Therapy
Basic therapy
Adapting to a healthy lifestyle is crucial not only to prevent type 2 diabetes,
but also to reduce the complex pharmacotherapy and the development and
progression of diabetic complications of type 2 diabetes. In this context, it
makes sense to address not only one, but as many risk factors as possible
through lifestyle modification [15 ].
Education and training
As an indispensable part of diabetes treatment, all persons affected by
diabetes mellitus and, if applicable, their family members should be offered
structured, evaluated and target group- and topic-specific training and
treatment programmes as well as, if necessary, problem-oriented follow-up
training [16 ].
Plasma glucose self-monitoring
In the case of an indication for plasma glucose self-monitoring, the
situations listed in [Tab. 2 ] should be
taken into account in people with type 2 diabetes. However, the measurements
should result in behavioural and therapeutic adjustments.
Tab. 2 Situations in which plasma glucose
self-monitoring is necessary or may be temporarily necessary in
people with type 2 diabetes1 .
Clinically defined situations
Diabetes stage
Newly diagnosed, adjustment phase
Diabetes along its course
Unstable with frequent hypoglycaemia (at this
point, measure before all meals until the therapy
goal is achieved, then return to targeted
situational measurements)
Therapy intensification
Temporarily after switching from insulin to oral
antidiabetic therapy
Additional illnesses/interventions
Serious infections
Planned operations
Mental illnesses with unreliable intake of
medication
During sport/exercise and blood
glucose-lowering substances, which may be
associated with hypoglycaemia, and corresponding
symptoms occur
Acute changes in diet due to illness
(e. g., diarrhoea/vomiting)
Diabetes therapy
Oral antidiabetics (OAD) with hypoglycaemia
potential (sulfonylureas, glinides, then
occasional measurements)
Insulin therapy and necessity of insulin dose
self-adjustment
Intensified conventional insulin therapy (before
all meals, occasionally at night)
Insulin pump therapy (before all meals,
occasionally at night)1
Situations with special hazards (e. g.
shift work, driving lorries, buses, cranes,
etc.)
1 G-BA decision of June 16, 2016 (BAnz AT 06.09.2016 B3):
Continuous interstitial glucose measurements with real-time
measuring devices (rtCGM) for therapy control in patients with
insulin-dependent diabetes mellitus can be provided under special
conditions as contracted medical services at the expense of the
health insurance funds. The costs for FGM (“flash
glucose monitoring”), also known as
“intermittent-scanning continuous glucose
monitoring” (iscCGM), are now also covered by insurance
companies.
Urine glucose analyses
These are not standard in the diagnosis, therapy decision-making and
monitoring, because urine glucose is only positive in the case of high blood
glucose values (renal glucose transport capacity is very different between
individuals, it is age-dependent, it is not systematically examined at
reduced kidney function, it lowers with certain diseases and is not useful
in pregnancy or with the use of drugs such as Sodium-glucose Cotransporter-2
(SGLT2) inhibitors). However, in the assessment of hyperglycaemic metabolic
derailment, however, the measurement of ketonuria is decisive for
therapy.
Nutritional therapy and consultation
Nutritional recommendations for people with type 2 diabetes should include
the following key points. These are just a few recommendations:
Motivation to maintain a healthy, well-balanced diet considering the
patient’s previous nutrition routine. At the same time, the
joy of eating should be preserved.
As far as possible, the use of industrially-processed food should be
avoided, and the intake of sucrose should be limited (World Health
Organization [WHO] recommendation<25 g/day).
The German Nutrition Society (DGE) recommends limiting mono- and
disaccharide consumption to<10% of daily energy
intake.
No generalized ban on sugar, but avoidance of large amounts of
regular sugar, fructose, sugar alcohols (e. g., sorbitol,
xylitol) or drinks containing these substances.
The estimation of type and amount of carbohydrates of each meal
should be used as an essential metabolic control strategy for people
with type 2 diabetes who inject insulin.
People with type 2 diabetes without insulin therapy should be able to
recognize foods which increase blood glucose.
For people with type 2 diabetes and renal insufficiency, a daily
protein intake of 0.8 g/kg is recommended. At the
dialysis therapy stage, the protein intake should be increased to
1.2–1.3 g/kg.
People with type 2 diabetes should be advised how to deal with
alcohol in a differentiated manner as part of the individual
consultation.
Practical recommendations for a healthy and balanced diet, a
Mediterranean diet at best [17 ]
[18 ]
[19 ]
[20 ]
[21 ].
Avoidance of large portions and frequent consumption of fatty foods,
e. g., fatty meat, fatty sausages, fatty cheese, fatty baked
goods, fatty ready-made products, fatty fast food, cream, chocolate,
chips, etc.
Choosing vegetable fats, e. g., oils, nuts, seeds.
Enriching meals with dietary fibres, e. g., vegetables, fresh
fruit, whole grain cereals.
The effectiveness of weight loss and improvement of the vascular risk profile
always depends on how the diet is designed: low-carb, vegan or Mediterranean
- how well the acceptance and adherence as well as the long-term management
of the dietary change succeed [21 ]
[22 ].
Weight reduction
Weight reduction in overweight and obese people with type 2 diabetes supports
the reduction of vascular risks, increases self-esteem, quality of life and
can lead to remission in the early stages of type 2 diabetes [20 ]
[23 ]
[24 ]
[25 ]
[26 ].
Physical activity (see [Fig.
1 ])
Fig. 1 Therapy algorithm for type 2 diabetes. 1
Lifestyle-modifying, non-drug therapy measures are the basic therapy
at every therapy level. AHA/ACC = American Heart Association
/ American College of Cardiology; ESC/EAS = European Society
of Cardiology / European Atherosclerosis Society (EAS); DDG
= German Diabetes Association; KDIGO = Kidney
Disease: Improving Global Outcomes; ADA = American Diabetes
Association
Increased physical activity and sport are essential therapeutic interventions
for all forms of diabetes. Physical activity is particularly beneficial for
people with type 2 diabetes for a number of reasons [27 ]
[28 ]
[29 ]. The structured
approach is outlined in the step-by-step programme [see Appendix] of the
guideline. Extensive practical recommendations can be found in this
supplement [30 ].
In brief:
People with type 2 diabetes should be motivated to increase their
physical activity.
It should be decided which types of exercise or sports are suitable
for people with type 2 diabetes on an individual basis.
Aerobic endurance training and strength training to build and
maintain musculature should be offered as structured movement
programmes.
At least 150 min of moderate intensity exercise are
recommended per week [31 ].
Lower intensity training shows lower drop-out rates and seems to be
more successful in the long run than high intensity exercise
training, either intermittently or continuously [32 ]. In particular, it is
recommended for people with type 2 diabetes in the second half of
their life to train dexterity, reactions, coordination, flexibility
and mobility.
Cessation of smoking
Active and passive smoking, in addition to being a preventable cause of
significantly increased morbidity and mortality, are also significant risk
factors for type 2 diabetes (Pan A, Wang Y, Talaei M et al. Relation of
active, passive, and quitting smoking with incident diabetes: a
meta-analysis and systematic review. Lancet Diabetes Endocrinol 2015;3 (12):
958–996). In a recently published meta-analysis, smoking was shown
to be an independent risk factor for the progression of albuminuria [33 ]. Albuminuria is one of the strongest
predictors for the development and progression of cardiovascular
complications. When appropriate to the situation, smokers should therefore
always be educated and specifically counselled about the particular risks of
smoking for type 2 diabetes, microvascular and macrovascular sequelae and
pulmonary disease. They should be strongly advised to stop smoking
tobacco.
