades físicas em conjunto com a modificação da dieta alimentar constitui a principal medida não farmacológicas para o controle e/ou prevenção do diabetes [2,20]. ...
... A Socieidade Brasileira de Diabetes (SBD) [2] esclarece que a educação voltada para a autogestão do DM abrange o processo de facilitação de conhecimentos, de habilidades e capacidades necessárias para o autocuidado da doença. Dentre os principais processos e resultados avaliados na autogestão do DM estão os psicossociais e comportamentais, que consideram conhecimento e crenças sobre o DM, satisfação no tratamento, qualidade de vida, realização de exercícios físicos e administração da dieta [23,20]. ...
Article
Objetivo Avaliar informações sobre a qualidade de vida de indivíduos portadores de diabetes Mellitus moradores da cidade de Campina Grande, Paraíba, e usuários do Sistema Único de Saúde. Métodos Estudo transversal de caráter descritivo e exploratório com abordagem quantitativa que usou o questionário Diabetes Quality of Life Measure Brasil-8. Resultados Dos 50 participantes voluntários, a maioria encontrava-se com mais de 65 anos, sendo 34% do total. No tocante aos escores do questionário Diabetes Quality of Life Measure Brasil-8, os domínios “Satisfação” e “Impacto” apresentaram valores próximos a 03 (3,02 e 3,11, respectivamente). O domínio “Preocupações sociais vocacionais” apresentou valor final de 1,12 (próximo a 01) e o domínio “Preocupações relacionadas ao diabetes” apresentou valor final de 3,77. Discussão A interpretação dos resultados indica uma inquietação quanto às complicações do diabetes Mellitus, sendo esse o principal fator de impacto na qualidade de vida dos participantes. Os domínios “Satisfação” e “Impacto” tiveram resultados medianos, que podem indicar desatenção nesses aspectos e possível agravamento das condições, posto que abrangem fatores interligados às complicações. ConclusãoConhecer e elucidar os aspectos que impactam a qualidade de vida dos pacientes com diabetes Mellituscontribuem para a melhoria da adesão ao tratamento.
... За последние несколько десятилетий во многих опубликованных систематических обзорах и метаанализах были обобщены данные о взаимосвязи пищевого поведения, качества рациона и заболеваемости СД2 [6]. Кроме того, результаты более ранних публикаций подтверждают, что модификация образа жизни и коррекция питания улучшают гликемический контроль, а также могут предотвратить развитие СД2 [7]. Именно поэтому продолжаются исследования, изучающие влияние различных систем питания с целью достижения ремиссии заболевания. ...
Article
Last time there are a lot of date, published systematic reviews and meta-analyses about relationship between eating behavior and incidence type 2 diabetes. It is known that more than 80% of cases of type 2 diabetes are associated with obesity. In this regard, the nutritional factor is of particular importance in the formation of treatment goals for patients with type 2 diabetes. Today, the disease is no longer a sentence, because achieving remission of type 2 diabetes is possible with the help of nutritional correction. The results of studies shown that a significant decrease in body weight is a predictor of remission of type 2 diabetes. However, the question of the best dietary recommendations for patients with type 2 diabetes remains open. Various dietary patterns (Mediterranean, paleo diet, DASH system, etc.) contribute to the improvement of glycemia, however, there is not enough data indicating a regression of symptoms of type 2 diabetes. In all likelihood, achieving remission is possible only with a significant reduction in daily caloric intake. The ketogenic diet, especially popular in recent times, also improves glycemic control. At the same time, according to the results of a meta-analysis, carbohydrates from whole grains and cereal fibers should not be neglected, because this category of products helps reduce the risk of developing diabetes.
... Such organ protection is also seen in people who do not have T2DM. Based on these criteria, all individuals with diabetes and established or subclinical CVD should be prescribed a drug with a demonstrated cardiovascular benefit from the glucagon-like peptide-1 receptor agonist (GLP-1 RA) or the SGLT2 inhibitor class, independent of HbA1c level or the presence of additional glucose-lowering medications [37]. Also, all people with diabetes and CKD (eGFR 60 mL/min per 1.73 m 2 or Urinary Albumin Creatinine Ration >3.0 mg/mmol/ >30 mg/g) should be given a medication from the SGLT2 inhibitor class that has a documented renal benefit. ...
Article
Full-text available
To cite this article: Popoviciu MS, Păduraru L, Rahman MM, Supti FA, Stoica RA, Reurean-Pintilei D, Bica CI, Cavalu S. The effects of SGLT2 inhibitors in patients with metabolic dysfunction-associated fatty liver disease; a narrative review.
... A Study have reported that shift in cardiac sympathovagal balance in patients with diabetes from parasympathetic to sympathetic can lead to increased cardiovascular morbidity and mortality [28]. Therefore, the American Diabetes Association and the European Association for the Study of Diabetes have emphasized the importance of CVD risk factor management in adults with diabetes [29]. The link between low-grade inflammation and early alterations in CAN in patients with T1DM and its clinical implications have reported previously [4,6,14]. ...
Article
Full-text available
Background Type 1 diabetes mellitus (T1DM) is an autoimmune disease characterized by the chronic inflammation and cause of endothelial dysfunction (ED). Heart rate variability (HRV) is a marker of sympathetic and parasympathetic autonomic nervous system dysfunction. We investigated the association of lipid profile, inflammatory biomarkers, endothelial dysfunction, and heart rate variability in adolescents with T1DM among UAE population. Method In this case-control study we recruited 126 adolescents (13–22 years) from Abu Dhabi, UAE (United Arab Emirates). Demographic, anthropometric, blood and urine samples were collected after an overnight fasting. HRV measurements were determined per Task Force recommendations. Independent t-test or Mann-Whitney U test and Pearson's Chi-squared test were used to compare groups. Adjusted conditional logistic regression model was used to identify the determinants independently associated with T1DM. Results The mean ages in control (n = 47) and patient (n = 79) groups were 17.5 ± 4.6 and 18.6 ± 4.8 years, respectively. A family history of diabetes and waist and hip circumferences significantly differed between the groups (p = 0.030 and 0.010). The patients with T1DM exhibited significantly higher levels of atherogenic markers than control. Endothelial dysfunction biomarkers such as levels of sICAM-1 (p < 0.001), adiponectin (p < 0.001) and 25-hydroxyvitamin D (p < 0.001) were significantly different in the control group compared with those in the T1DM group. There was a significant difference in SDNN intervals, NN50, pNN50, and SD1/SD2 among the two groups. In adjusted analysis, total cholesterol (adjusted Odds Ratio (aOR): 2.78, 95 % CI:1.37–5.64; p = 0.005), LDL (2.66, 95%CI:1.19–5.92; p = 0.017), and triglycerides (5.51, 95%CI:1.57–19.41; p = 0.008) were significantly associated with developing T1DM. The HRV indicators were significantly associated with decrease odds of T1DM after controlling for SBP, BMI, and family history of DM. Conclusion In this study, adolescents with T1DM showed a significant association with lipid profile, ED, and HRV compared with controls. Thus, an early attention to diabetes control is required to reduce the risk of cardiac autonomic neuropathy leading to various cardiovascular diseases.
Article
Aim To assess the differential association of risk factors with severe and non‐severe hypoglycaemia. Materials and Methods The Hypoglycaemia Assessment Tool study evaluated the risk of hypoglycaemia over a 4‐week period in patients with type 1 diabetes (T1D) and type 2 diabetes (T2D) on insulin in 24 countries. Negative binomial regressions were applied to examine the associations of several risk factors with severe and non‐severe hypoglycaemia. Results The median age was 41 years in 5949 patients with T1D and 62 years in 12 914 patients with T2D. The 4‐week rates of non‐severe hypoglycaemic were 5.57 and 1.40 episodes per person in T1D and T2D, respectively; the corresponding rates for severe hypoglycaemia were 0.94 and 0.30. The excess risk was 42% higher for severe than non‐severe hypoglycaemia in females versus males with T2D; 27% higher in patients with T2D with versus without a continuous glucose monitoring (CGM); and 47% lower in patients with T1D with versus without an insulin pump. The excess risk also differed across geographical areas and was marginally lower for severe than non‐severe hypoglycaemia for higher values of HbA1c in patients with T2D. Associations with severity of hypoglycaemia were not different for age, diabetes and insulin therapy duration, previous hypoglycaemic episodes and insulin regimen. Conclusions The risk of severe versus non‐severe hypoglycaemia differs in patients with T1D and T2D; sex, the use of a CGM and insulin pump, and geographical areas were differently associated with one type of hypoglycaemia than the other.