Further information on tobacco cessation and support for quitting smoking can
be found in the S3 guideline “Smoking and Tobacco Dependence:
Screening, Diagnosis and Treatment”, Update 2021 [34 ] and in the Tobacco Atlas Germany [35 ].
Smokers who are willing to change should receive regular counselling
regarding possible tobacco cessation procedures (see Appendix; [Fig. 2 ]).
Fig. 2 Algorithm for drug therapy in type 2 diabetes. eGFR
= Estimated Glomerular Filtration Rate; DPP4-inhibitor
= Dipeptidyl peptidase-4 inhibitor; GLP-1 RA =
Glucagon-like peptide-1 receptor agonist; SGLT2 =
Sodium-glucose cotransporter-2
The basic therapy at every therapy level comprises lifestyle-modifying,
non-drug therapy measures, but these are often not sufficient on their own.
In patients for whom lifestyle-modifying measures are not expected to be
sufficiently successful (due to severity of metabolic derailment, adherence
problems, multimorbidity), these measures should be combined with metformin
and, if contraindicated or intolerant, with another antidiabetic drug. Most
people with type 2 diabetes have multimorbidity and thus, depending on the
individual therapy goal, there is a need for polypharmacy with
prioritisation according to the severity of vascular risks ([Fig. 1 ]).
Pharmacotherapy
The step-by-step procedure provided in the therapy algorithm ([Figs. 1 ]
[2 ]) refers to the time of clinical diagnosis of type 2 diabetes in the
stage of relative metabolic compensation. Newly-diagnosed patients with
metabolic decompensation should receive basic therapy and pharmacotherapy
(e. g., even insulin) at the same time.
Risk assessment
Before starting drug treatment, a detailed risk assessment is absolutely
necessary, because this determines the choice and possible combination of
antidiabetic and organ-protective drugs. In [Tab. 3 ], important risk factors are listed in accordance with the
National Care Guideline:
Tab. 3 Risk factors for which early use of
organ-protective drugs is indicated. Data source: [1 ]
Duration of diabetes (>10 years)
(Biological) age
Gender (male>female)
Lifestyle: unbalanced diet/physical
inactivity
Family history of early cardiovascular
disease
(Men<55 years; women<60
years)
Hypertension or antihypertensive therapy
Dyslipidaemia or lipid-lowering therapy
Obesity (>30 kg/m2)
Renal insufficiency
(eGFR<60 ml/min.)
Albuminuria (>30 mg/g U
creat.)
Smokers and ex-smokers
Subclinical arteriosclerosis or cardiovascular
disease
Left ventricular hypertrophy
Obstructive sleep apnoea syndrome
Due to the complexity and the large number of risk factors ([Tab. 3 ]), which have not been evaluated in
their entirety, the risk assessment cannot be depicted in the form of
scores. The analysis of important RCTs impressively shows how heterogeneous
the inclusion criteria for the study participants were (
[Tab. 4 ]). In addition, most RCTs (strict
inclusion and exclusion criteria) only represent a maximum of
4–50% of real-world patients. In order to assess the
effectiveness of interventions in Randomised controlled trials (RCTs) in
real-world settings, pragmatic and register studies with the same patient
characteristics as in corresponding RCTs are therefore necessary. Thus, only
an individual careful assessment of the risk for cardiovascular and renal
diseases before implementation of the corresponding therapy algorithm is
helpful at present ([Fig. 2 ]
[3 ]).
Fig. 3 Algorithm for insulin therapy [1 ] in addition to [Fig. 2 ]. The algorithm does not
refer to people with severe metabolic decompensation or emergency
situations. Current specialist information must be taken into
account. Review the therapy strategy and the therapy goal in
3–6 months at the latest. GLP-1 RA = Glucagon-like
peptide-1 receptor agonist; SGLT2 = Sodium-glucose
cotransporter-2
Tab. 4 Criteria used to diagnose high cardiovascular
risk (in patients without manifest atherosclerotic heart
disease) in 12 published cardiovascular
“Outcome” studies on the effect of GLP-1
receptor agonists or SGLT2 inhibitors: EMPA-REG, CANVAS-Program,
DECLARE TIMI-58, VERTIS CV, ELIXA, LEADER, SUSTAIN-6, EXSCEL,
REWIND, HARMONY Trials, PIONEER-6, AMPLITUDE-O.
Criteria
Frequency (n)
Frequency (%)
Comment
Age≥50, 55, or 60 years
6
100
Basic criterion, requires additional risk
factors
Plus reduced renal function (eGFR
25–59.9 ml/min.)
1
17
Also occurs as CHD-equivalent
Plus≥1 (n=4) or≥2
(n=2) further risk factors (see
below)
6
100
Further risk factors (see below)
Diabetes duration≥10 years
1
17
Main criterion according to ESC
Arterial hypertension (>140
and>90 mmHg or antihypertensive
medication)
3
50
Surprisingly low rated
Smoking/tobacco use
3
50
Surprisingly low rated
Micro- or macroalbuminuria
5
83
Central and meaningful criterion
HDL cholesterol low
(e. g.,<1 mmol/l or
42.5 mg/dl)
2
33
Surprisingly low rated
LDL cholesterol elevated
(e. g.,>3.36 mmol/l/or
130 mg/dl)
2
33
Surprisingly low rated
Lipid-modifying therapy
1
17
Surprisingly low rated
Left ventricular hypertrophy (in arterial
hypertension)
3
50
Hypertension with end organ damage
Left ventricular systolic or diastolic
dysfunction
3
50
Heart failure
Ankle-brachial index<0.9 (≥1 leg
affected)
3
50
Is also used for already manifested PAD
Obesity
1
17
Surprisingly low rated
First-degree relative(s) with coronary heart disease with
manifestation≤55 years (men) or≤65 years
(women)
1
17
Seldom mentioned
6 of 12 cardiovascular “outcome” studies recruited
patients without manifest disease due to risk factors. The
percentages refer to this total number (6 studies). Criteria that
were used consistently often (≥50%) are highlighted
in bold. All other criteria were suggested in a maximum of
33% of the studies.
Overview with regard to metabolic effects and clinical endpoints
[Tab. 5 ] allows a quick, orientating
overview with regard to metabolic effects and clinical endpoints of the
pharmaceuticals discussed in this Clinical Practice Guideline - apart from
oral semaglutide, which was not inferior to subcutaneous semaglutide in
terms of clinical endpoints. The table is a careful interpretation of the
available evidence from randomised controlled trials and meta-analyses,
which was compiled and consulted by the Medical Centre for Quality in
Medicine and the National Care Guidelines working group
(www.leitlinien.de/nvl/diabetes; AWMF Register No. 001;
[1 ] and supplemented by the author
group of this Clinical Practice Guideline because of new study results.
Tab. 5 Informative, comparative consideration of the substance classes (supplement to the
algorithm (▶Fig. 2)). This table is a summary interpretation of the evidence. For
a detailed presentation of the
evidence, see Type 2 Diabetes. Long version. 2nd edition, 2020. consultation version.
AWMF Register No.: nvl-001 [rerif].
Medicine
Total mortality
Cardiovascular endpoints
Microvascular endpoints 1
Renal endpoints
Hypoglycaemias
HbA1c Weight
Comments/selected safety information
Metformin
0
0
0
0
↔↑
HbA1c ↓↓ Weight:
↔↓
Risk of lactic acidosis, taking a break in form of sick
days when unwell
SGLT2 inhibitors
HbA1c ↓↓ Weight: ↓
Risk of genital infections, atypical ketoacidosis,
Fournier gangrene Taking a break in form of sick
days when unwell Weight reduction (undesired in
cases of frailty)
Dapagliflozin
0* sinks with patients with HF
MACE: 0 CV death: 0 heart failure-related
hospitalization: ↓ sinks
Not specified: retinopathy, neuropathy, amputations:
0
↓ sinks
↔
HbA1c ↓↓ Weight: ↓
Empagliflozin
↓ sinks*
MACE: ↓ sinks CV death: ↓
sinks heart failure-related hospitalization:
↓ sinks
Not specified
↓ sinks
↔
HbA1c ↓↓ Weight: ↓
Ertugliflozin
MACE: 0 CV death: 0 heart failure-related
hospitalization: ↓ sinks
0 (eGFR decrease is reduced)
GLP-1-RA
Gastrointestinal side effects, gallstones Low risk
of pancreatitis Injections
necessary Weight reduction (undesired in cases of
frailty)
Dulaglutide
0
MACE: ↓ sinks CV death: 0 heart
failure-related hospitalization: 0
Retinopathy: 0 Not specified: neuropathy,
amputations
↓ sinks
↔
HbA1c ↓↓ Weight: ↓
Exenatide
↓ sinks*
MACE: 0 CV death: 0 heart failure-related
hospitalization: 0
Amputations: 0
Not specified
↔
HbA1c ↓↓ Weight: ↓
Liraglutide
↓ sinks*
MACE: ↓ sinks CV death: ↓ sinks
heart failure-related hospitalization: 0
Retinopathy: 0 Not specified: neuropathy,
amputations
↓ sinks
↔
HbA1c ↓↓ Weight: ↓
Lixisenatide
0*
MACE: 0 CV death: 0 heart failure-related
hospitalization: 0
Not specified: retinopathy, neuropathy, amputations
Not specified
↔
HbA1c ↓↓ Weight: ↓
Semaglutide
0*
MACE: ↓ sinks CV death: 0 heart
failure-related hospitalization: 0 For ORAL
semaglutide: MACE: 0 CV death: ↓
sinks heart failure-related hospitalization: 0
Retinopathy: ↑ Not specified: neuropathy,
amputations
↓ sinks
↔
HbA1c ↓↓ Weight: ↓
Caution in case of pre-existing retinopathy
Sulfonylureas
(0)
MACE: (0)* CV death:
(0) heart failure-related hospitalization:
(0)
(0 to ↓)
(0 to ↓)
↑↑
HbA1c ↓↓ Weight: ↑
-Risk of severe, prolonged hypoglycaemias -CVOT
study: no difference in the primary CV endpoint in
direct comparison to CV-neutral linagliptin
DPP-4 inhibitors
(0)
MACE: (0) certain CV death: (0) heart
failure-related hospitalization: (0)
(0)
(0)
↔
HbA1c ↓↓ Weight: ↔
Very rare: pancreatitis, inflammation bowel
diseases CVOT present for sitagliptin,
saxagliptin, linagliptin Vildagliptin has NO
CVOT Saxagliptin is not recommender with
pre-existing heart failure
Possibly as of stage 3 of the algorithm
Insulin
(0)
(0)
(↓)
(0)
↑↑
HbA1c ↓↓(depending on the
dose) Weight: ↑↑
Effects on endpoints: ↓: positive effect (endpoint was
reached less frequently in the studies), ↑: negative effect
(endpoint was reached more frequently in the studies); 0: endpoint
was not affected in the studies considered, assumptions in
parentheses () are from studies with low methodological quality, or
there was insufficient evidence to assess.; All-cause mortality
endpoint* : The study was not laid out for the
endpoint all-cause mortality.; MACE: cardiovascular death, stroke,
myocardial infarction (for exact definition, see cardiovascular
endpoint studies); CV death: cardiovascular death, HHI:
hospitalization for heart failure.; CVOT: cardiovascular outcome
studies. n.s.: not specified (effect sizes were not reported or were
reported without confidence interval in the main publication).;
Hypoglycaemia: ↑ increased risk, ↔ low risk; HbA1c:
↓: decrease; weight: ↑: weight gain, ↓:
weight loss.; Compared with linagliptin in CVOT, dapagliflozin and
ertugliflozin are approved for the treatment of chronic heart
failure. This applies to patients with impaired left ventricular
function (HFrEF). Then dapagliflozin can be given up to an eGFR of
30 ml/min. and empagliflozin up to an eGFR of
20 ml/min.; Safety aspects and effects listed
represent the state of discussion of the available evidence in the
expert group and should not be considered a comprehensive
presentation.; 1 microvascular endpoints: retinopathy,
neuropathy, amputations.
Reasons for the therapy level non-drug basic therapy
Basic therapy includes all lifestyle-modifying, non-drug measures. These
include education and training of the patient, nutritional therapy,
increasing physical activity and smoking cessation, as well as stress
management strategies. An important goal is to strengthen the will to lead a
healthy lifestyle (refraining from smoking, maintaining a
diabetes-appropriate diet, increased physical activity, limiting alcohol
consumption) ([Fig. 2 ]
[3 ]). Digital tools and telemedical support
are becoming increasingly important for the implementation of a personalised
basic therapy [36 ].
Since many people with type 2 diabetes have a variety of other vascular risk
factors in addition to chronic hyperglycaemia or already have
cardiovascular, renal and other diseases, the treatment of these people is
complex and should take into account all vascular risk factors and
manifested clinical diseases individually. To emphasise this more clearly,
the previous treatment algorithm has been expanded to address major
cardiovascular risks in more detail.
Reasons for pharmacotherapy therapy level
The basic therapy plays an important role in every further level of therapy
modification. Pharmacotherapy is indicated to achieve the individual therapy
goals if these lifestyle-modifying measures cannot be implemented or cannot
be implemented adequately by the person with diabetes and are therefore not
successful or do not make sense in the foreseeable future (2–3
months). Whenever possible, the advantages of metformin (see appendix)
should be used and doses should start gradually and increase slowly
(e. g., starting with 500 mg with the main meal and
increasing by another 500 mg each week up to a total dose of
2×1000 mg per day).
In case of contraindications (eGFR!) or poor tolerability of metformin
(mainly dose-dependent gastrointestinal complaints), other options for
monotherapy are available and should be used according to the patient risk
profile (cardiorenal risks and morbidity) and the other patient-relevant
benefits (influence on body weight, risk of hypoglycaemia, metabolic
effects, side effect profile and clinical endpoints). It is essential that
patient preferences are taken into account, as this is the only way to
ensure good treatment adherence.
In patients with cardiovascular or renal diseases or a very high
cardiovascular risk ([Tab. 3 ]),
substances that reduce evidence-based cardiovascular and renal diseases as
well as mortality (SGLT2 inhibitors, GLP-1 receptor agonists) should be used
primarily in combination with metformin
(eGFR>30 ml/min.!). For people with type 2 diabetes
with HbA1c levels significantly outside the individual glucose target range
(e. g.,>1.5% above the target range) at diagnosis,
initial pharmacotherapy, including the use of multiple combinations
including insulin, if necessary, is warranted. After reaching the HbA1c
target value, the therapy should be adjusted at individually agreed
intervals.