Article
Full-text available
Randomized controlled trials (RCTs) show a reduction in acute kidney injury, renal impairment and acute renal failure after initiation of a sodium glucose cotransporter‐2 inhibitor. Observational literature on the association is conflicting, but important to understand for populations with a higher risk of medication‐related adverse renal events. We aimed to systematically review the literature to summarize the association between sodium glucose cotransporter‐2 inhibitor use and acute kidney injury, renal impairment and acute renal failure in three at‐risk groups: older people aged >65 years, people with heart failure and people with reduced renal function. A systematic search of Embase (1974 until 23 February 2024) and PubMed (1946 until 23 February 2024) was performed. RCTs were included if they reported numbers of acute kidney injury or acute renal failure in people using sodium glucose cotransporter‐2 inhibitors compared to other diabetic therapies. Studies needed to report results by level of renal function, heart failure status or age. Of 922 results, eight studies were included. The absolute risk of acute kidney injury or acute renal failure was higher in people >65 years compared to those <65 years, higher in people with heart failure (vs without) and higher in people with reduced kidney function (vs preserved kidney function), but insufficient evidence to determine if the relative effect of sodium glucose cotransporter‐2 inhibitors on this risk was similar for each group. At‐risk cohorts are associated with a higher incidence of acute kidney problems in users of sodium glucose cotransporter‐2 inhibitors.
Article
Full-text available
Introduction The purpose of this study was to evaluate whether bone mineral density (BMD) ≥ −2.5 SD could be used as the treat-to-target (T2T) goal when treating osteoporosis with teriparatide (TPTD) and alendronate (ALN), and to investigate the relationship with incident vertebral fracture by re-analyzing data from a randomized, controlled trial (JOINT-05) involving postmenopausal Japanese women at high fracture risk. Materials and methods Participants received sequential therapy with once-weekly TPTD for 72 weeks, followed by ALN for 48 weeks (TPTD-ALN group) or ALN monotherapy for 120 weeks (ALN group). BMDs were measured at the lumbar spine (L2-4), total hip, and femoral neck at 0, 24, 48, 72, and 120 weeks by dual-energy X-ray absorptiometry. The T2T goal was BMD ≥ −2.5 SD, and the endpoint was the proportion of participants with baseline BMD < −2.5 SD in three measurement sites achieving BMD ≥ −2.5 SD. Results A total of 559 participants were selected. BMD ≥ −2.5 SD at 120 weeks in the L2-4, total hip, and femoral neck sites was achieved in 20.5%, 23.1%, and 5.9%, respectively, in the TPTD-ALN group and 22.2%, 11.7%, and 7.3%, respectively, in the ALN group. Incident vertebral fractures occurred in areas of both lower and high BMD. Conclusion During the 1.5-year treatment period, more than 20% of participants achieved BMD ≥ −2.5 SD as a T2T goal at L2-4. Since the achievement level differed depending on the BMD measurement site, the appropriate site should be selected according to the baseline BMD level.