Reason for combination therapies
A dual combination is necessary for many patients for metabolic reasons and
is more favourable with regard to side effects of the individual substances,
since in some cases lower doses can be used in the combination.
An early combination therapy should be aimed for in order to avoid derailing
the metabolic parameters far from the agreed target range [37 ]
[38 ].
The target values should usually be checked at 3-month intervals. There is
now a large number of publications with good evidence for the selection of
combinations. Patient preferences, individual therapy goals, simplicity of
treatment, existing cardiovascular diseases and possible contraindications
also play an important role. If the number of oral medications becomes too
complex due to the complexity of the therapy, vascular risk factors or
comorbidities (including COPD, depression, chronic pain conditions, etc.),
fixed combinations should be used wherever possible. Parenteral blood
glucose-lowering principles (GLP-1 RAs, insulins) can also be useful and
helpful for these patients and significantly increase therapy adherence. The
higher the HbA1c level, the more likely the use of insulin, but this does
not mean that initial insulin therapy must be continued after metabolic
recompensation. De-escalation strategies should be considered for each
patient.
The administration of more than 2 oral antidiabetic agents may be
individually-appropriate if therapy with a GLP-1-RA or insulin is not yet
indicated ([Fig. 3 ]), the patient is not
yet comfortable with injection therapy, or this therapy should be delayed
for other reasons.
Oral triple therapy in the combination of metformin, a DPP4 inhibitor and an
SGLT2 inhibitor is a safe, effective and simple therapy. Potentiation of
side effects has not been observed with oral triple combination; they are
essentially the same as those observed with monotherapy for the respective
substance.
In case of non-response to therapy, the patient’s compliance with
therapy should always be discussed before increasing the dose or changing
the treatment.
Reasons for injection therapy
Due to lower hypoglycaemia rates and a favourable body weight progression
(compared to intensified insulin therapy), starting with GLP-1 RA-assisted
therapy or basal insulin in combination with oral antidiabetics is
recommended for most cases ([Fig.
3 ]).
Insulin dose reduction should absolutely be considered in case of worsening
renal function in order to avoid severe hypoglycaemia.
A combination of GLP-1-RA with oral antidiabetic drugs (except DPP4
inhibitors) is an effective treatment if the individual therapy goal was not
achieved with the previous oral antidiabetic drugs in mono- or multiple
combinations or if side effects make a new therapy strategy absolutely
necessary. In principle, the use of GLP-1-RA should be considered before
starting a therapy with insulin, especially because of the very low
hypoglycaemia risk of the substance class, the favourable weight progression
and the favourable cardiovascular and renal outcome data of these
substances.
Combinations of a GLP-1-RA with a basal insulin lead to a significant delay
in the intensification of antidiabetic therapy (e. g., escalation of
the basal insulin dose or additional administration of prandial insulin), to
significantly better metabolic control without a significant increase in the
risk of hypoglycaemia and to favourable weight effects [39 ]
[40 ]
[41 ]
[42 ]
[43 ].
Only when these combination therapies are no longer sufficiently effective or
indicated will a further intensification of insulin therapy with prandial
insulin be required in a next step.
Flexibility of therapy decisions due to the heterogeneity of type 2 diabetes
and individual therapy goals is necessary at every stage of treatment. In
most cases, persuasion to accept injection treatment and extensive
education/training of the patient are necessary. In individual
cases, a Continuous Subcutaneous Insulin Infusion (CSII) is indicated if the
therapy goals are not achieved sufficiently under intensified conventional
therapy (ICT).
Treatment of dyslipidaemia
Dyslipidaemia is common in people with type 2 diabetes and is an important
vascular risk factor. Detailed information on the treatment of dyslipidaemia
can be found in the ESC/EAS guideline [6 ] and in the practice recommendation of this supplement [44 ].
Treatment of arterial hypertension
Arterial hypertension is an important cardiovascular and renal risk factor
that should be treated early and consistently. Structured training on
hypertension, including practical training of patients to self-monitor their
blood pressure, is helpful [45 ]. Detailed
information on the treatment of hypertension has been discussed in
guidelines [7 ]
[46 ]
[47 ]
[48 ] and other publications [49 ]
[50 ].
Appendix
Medical history and clinical examinations
Tab. 1 Medical history and clinical examinations in people with type 2 diabetes.
History and examination
It should be noted that type 2 diabetes is frequently poor in
symptoms or asymptomatic and that the symptoms are often
overlooked.
Excess weight/obesity
High blood pressure
Lipid metabolism disorders
Thirst
Frequent urination
Involuntary weight loss
Tendency to infection – especially infections of the skin or mucous membranes
Exhaustion, fatigue, weakness
Physical inactivity
Drug intake (e. g., glucocorticoids, psychotherapeutics)
Alcohol consumption
Smoking
Depression
Exertional dyspnea
NYHA Class
Angina symptoms
Intermittent claudication (walking distance)
Memory deficits, cognitive dysfunction
Visual disturbances, retinopathy
Periodontitis
Erectile dysfunction
Birth of children > 4000 g
Family history
Physical examination
Height
Weight (BMI)
Waist circumference (in the middle between lower rib-bone and upper iliac
crest right after exhaling normally)
Cardiovascular system
Blood pressure
Peripheral arteries, pulse status [1]
Peripheral nervous system [2]
Skin
Oral hygiene (periodontitis) [230]
Eye examinations [3]
Foot examinations [4]
Laboratory values
Optional GAD: antibodies test for the sometimes-difficult differentiation
to type 1 diabetes or LADA and insulin or better C-peptide (with
HOMA2-B and HOMA2-IR) in cases of unclear differential diagnosis
or for subtyping if this results in a therapeutic consequence (see also
the Clinical Practice Guideline “Definition, classification and
diagnosis of diabetes mellitus’ in this supplement)
Plasma glucose
Blood count
HbA1c
Creatinine
eGFR
Potassium
Lipid profile
Gamma GT
AST
ALT [5]
Uric acid [6]
Urinalysis incl. albuminuria or UACR (albumin mg/g creatinine), ketones in urine
or blood (only for high glucose values; for SGLT-2 inhibitor therapy, also at
plasma glucose values < 250 mg/dl [13.9 mmol/l])
Technical examinations
Resting and exercise ECG [7, 7]
Echocardiography with or without pharmacological stress as an alternative to a
stress ECG; ask about (HFpEF/HFrEF)
Abdominal sonography (fatty liver and others)
Eye examination
Ankle-brachial index for weak or not palpable pulses in the feet (consider:
media sclerosis)
NYHA class = New York Heart Association; BMI = Boy Mass Index; GAD = glutamic acid
decarboxylase; LADA = Latent Autoimmune Diabetes in Adults;
HOMA2-B = Homeostatic Model Assessment2-beta cell function; HOMA2-IR = Homeostatic
Model Assessment2-Insulin resistance; HbA1c =
Haemoglobin A1c; eGFR = Estimated Glomerular Filtration Rate; Gamma GT = Gamma Glucose
Tolerance; AST = Aspartate transaminase; ALT =
Alanine transaminase; UACR = Urinary Albumin-to-Creatinine Ratio; SGLT-2 = Sodium
Glucose CoTransporter 2; ECG = Echocardiography; HFpEF =
Heart Failure with Preserved Ejection Fraction; HFrEF = Heart Failure with Reduced
Ejection Fraction
Tab. 2 Monitoring of people with type 2 diabetes.
History/Examination/Screening
History
Diabetes duration
Weight/BMI, waist-height ratio if applicable
(weight progression, excess weight)
Blood pressure
Foot status
Previous therapy (complete medication plan if
possible)
Physical activity
Eating habits
Smoking
Diabetes education and training programme carried out,
blood glucose self-monitoring
Hypoglycaemia (frequency and severity)
Anxiety
Depression
Erectile dysfunction
Physical examination
Laboratory values Screening for oral hygiene
HbA1c
Creatinine clearance rate (eGFR)
Lipid profile including LDL-, HDL-cholestrol
Urinalysis incl. albuminuria or UACR (albumin
mg/g creatinine), ketones in urine or blood
(only for high glucose values; for SGLT-2 inhibitor
therapy)
People with type 2 diabetes should be regularly checked
for periodontitis
Screening for diabetic neuropathy [2]
People with type 2 diabetes neuropathy should be screened once
per year from the moment of diagnosis for sensorimotor and
autonomic neuropathy.
Screening for foot lesions [4]
People with type 2 diabetes also with no clinical findings of
sensorimotor neuropathy should be examined for foot lesions at
least once a year. If clinical findings of sensorimotor
neuropathy are already present, regular examinations for foot
lesions should be carried out every 3–6 months.
Screening for nephropathy [9]
People with type 2 diabetes should be examined for albuminuria at
least once a year, as this allows a significant additional risk
assessment for cardiovascular and renal complications. In
addition, the eGFR should be determined, whereby the frequency
of the measurement varies depending on the stage of the renal
disease and possible renal complications (nephrotoxic
substances, contrast agents, hypovolemia).
Screening for retinal complications [3]
An ophthalmic screening should be performed:
For type 2 diabetes upon diagnosis (initial examination).
If no diabetic retinal change is detected, the screening
interval should be
2 years in case of known low risk (=no
ophthalmological risk and no general risk),
1 year for all other risk constellations.
If the ophthalmologist does not know the general risk factors,
he/she should treat the patient as with an unfavourable
general risk profile. Patients with diabetic retinopathy changes
(=ophthalmic risk) should be examined annually or more
frequently, depending on the findings. In the case of
newly-occurring symptoms such as deterioration of vision,
distorted vision, blurred vision and/or floaters, an
examination should be carried out promptly at the
ophthalmologist.
Assessment of macro- and microvascular overall risk
People with type 2 diabetes should be examined for vascular risks
(hypertension) at least once a year and they should be asked
whether they smoke. In addition, HbA1c, lipids, uric acid and
circulatory parameters (blood pressure measurement and pulse
measurement at different sites) should be controlled and a
micro-/macroalbuminuria should be measured
quantitatively. Looking for symptoms of heart insufficiency
should be done at least twice a year.
BMI = Body Mass Index; GLP-1-RA =Glucagon-like Peptide-1 Receptor Agonists; FGM =
Flash Glucose Monitoring; CGM = Continuous Glucose
Monitoring; HbA1c = Hemoglobin A1c; eGFR = Estimated Glomerular Filtration Rate; LDL-cholestrol
= Low Density Lipoprotein-cholestrol; HDL-cholestrol
= High Density Lipoprotein-cholestrol; UACR = Urinary Albumin-to-Creatinine Ratio;
SGLT-2 = Sodium Glucose CoTransporter 2
Physical exercise
Regular exercise is particularly important for people with type 2 diabetes
[10–17].
Tab. 3 Benefits of regular physical activity
Lowers blood pressure
Reduces heart rate at rest and under stress
Improves dyslipidaemia
Reduces cardiovascular risk
Reduces insulin resistance
Supports weight loss
Improves the flow of blood and thus the supply of muscles and organs
Reduces the risk of thrombosis
Relieves chronic pain
Prevents certain types of cancer
Strengthens the immune system
Strengthens confidence in one’s own ability and thus self-esteem
Lifts the mood and reduces stress
Promotes mobility and coordination, especially in older people
Promotes general well-being
Lifestyle measures – Figure
Fig. 1 Step programme for physical activity. Data source: [231].
Smoking cessation
Fig. 2 Algorithm for the approach to smoking. Source: German Medical Association (BÄK),
National Association of Statutory
Health Insurance Physicians (KBV), Association of the Scientific Medical Societies
(AWMF). National Health Care Guideline Therapy
of Type 2 Diabetes - Long version, 1st edition. Version 4. 2013, last modified: November
2014. Available from: www.dmtherapie.versorgungsleitlinien.de; [cited: 15.08.2018]; DOI: 10.6101/AZQ/000 213 [rerif].
Critical presentation of the individual antidiabetic pharmaceuticals
Metformin
Thanks to its effectiveness in lowering the HbA1c value, its well-known safety
profile, the approval conditions of other substances with positive effects in CVOTs,
extensive experience with it and its’ low costs, metformin continues to be
the antidiabetic drug of first choice for the treatment of type 2 diabetes. The low
risk of hypoglycaemia (caveat: simultaneous alcohol consumption) and the beneficial
effect of slightly reducing weight are also advantageous. The indication as
monotherapy and in combination therapy with metformin was expanded in February 2017
[18]:
Patients with a renal insufficiency up to degree 3b (>eGFR
30 ml/min) can be treated with metformin if there are no
other contraindications.
Maximum daily dose is 1000 mg (500–0–500 mg)
for an eGFR of 30–44 ml/min. At this eGFR, a
metformin therapy should not be started.
Maximum daily dose is 2000 mg for an eGFR of
45–59 ml/min.
To be on the safe side, a dose reduction to 500 mg per day can be
carried out at an eGFR of 30–44 ml/min, because the
eGFR can worsen acutely at this level, particularly in elderly people with
exsiccosis or due to kidney toxic drugs.
The pros and cons of metformin therapy at an eGFR of
30–44 ml/min must be explained to the patient.
In the population-based large study involving 75 413 patients of the Geisinger Health
System, an analysis of all patients with regard to hospitalisation due to acidosis
was carried out. 2335 hospitalizations due to acidosis were found in the period from
2004 to 2017 (mean follow-up time of 5.7 years). In this clinical real-world setting
and compared to other antidiabetic drugs (excluding insulin), metformin was only
associated with lactate acidosis if the eGFR was lower
than<30 ml/min. [19]
As far as clinical endpoints are concerned, despite the frequent use of metformin,
the data are inconclusive. Positive data from the UKPDS can be found in a relatively
small number of overweight patients and from several small studies. In a recent
meta-analysis, neither significant positive nor negative effects of metformin on
cardiovascular endpoints were found [20]; however, the authors admit that the
numbers are too small for a meta-analysis and a large controlled study would be
necessary to clarify the question. Correspondingly, there is no evidence of an
advantage of metformin for a given combination therapy with respect to
cardiovascular endpoints and all-cause mortality [21,22]. The European Society of
Cardiology Guidelines have replaced primary therapy with metformin with SGLT2
inhibitors and GLP-1-RA in patients with newly-diagnosed type 2 diabetes and
atherosclerotic cardiovascular disease, as there is no cardiovascular outcome study
for metformin in this population. In addition, further analyses of endpoint studies
with GLP-1-RA or SGLT2 inhibitors show that metformin use had no modulating effect
on the cardioprotective effect of these agents [22a]. However, there is no
definitive evidence to support the benefit of this recommendation as there have been
no controlled trials to date [23]. Strictly observing the contraindications for
metformin, one should therefore continue to start with metformin as primary therapy
and, if clinically indicated (manifest cardiovascular and renal diseases or patients
with a high cardiorenal risk (Part 1; Tab. 3, 4), start combination therapy with
SGLT2 inhibitors and/or GLP1-RA early (within 1–2 months).