Article
Full-text available
Objectives To assess the efficacy and tolerability of iGlarLixi—a novel, fixed-ratio, soluble combination of insulin glargine and lixisenatide—for the treatment of type 2 diabetes (T2D). Methods The PubMed, Embase, Cochrane Library and ClinicalTrials.gov databases were searched from inception to November 15, 2023 to identify randomized controlled trials (RCTs) comparing iGlarLixi with a placebo or any other antidiabetic agent in adults with T2D. Risk ratios (RRs) and mean differences (MDs) with 95% confidence intervals (CIs) were calculated to evaluate the outcomes. Results A total of 10 trials enrolling 6071 T2D patients were included. Compared with placebos or other antidiabetic agents, iGlarLixi exerted beneficial effects on changes in HbA1c, the percentage of patients who achieved an HbA1c < 7%, the percentage of patients who achieved an HbA1c < 6.5%, the percentage of patients who achieved an HbA1c < 7.0% without weight gain and/or without severe or blood glucose-confirmed hypoglycemic episodes, changes in fasting plasma glucose, and changes in self-measured plasma glucose. Regarding safety, iGlarLixi did not increase the incidence of severe hypoglycemia or serious adverse events but did increase the incidence of gastrointestinal adverse events, symptomatic hypoglycemia, and adverse events (e.g., nausea, vomiting, diarrhea). Conclusions iGlarLixi showed improved efficacy and safety in patients with T2D. Additional large, multicenter RCTs are warranted to obtain deeper insights into the efficacy and safety of iGlarLixi, thereby providing guidance for clinical treatment decisions.
Article
ZUSAMMENFASSUNG In der vorliegenden Auswertung aus dem DPV-Register wird der Einsatz von unterschiedlichen Antidiabetika zwischen den Jahren 2005 bis 2021 ausgewertet und analysiert. Es konnten Daten von insgesamt 405 790 Menschen mit Diabetes ausgewertet werden (2 464 078 Beobachtungsjahre). Die Population (53,4 % männlich) war im Median 70,91 Jahre alt bei einer medianen Diabetesdauer von ca. 9 Jahren. Im Laufe des Auswertungszeitraumes nahm der Anteil der über 80-Jährigen kontinuierlich zu. Seitens der Therapie nahm der Anteil der lediglich mit Insulin behandelten Patienten ebenso ab wie der Anteil der lediglich mit Lebensstil behandelten Patienten. Metformin wurde tendenziell zunehmend häufiger eingesetzt. Für Sulfonlyharnstoffe zeigte sich eine komplette gegensätzliche Entwicklung gegenüber den DPP-IV-Inhibitoren mit einer Zunahme dieser seit 2007 etablierten Substanzgruppe. Seit ungefähr 2015 zeigen sich deutlich zunehmende Verordnungshäufigkeiten für SGLT-2-Inhibitoren sowie GLP-1-Rezeptoragonisten bei gleichzeitig zu erkennendem allmählichem Rückgang der DPP-IV-Inhibitorverordnungen. Während der Anteil der Metformin-Verordnungen im Beobachtungszeitraum in allen Altersgruppen kontinuierlich und weitgehend parallel anstieg, sind für SGLT-2-Inhibitoren und GLP-1-Rezeptoragonisten anfänglich bei der Gruppe der unter 60-jährigen und erst zeitversetzt auch der älteren Patienten eingesetzt worden. Seitens der Geschlechterunterschiede in den Verordnungen gibt es für GLP-1-Rezeptoragonisten quasi keine Unterschiede, bei Metformin gibt es ein ganz leichtes Überwiegen der männlichen Patienten, welches bei SGLT-2-Inhibitoren deutlicher wird. Die mittlere glykämische Kontrolle ausgedrückt durch den HbA1c-Wert liegt in der gesamten Kohorte im Median bei 7,11 % (54,2 mmol/Mol) mit einem weitgehend unveränderten Verlauf über den Beobachtungszeitraum und einer klaren Differenzierung dahingehend, dass jüngere Patienten den höchsten medianen HbA1c aufweisen und die älteste Kohorte den niedrigsten. Die Daten werden im Kontext sich ändernder Leitlinien und Empfehlungen diskutiert und analysiert.