Metformin is currently gaining great interest due to interesting pleiotropic effects
that influence changes at the epigenetic level and gene expression and are thus
potentially protective against carcinomas [24–32].
Metformin and COVID-19
A number of observational studies have shown that hospitalised COVID-19 infections
in
people with diabetes on pre-hospital metformin therapy, are associated with
significantly lower mortality [33,34]. This was confirmed in a recent meta-analysis,
which found a significant reduction in the odds ratio for mortality in COVID-19
patients with diabetes treated with metformin compared to those not treated with
metformin: OR 0.62; 95%-CI: 0.43–0.89 [35]. In some of the studies,
the confounding variables were not or only insufficiently taken into account. As
long as no controlled studies are available, metformin should be maintained [36,37]
or used with great caution in seriously-ill inpatients infected with COVID-19
because of the risk of lactic acidosis.
Summary of the therapy with metformin:
Kidney function must be checked regularly (every 3–6 months). Caveat:
metformin must be discontinued immediately if eGFR drops
to<30 ml/min.
Beware of diseases which increase the risk of lactic acidosis (e. g.,
acute deterioration of kidney function due to gastroenteritis, respiratory
insufficiency, acute diseases and infections or non-steroidal
anti-inflammatory drugs).
Caution when initiating therapy with ACE inhibitors or AT-1 receptor
blockers, diuretics, at the beginning of therapy with non-steroidal
anti-inflammatory drugs.
When administering x-ray contrast media, prior to interventional or major
surgical procedures, the patient should discontinue the use of metformin and
only restart taking it after 48 h, and only if the eGFR has not
deteriorated significantly postoperatively and the patient can eat
again.
In cardiovascular and renal high-risk individuals or people with manifest
cardiorenal disease, extreme caution is advised.
Sulfonylureas
Sulfonylureas have been used for decades because they effectively lower blood
glucose, are well tolerated and are inexpensive.
Due to their ability to increase insulin secretion by inhibiting the potassium
channels of the β-cells independently of glucose, they have the highest
hypoglycaemic potential of all oral antidiabetics, with the risk of sometimes severe
and prolonged hypoglycaemia, especially in older people with impaired renal function
and polypharmacy. Sulfonylureas are largely contraindicated with decreasing renal
function (eGFR<30 ml/min) with the exception of gliclazide
and gliquidone. Due to the high risk of severe hypoglycaemia in patients with
cardiovascular and renal complications, sulfonylureas should not be used in these
people. Sulfonylureas usually lead to moderate weight gain.
Favourable effects on microvascular endpoints were found in the UKPDS more than 6
years after treatment initiation for chlorpropramide and glibenclamide (mainly
reduced rate of photocoagulation). In the ADVANCE trial, gliclazide was found to
have positive effects on microvascular complications, mainly by reducing nephropathy
[38,39].
In the prospective, randomised, controlled CAROLINA study (mean observation time 6.3
years, approx. 3000 patients in each study arm; in both study arms 42% of
the participants already suffered from clinically manifest cardiovascular
complications at baseline), a comparison was made between linagliptin
(5 mg/d) and glimepiride (1–4 mg/d) with
regard to cardiovascular endpoints, hypoglycaemia and weight progression. There was
no difference when comparing the two study arms for 3P-MACE, 4P-MACE, all-cause and
cardiovascular death, and mortality with overall comparable HbA1c levels [40].
Weight progression was more favourable with linagliptin compared with glimepiride
(−1.54 kg), and rates of all, moderate and severe hypoglycemic
events requiring hospitalization were significantly lower with linagliptin compared
with glimepiride at all doses between 1 and 4 mg (1 mg: HR 0.23;
95% CI 0.21–0.26; p<0.0001, 2 mg: HR 0.18;
95% CI 0.15–0.21; p<0.0001, 3 mg: HR 0.15;
95% CI 0.08–0.29; p<0.0001, 4 mg: HR 0.07;
95% CI 0.02–0.31; p=0.0004). The authors concluded from the
CAROLINA trial data that there are no reasons, other than the lower cost of
glimepiride, to use glimepiride more preferentially than linagliptin in antidiabetic
therapy [40].
In several retrospective observational studies, in a large randomised pragmatic
trial, analyses from registry data and their meta-analyses, and Cochrane reviews,
sulfonylureas were shown to have no benefits in terms of macrovascular endpoints,
either in monotherapy or in combination therapy. Rather, increased cardiovascular
morbidity and mortality were described [24,41–49].
Repaglinide
Due to a decision of the Federal Joint Committee (G-BA), a comprehensive prescription
restriction for glinides was implemented as of 01.07.2016. The prescription
restriction reads: “The treatment of patients with renal insufficiency and a
creatinine clearance<25 ml/min with repaglinide is excluded
if no other oral antidiabetic agents are suitable and insulin therapy is not
indicated. Despite a detailed evidence-based statement (see also
http://www.deutsche-diabetes-gesellschaft.de/stellungnahmen)
to the G-BA and Federal Ministry of Health (BMG), the G-BA decision still
stands.
DPP-4 inhibitors
DPP-4 inhibitors are increasingly replacing therapy with sulfonylureas for reasons
of
a favourable safety profile, even in progressive renal insufficiency and a good
tolerability, which is particularly important for elderly people. Therapy adherence
and persistence with DDP-4 inhibitors (in 594,138 patients) were suboptimal despite
good tolerability: after 1 year of therapy, adherence was 56.9% (95%
CI 49.3–64.4) and after 2 years, 44.2% (95% CI
36.4–52.1) [50].
With the exception of linagliptin, the dosage of all DPP-4 inhibitors on the market
must be adjusted to the kidney function. In addition, DPP-4 inhibitors show largely
weight-neutral effects with similar antihyperglycaemic effects and low hypoglycaemic
rates. DPP-4 inhibitors seem to exert better metabolic control for longer than
sulfonylureas (observation period 104 weeks) [51].
The results of the CAROLINA study [40] (see section on sulfonylureas) were examined
in a real-world study with inclusion criteria as in the CAROLINA study in a
propensity score matching (PSM) [52]. There were 24 131 study pairs for linagliptin
and glimepiride to be analysed. As in the CAROLINA study, no differences were found
with regard to cardiovascular safety.
The results of the RCTs SAVOR TIMI 53® (saxagliptin [53]), EXAMINE®
(alogliptin [54]), TECOS® (sitagliptin [55]), CARMELINA®
(linagliptin [56,57]) on the effect of DPP-4 inhibitors on cardiovascular and renal
endpoints each show cardiovascular safety across all eGFR ranges
(<30 ml/min.->60 ml/min.) of the
investigated DPP-4 inhibitor in their primary endpoint, which was also confirmed in
extensive meta-analyses [58–64]. In a large US database, a 3-year follow-up
showed that DDP-4 inhibitors reduced the risk of the composite clinical endpoint
(eGFR decline>50%, end-stage renal failure or all-cause mortality)
more significantly compared with sulfonylureas but were less effective than GLP-1-RA
and SGLT2 inhibitors [65].
DPP-4 inhibitors are therefore effective antidiabetics with few side effects and can
be used very well as mono- and combination therapy if contraindications to the use
of metformin are present and there is a corresponding patient preference. Another
advantage is that DPP-4 inhibitors act largely weight-neutrally, hardly induce
hypoglycaemia and the use of linagliptin is not contraindicated even in
(pre)terminal renal insufficiency.
Hospitalisation for heart failure was not increased with the use of DPP-4 inhibitors,
except for saxagliptin (SAVOR TIMI 53). In a large meta-analysis on the risk of
DPP-4 inhibitors with regard to heart failure or hospitalisation for heart failure
including RCTs and observational studies, the authors concluded that the effect of
DPP-4 inhibitors on heart failure remains uncertain (due to relatively short
observation periods and overall weak data) [60]. A recent meta-analysis of
alogliptin, linagliptin, saxagliptin and sitagliptin showed a neutral effect on
myocardial infarction, stroke, heart failure (OR 1.06; 95% CI
0.96–1.18) and cardiovascular death [66].
Based on NAFLD and NASH studies with imaging and liver histology, DPP-4 inhibitors
showed no significant benefit in people with type 2 diabetes and NAFLD, in contrast
to GLP-1 RAs or SGLT2 inhibitors [67].
DPP-4 inhibitors in hospitalised patients
The use of DPP-4 inhibitors in people with type 2 diabetes and moderate, relatively
stable hyperglycaemia has been shown in a number of RCTs to have a good safety
profile, effective blood glucose-lowering and insulin savings with insulin
co-medication [68].
DPP-4 inhibitors may be able to slow down the over-activated immune system in people
with Sars-CoV-2 infection and thus contribute to a more favourable cardiovascular
outcome [69]. However, in the absence of randomised trials, the observational
studies available to date do not provide robust evidence to use DPP-4 inhibitors in
COVID-19 infection [70].
Safety aspects
In the meta-analysis of the 3 RCTs on DPP-4 inhibitors (SAVOR TIMI 53, EXAMINE and
TECOS), an increased incidence of acute pancreatitis was found compared with
corresponding controls (odds ratio 1.79; 95% CI 1.13–2.82;
p=0.013), although the absolute risk of acute pancreatitis was low overall
and only 0.13% higher in absolute terms under DPP-4 inhibitors [71]. A newer
meta-analysis found an association between DPP-4 inhibitors and the risk of acute
pancreatitis (OR 1.72; 95% CI 1.18–2.53). However, the authors
stated that the number of cases was too small to make a definite statement [72].
Therefore, great caution should be exercised when using DPP-4 inhibitors in people
with type 2 diabetes and a history or risk of pancreatitis.
A clear association between DPP-4 inhibitor therapy and bullous pemphigoid has
been seen in a number of cases [73].
It has also been shown that DPP-4 inhibitors are not associated with a higher rate
of
carcinoma [74].
DPP-4 inhibitors were associated with a significantly higher incidence of
inflammatory bowel disease in type 2 diabetes in a large population-based
study (HR 1.75; 95%- CI 1.22–2.49) [75]. This association was
highest 3–4 years after DPP-4 inhibitor therapy but became significantly
lower thereafter. The association started 2–4 years after the start of
therapy. However, in a recent meta-analysis of 13 studies, no association was found
between DPP-4 inhibitors and inflammatory bowel disease [76].
In combination with metformin, sitagliptin was certified by the G-BA as having a low
added benefit (BAnz AT 29.04.2019). However, neither in monotherapy nor in
combination therapy was saxagliptin granted an added benefit (BAnz AT 18.01.2017,
BAnz AT 13.03.2018 B2). The combination of linagliptin and empagliflozin was also
not considered to be of additional benefit (BAnz AT 24.12.2019 B3).
SGLT-2 inhibitors
SGLT-2 inhibitors (canagliflozin, dapagliflozin, empagliflozin, ertugliflozin) are
effective antihyperglycaemic substances in the treatment of type 2 diabetes in both
mono- and combination therapy with all other glucose-lowering drugs.
Their efficacy profile is favourable, also because the risk of hypoglycaemia is low,
patients lose weight and there is a clinically-relevant reduction in systolic blood
pressure [77–87].
Approved in Germany: Dapagliflozin, empagliflozin and ertugliflozin.
Not approved in Germany: Canagliflozin and sotagliflozin.
Safety aspects
However, there is a significantly increased risk of genital infections with
SGLT-2 inhibitors in RCTs [88,89]. The relative risk of SGLT-2 inhibitors for
genital infections was more than 3 times higher than placebo (RR 3.37; 95%
CI 2.89–3.93) and almost 4 times higher than an active comparator (RR 3.89;
95% CI 3.14–4.82). By contrast, the risk of urinary tract infections
was not significantly increased by SGLT-2 inhibitors compared to placebo (RR 1.03;
95% CI 0.96–1.11) or an active comparator therapy (RR 1.08;
95% CI 0.93–1.25). In a large retrospective cohort study of a US
database, an approximately 3-fold higher risk of genital infection was found with
SGLT2 inhibitors compared to DPP-4 inhibitors, starting in the first 4 weeks of
therapy and as long as therapy was continued [90]. Comparable results were also seen
in the real-world analysis of people with diabetes at a relatively advanced age
(71.8±5 years) [91]. Patients with a history of genital infections were
particularly at risk of infection when taking SGLT2 inhibitors [92].
A necrotizing fasciitis of the perineum and genitals (Fournier gangrene) is a
very rare, severe infection with the need for immediate antibiotic and usually
surgical intervention. Diabetes is one of the risk factors. With the introduction
of
SGLT-2 inhibitor therapy, a few cases of Fournier gangrene under SGLT-2 inhibitor
therapy were described. A Red Hand letter was published in consultation with the
European Medicines Agency (EMA) and the Federal Institute for Drugs and Medical
Products/Bundesinstitut für Arzneimittel und Medizinprodukte (BfArM)
to clarify the ‘Risk of a Fournier gangrene (necrotizing fasciitis of the
perineum) when using SGLT-2 inhibitors (sodium glucose cotransporter-2
inhibitors)”.
A recently published real-world study investigated the incidence of Fournier gangrene
in patients after starting therapy with SGLT2 inhibitors (n=93,197) or with
DPP-4 inhibitors. No increased risk of this gangrene was found with SGLT2 inhibitor
therapy compared with persons with DPP-4 inhibitor treatment [93].
In a recent meta-analysis of all randomised controlled trials of SGLT2 inhibitors
(n=84) in patients with type 2 diabetes, no differences were found in the
risk of Fournier gangrene, abscess, cellulitis or erysipelas with SGLT2 inhibitors
vs. comparators or placebo. The rate of Fournier gangrene was very low at 3.53 per
100 000 patient-years [94].
The European Medicines Agency (EMA) has started a review process to investigate
whether canagliflozin therapy leads to an increased rate of amputations
(mostly toes): In 2016, the EMA’s Pharmacovigilance Risk Assessment
Committee (PRAC) extended the review to include dapagliflozin and empagliflozin
[95].