Article
Full-text available
Importance In clinical trials of patients with type 2 diabetes, long-acting insulin analogs modestly reduced the risk of nocturnal hypoglycemia compared with human neutral protamine Hagedorn (NPH) insulin, but cost 2 to 10 times more. Outcomes in clinical practice may differ from trial results. Objective To compare the rates of hypoglycemia-related emergency department (ED) visits or hospital admissions associated with initiation of long-acting insulin analogs vs human NPH insulin in patients with type 2 diabetes. Design, Setting, and Participants A retrospective observational study using data from Kaiser Permanente of Northern California from January 1, 2006, through September 30, 2015. Patients with type 2 diabetes who initiated a long-acting insulin analog or NPH insulin were included and censored at death, loss of health plan coverage, change in insulin treatment, or study end on September 30, 2015. Exposure Initiation of basal insulin analogs (glargine or detemir) vs NPH insulin. Main Outcomes and Measures The primary outcome was the time to a hypoglycemia-related ED visit or hospital admission and the secondary outcome was the change in hemoglobin A1c level within 1 year of insulin initiation. Results There were 25 489 patients with type 2 diabetes who initiated basal insulin therapy (mean age, 60.2 [SD, 11.8] years; 51.9% white; 46.8% female). During a mean follow-up of 1.7 years, there were 39 hypoglycemia-related ED visits or hospital admissions among 1928 patients who initiated insulin analogs (11.9 events [95% CI, 8.1 to 15.6] per 1000 person-years) compared with 354 hypoglycemia-related ED visits or hospital admissions among 23 561 patients who initiated NPH insulin (8.8 events [95% CI, 7.9 to 9.8] per 1000 person-years) (between-group difference, 3.1 events [95% CI, −1.5 to 7.7] per 1000 person-years; P = .07). Among 4428 patients matched by propensity score, the adjusted hazard ratio was 1.16 (95% CI, 0.71 to 1.78) for hypoglycemia-related ED visits or hospital admissions associated with insulin analog use. Within 1 year of insulin initiation, hemoglobin A1c level decreased from 9.4% (95% CI, 9.3% to 9.5%) to 8.2% (95% CI, 8.1% to 8.2%) after initiation of insulin analogs and from 9.4% (95% CI, 9.3% to 9.5%) to 7.9% (95% CI, 7.9% to 8.0%) after initiation of NPH insulin (adjusted difference-in-differences for glycemic control, −0.22% [95% CI, −0.09% to −0.37%]). Conclusions and Relevance Among patients with type 2 diabetes, initiation of a basal insulin analog compared with NPH insulin was not associated with a reduced risk of hypoglycemia-related ED visits or hospital admissions or with improved glycemic control. These findings suggest that the use of basal insulin analogs in usual practice settings may not be associated with clinical advantages for these outcomes.
Article
Full-text available
AIM To assess the efficacy and safety of semaglutide, a recently approved glucagon‐like peptide 1 receptor agonist (GLP‐1 RA) for type 2 diabetes. METHODS We searched major electronic databases and grey literature sources for randomised controlled trials comparing semaglutide with placebo or other antidiabetic agents. Primary outcome was change from baseline in HbA1c. Secondary endpoints included change from baseline in body weight, blood pressure, heart rate, and incidence of hypoglycaemia, gastrointestinal adverse effects, pancreatitis and diabetic retinopathy. RESULTS Six placebo‐controlled and seven active‐controlled studies with subcutaneous semaglutide were included. We identified only one trial with oral semaglutide. Compared with placebo, subcutaneous semaglutide 0.5 and 1 mg reduced HbA1c by 1.01% (95% CI 0.56 to 1.47) and 1.38% (1.05 to 1.70) respectively. Both doses demonstrated superior glycaemic efficacy compared to other antidiabetic agents, including sitagliptin, exenatide, liraglutide, dulaglutide and insulin glargine. Semaglutide also had a beneficial effect on body weight (mean difference versus placebo –4.11 kg, 95% CI –4.85 to –3.37 for semaglutide 1 mg) and systolic blood pressure. We did not observe increased hypoglycaemia rates with semaglutide; nevertheless, we noted an increased incidence of nausea, vomiting and diarrhoea. Cases of pancreatitis were infrequent and odds ratio for diabetic retinopathy compared with placebo was 1.32 (95% CI 0.98 to 1.77). CONCLUSIONS Semaglutide is a potent once‐weekly GLP‐1 RA, reducing significantly HbA1c, body weight and systolic blood pressure. However, it is associated with increased incidence of gastrointestinal adverse events. Results for pancreatitis and retinopathy require further assessment in post‐approval pharmacovigilance studies. This article is protected by copyright. All rights reserved
Article
Full-text available
BACKGROUND : Canagliflozin is a sodium glucose cotransporter 2 inhibitor that reduces the risk of cardiovascular events. We report the effects on heart failure and cardiovascular death overall, in those with and without a baseline history of heart failure, and in other participant subgroups. METHODS : The CANVAS Program (Canagliflozin Cardiovascular Assessment Study) enrolled 10 142 participants with type 2 diabetes mellitus and high cardiovascular risk. Participants were randomly assigned to canagliflozin or placebo and followed for a mean of 188 weeks. The primary end point for these analyses was adjudicated cardiovascular death or hospitalized heart failure. RESULTS : Participants with a history of heart failure at baseline (14.4%) were more frequently women, white, and hypertensive and had a history of prior cardiovascular disease (allP<0.001). Greater proportions of these patients were using therapies such as blockers of the renin angiotensin aldosterone system, diuretics, and β-blockers at baseline (allP<0.001). Overall, cardiovascular death or hospitalized heart failure was reduced in those treated with canagliflozin compared with placebo (16.3 versus 20.8 per 1000 patient-years; hazard ratio [HR], 0.78; 95% confidence interval [CI], 0.67-0.91), as was fatal or hospitalized heart failure (HR, 0.70; 95% CI, 0.55-0.89) and hospitalized heart failure alone (HR, 0.67; 95% CI, 0.52-0.87). The benefit on cardiovascular death or hospitalized heart failure may be greater in patients with a prior history of heart failure (HR, 0.61; 95% CI, 0.46-0.80) compared with those without heart failure at baseline (HR, 0.87; 95% CI, 0.72-1.06;Pinteraction =0.021). The effects of canagliflozin compared with placebo on other cardiovascular outcomes and key safety outcomes were similar in participants with and without heart failure at baseline (all interactionPvalues >0.130), except for a possibly reduced absolute rate of events attributable to osmotic diuresis among those with a prior history of heart failure (P=0.03). CONCLUSIONS : In patients with type 2 diabetes mellitus and an elevated risk of cardiovascular disease, canagliflozin reduced the risk of cardiovascular death or hospitalized heart failure across a broad range of different patient subgroups. Benefits may be greater in those with a history of heart failure at baseline. CLINICAL TRIAL REGISTRATION : URL: https://www.clinicaltrials.gov. Unique identifiers: NCT01032629 and NCT01989754.
Article
Aim: To assess the efficacy of ambulatory glucose profiling (AGP) generated by FreeStyle LibrePro™ flash glucose monitoring (FCGM) on glycemic control in patients with uncontrolled type 1 diabetes (T1D) and type 2 diabetes (T2D). Methods: Clinical and biochemical data were obtained from 5072 patients with diabetes who had an A1c ≥7% (2536 who had been initiated on FCGM-based AGP between March 2015 and October 2016 [cases] and 2536 age-, gender-, A1c-, site- and time-matched controls who were not initiated on AGP) across seven diabetes clinics in India. Anthropometric and clinical measurements were obtained through standardized techniques. Fasting and postprandial plasma glucose and glycated hemoglobin(A1c) were estimated before and after initiation of AGP. Results: Overall, there was a significant decrease in A1c both in cases and controls; however, the magnitude of reduction was higher among cases (1% vs.0.7%; P < 0.001).The overall reduction in A1c among cases was higher in T2D (9.2% to 8.3%) compared with T1D (9.6% to 9.4%); however, the absolute difference in A1c reduction between cases and controls was higher among T1D (0.5% vs. 0.2%) patients. The reduction in glycemic parameters was irrespective of age or gender (P for trend <0.001) across all study sites. The greatest reductions in A1c were noted within 6 months of AGP initiation. Multiple logistic regression showed that those who did not use AGP had a 1.42 higher risk (95% CI: 1.24-1.64) of not achieving even 0.1% reduction in A1c compared with those who were initiated on AGP even after adjusting for age, gender, body-mass index, systolic blood pressure, time to follow-up A1c, and medication use. Conclusions: This study shows that FCGM-based AGP with FreeStyle LibrePro is associated with significant reductions in A1c levels in both T1D and T2D. In addition, improvement in A1c levels was maintained across all age groups and in patients enrolled at different diabetes clinics in India.
Article
Nutrition therapy is considered a key component of diabetes management, yet evidence around the ideal macronutrient composition of the diet remains inconclusive. A systematic review and meta-analysis was performed to assess the effects of carbohydrate-restricted diets (≤ 45% of total energy) compared to high carbohydrate diets (> 45% of total energy) on glycemic control in adults with diabetes mellitus. Six databases were searched for articles published between January 1980 and August 2016. Primary outcome was between-group difference in HbA1c change. Individual effect sizes were standardized, and a meta-analysis performed to calculate pooled effect size using random effects. 25 RCTs involving 2412 participants were included. Carbohydrate-restricted diets, in particular those that restrict carbohydrate to < 26% of total energy, produced greater reductions in HbA1c at 3 months (WMD -0.47%, 95% CI: -0.71, -0.23) and 6 months (WMD -0.36%, 95% CI: -0.62, -0.09), with no significant difference at 12 or 24 months. There was no difference between moderately restricted (26-45% of total energy) and high carbohydrate diets at any time point. Although there are issues with the quality of the evidence, this review suggests that carbohydrate-restricted diets could be offered to people living with diabetes as part of an individualised management plan.
Article
Sulphonylureas have been commercially available since the 1950s, but their use continues to be associated with controversy. Although adverse cardiovascular outcomes in some observational studies have raised concerns about sulphonylureas, findings from relatively recent, robust, and high-quality systematic reviews have indicated no increased risk of all-cause mortality associated with sulphonylureas compared with other active treatments. Results from large, multicentre, randomised controlled trials such as the UK Prospective Diabetes Study and ADVANCE have confirmed the microvascular benefits of sulphonylureas, a reduction in the incidence or worsening of nephropathy and retinopathy, and no increase in all-cause mortality, although whether these benefits were due to sulphonylurea therapy and not an overall glucose-lowering effect could not be confirmed. A comparison of sulphonylureas and pioglitazone in the TOSCA.IT trial also confirmed the efficacy and cardiovascular safety of sulphonylureas. Investigators of randomised controlled trials have reported an increased risk of hypoglycaemia and weight gain with sulphonylureas, but data from observational studies suggest that the incidence of severe hypoglycaemia is lower in people taking sulphonylurea than in people taking insulin, and weight gain with sulphonylureas has been relatively modest in large cohort studies. 80% of people with diabetes live in low-to-middle income countries, so the effectiveness, affordability, and safety of sulphonylureas are particularly important considerations when prescribing glucose-lowering therapy. Results of ongoing head-to-head studies with new drugs, such as the comparison of glimepiride with linagliptin in the CAROLINA study and the comparison of various therapies (including sulphonylureas) for glycaemic control in the GRADE study, will determine the place of sulphonylureas in glucose-lowering therapy algorithms for patients with type 2 diabetes.
Article
Cardiovascular disease is a major cause of morbidity and mortality, in particular in people with diabetes and is also the largest contributor to healthcare costs in this population ⁽¹⁾. However, good quality randomised controlled trials and systematic reviews have shown that management of risk factors such as blood pressure, dyslipidaemia and glucose can lead to improved microvascular and macrovascular complications in people with type 2 diabetes mellitus (T2DM). Legacy effect or metabolic memory has been a phenomenon used to describe the prolonged benefits of glucose, blood pressure or lipid control in people with cardiovascular disease, diabetes or in primary prevention of cardiovascular disease by early risk factor control. This paper reviews the evidence in relation to longer term benefits of early risk factor control in people with diabetes.
Article
Background: Despite common mechanisms of actions, glucagon-like peptide-1 receptor agonists differ in structure, pharmacokinetic profile, and clinical effects. This head-to-head trial compared semaglutide with dulaglutide in patients with inadequately controlled type 2 diabetes. Methods: This was an open-label, parallel-group, phase 3b trial done at 194 hospitals, clinical institutions or private practices in 16 countries. Eligible patients were aged 18 years or older and had type 2 diabetes with HbA1c 7·0-10·5% (53·0-91·0 mmol/mol) on metformin monotherapy. Patients were randomly assigned (1:1:1:1) by use of an interactive web-response system to once a week treatment with either semaglutide 0·5 mg, dulaglutide 0·75 mg, semaglutide 1·0 mg, or dulaglutide 1·5 mg subcutaneously. The primary endpoint was change from baseline in percentage HbA1c; the confirmatory secondary endpoint was change in bodyweight, both at week 40. The primary analysis population included all randomly assigned patients exposed to at least one dose of trial product obtained while on treatment and before the onset of rescue medication. The safety population included all randomly assigned patients exposed to at least one dose of trial product obtained while on treatment. The trial was powered for HbA1c non-inferiority (margin 0·4%) and bodyweight superiority. This trial is registered with ClinicalTrials.gov, number NCT02648204. Findings: Between Jan 6, 2016, and June 22, 2016, 1201 patients were randomly assigned to treatment; of these, 301 were exposed to semaglutide 0·5 mg, 299 to dulaglutide 0·75 mg, 300 to semaglutide 1·0 mg, and 299 to dulaglutide 1·5 mg. 72 (6%) patients withdrew from the trial (22 receiving semaglutide 0·5 mg, 13 receiving dulaglutide 0·75 mg, 21 receiving semaglutide 1·0 mg, and 16 receiving dulaglutide 1·5 mg). From overall baseline mean, mean percentage HbA1c was reduced by 1·5 (SE 0·06) percentage points with semaglutide 0·5 mg versus 1·1 (0·05) percentage points with dulaglutide 0·75 mg (estimated treatment difference [ETD] -0·40 percentage points [95% CI -0·55 to -0·25]; p<0·0001) and by 1·8 (0·06) percentage points with semaglutide 1·0 mg versus 1·4 (0·06) percentage points with dulaglutide 1·5 mg (ETD -0·41 percentage points [-0·57 to -0·25]; p<0·0001). From overall baseline mean, mean bodyweight was reduced by 4·6 kg (SE 0·28) with semaglutide 0·5 mg compared with 2·3 kg (0·27) with dulaglutide 0·75 mg (ETD -2·26 kg [-3·02 to -1·51]; p<0·0001) and by 6·5 kg (0·28) with semaglutide 1·0 mg compared with 3·0 kg (0·27) with dulaglutide 1·5 mg (ETD -3·55 kg [-4·32 to -2·78]; p<0·0001). Gastrointestinal disorders were the most frequently reported adverse event, occurring in 129 (43%) of 301 patients receiving semaglutide 0·5 mg, 133 (44%) of 300 patients receiving semaglutide 1·0 mg, 100 (33%) of 299 patients receiving dulaglutide 0·75 mg, and in 143 (48%) of 299 patients receiving dulaglutide 1·5 mg. Gastrointestinal disorders were also the most common reason for discontinuing treatment with semaglutide and dulaglutide. There were six fatalities: one in each semaglutide group and two in each dulaglutide group. Interpretation: At low and high doses, semaglutide was superior to dulaglutide in improving glycaemic control and reducing bodyweight, enabling a significantly greater number of patients with type 2 diabetes to achieve clinically meaningful glycaemic targets and weight loss, with a similar safety profile. Funding: Novo Nordisk.
Article
Bariatric surgery has many benefits beyond weight loss, including improved control of glycaemia, blood pressure, and dyslipidaemia; hence, such surgery has been rebranded as metabolic surgery. The operations are, unfortunately, also associated with major surgical and medical complications. The medical complications include gastro-oesophageal reflux disease, malnutrition, and metabolic complications deriving from vitamin and mineral malabsorption. The benefits of surgery can be optimised by implementing specific protocols before and after surgery. In this Review, we discuss the assessment of the risk of major cardiac complications and severe obstructive sleep apnoea before surgery, and the provision of adequate lifelong postsurgery nutritional, vitamin, and mineral supplementation to reduce complications. Additionally, we examine the best antidiabetic medications to reduce the risk of hypoglycaemia after gastric bypass and sleeve gastrectomy, and the strategies to improve weight loss or reduce weight regain. Although optimising clinical pathways is possible to maximise metabolic benefits and reduce the risks of complications and micronutrient deficiencies, evolution of these strategies can further improve the risk-to-benefit ratio of metabolic surgery.