The canagliflozin CANVAS programme [96] trials confirmed the suggestion of a higher
risk of amputations (predominantly toe and metatarsal areas) with canagliflozin
compared with placebo (event rate 6.3 vs. 3.4 persons per 1000 patient-years; HR
1.97; 95% CI 1.41–2.75; p<0.001). For SGLT2 inhibitors,
higher rates of amputations are also found in RCTs in pharmacovigilance reports
[97]. In contrast, current studies and searches did not find higher amputation rates
with dapagliflozin [98] and empagliflozin [99]. The large CREDENCE study with
canagliflozin also found no signal for an increased amputation rate [100]. The
meta-analysis by Huang et al. [101] also found no evidence that SGLT2 inhibitors
were associated with an increased risk of amputation.
The FDA has also issued a warning about an increased fracture risk due to
reduced bone density under canagliflozin
(http://www.fda.gov/Drugs/DrugSafety/ucm461449.htm).
Indeed, the fracture event rate was significantly higher with canagliflozin compared
with placebo: 15.4 vs. 11.9 per 1000 patient-years (p=0.02) [102]. However,
careful elaboration of the CANVAS and CANVAS-R data showed significant heterogeneity
of fracture risk in both studies: in the CANVAS study (n=4330: HR 1.55;
95% CI 1.21–1.97) the risk was significantly increased, whereas this
could not be demonstrated in the CANVAS-R study (n=5812: HR 0.86;
95% CI 0.62–1.19) [103]. In the recently published large RCT
(CREDENCE study) with canagliflozin, there was also no signal for an increased
fracture risk [104].
A recent fracture analysis of people with type 2 diabetes (n≥12 000) treated
with empagliflozin (pooled data from placebo-controlled trials and a head-to-head
trial vs. glimepiride) found no significantly increased rate of fractures [105].
Numerous meta-analyses also showed no significant increase in fracture rates with
SGLT2 inhibitor therapy [106–108].
Real-world studies and analyses of health care data also showed no increased fracture
rate with SGLT2 inhibitor therapy [109].
When SGLT2 inhibitors were used, ketoacidosis was occasionally observed in
people with type 2 diabetes [110, 111]. The SGLT2 inhibitor manufacturers in Germany
already informed physicians and pharmacists about the situation in 2015.
A comprehensive analysis of all reports of ketoacidosis cases with a possible
connection to SGLT2 inhibitors that were listed in the US Food and Drug
Administration Adverse Event Reporting System (FAERS) between January 2014 and
October 2016 has been published [112]. They found a Proportional Reporting Ratio
(PPR) of 7.9 (95% CI 7.5–8.4). The PPR is the ratio of spontaneous
reports for a specific drug (in this case SGLT2 inhibitors) associated with a
specific adverse event (=ketoacidosis) divided by the corresponding ratio
for all or some other drugs with this adverse event. However, the PPR does not
describe a relative risk, i. e., the real risk for ketoacidosis. Detailed
analysis of 2397 reports of ketoacidosis in FAERS showed a predominance in people
with type 1 diabetes, in women, across a wide age and body weight range, and high
variability in the duration of SGLT2 inhibitor therapy. 37 people (1.54%)
died from ketoacidosis. In the large randomised controlled trials of SGLT2
inhibitors, the risk of ketoacidosis was significantly increased with SGLT2
inhibitors in type 2 diabetes but was less than 1%. The meta-analysis
published last year (39 RCTs with 60 580 patients) again confirmed a statistically
significant increased rate of ketoacidosis with SGLT2 inhibitors (0.18%)
compared to controls (0.09%) with an OR of 2.13 (95% CI
1.38–3.27). Older age and longer use of SGLT2 inhibitors played a role
[113].
Normoglycaemia or mild hyperglycaemia does not exclude a ketoacidosis with SGLT-2
inhibitors. Risk factors for the development of a (euglycaemic) ketoacidosis with
SGLT-2 inhibitors included a rapid and significant reduction of the insulin dose,
severe dehydration, and alcohol consumption; almost all patients with ketoacidosis
were in a catabolic state (operations, myocardial infarction, severe infections,
long fasting, excessive physical strain).
Therefore, the German Diabetes Association (DDG) recommends that the following be
considered when dealing with SGLT-2 inhibitors:
Discontinuation of SGLT2 inhibitors at least 3 days (=about 5
half-life times equivalent to 11–13 hours) before major
elective surgery [114, 115], immediate pause of SGLT2 inhibitor therapy in
emergencies and acute illness,
Caution during ongoing insulin therapy (avoid significant reduction or
discontinuation of insulin therapy),
Avoidance of prolonged periods of fasting, ketogenic/extremely
low-carbohydrate diets and excessive alcohol consumption.
The combination of SGLT-2 inhibitors with metformin increases the risk of
ketoacidosis [116] and
If symptoms are present, consider the possibility of euglycaemic ketoacidosis
and initiate the appropriate diagnostic procedures (plasma glucose and
ketones in blood, possibly also necessary venous blood gas).
Effects on cardiovascular and renal endpoints
Dapagliflozin
The DECLARE-TIMI 58 study with dapagliflozin [117] included 6974 patients
(40.6%) with known cardiovascular diseases and 10 186 (59.4%) with
multiple risk factors for arteriosclerotic cardiovascular diseases. The mean
follow-up of the patients was 4.2 years. A total of 3962 patients stopped the study
prematurely (=5.7% per year): 1811 of the 8574 patients
(21.1%) on dapagliflozin and 2151 of 8569 (25.1%) in the control
group. Dapagliflozin resulted in a significantly lower hospitalization rate for
heart failure compared to placebo (HR 0.73; 95% CI 0.61–0.88). There
was no difference between the dapagliflozin group and the placebo group in the rate
of 3P-MACE (8.8 vs. 9.4%; HR 0.93; 95% CI 0.84–1.03;
p=0.17), cardiovascular morality (HR 0.98, 95% CI 0.82–1.17)
and all-cause mortality (HR 0.93, 95% CI 0.82–1.04). In the renal
composite secondary endpoint (≥40% reduction in eGFR,
newly-developed terminal renal failure or death of renal or cardiac genesis),
dapagliflozin led to a significant reduction in renal endpoints (HR 0.76;
95% CI 0.67–0.87).
Extensive sub-analyses of the DECLARE-TIMI 58 population confirmed the beneficial
effects of dapagliflozin on the development and progression of renal [118] and
cardiovascular endpoints [119, 120].
In the DAPA-HF study, at a median follow-up of 18.2 months of 2373 study
participants, the primary composite endpoint of worsening heart failure
(hospitalisation or intravenous therapy for heart failure) or cardiovascular death
was met in 386 (16.3%) in the dapagliflozin group and 502 (21.2%) in
the placebo group: HR 0.74, 95% CI 0.65–0.85; p<0.001. The
primary endpoints were comparable between people with (42% of the study
population) and without diabetes (HR 0.75, 95% CI 0.63–0.90 vs HR
0.73, 95% CI 0.60–0.88). Dapagliflozin reduced numerous secondary
endpoints such as total number of hospitalisations for heart failure (first and
recurrent), reduction in all-cause mortality and improvement in quality of life
[121].
In the recently-published multicentre RCT DAPA-CKD [122], patients (n=4304;
68% of patients had type 2 diabetes) with an albumin:creatinine ratio of
200–5000 mg/g and an eGFR of
25–75 mL/min were randomised 1:1 to dapagliflozin
(10 mg/d) or placebo. The median follow-up was 2.4 years. The
primary endpoint was composed of a decrease in eGFR of more than 50%, ESRD,
renal or cardiovascular death. Secondary endpoints were the primary endpoint other
than cardiovascular death, a composite endpoint of cardiovascular death or
hospitalisation for heart failure and all-cause mortality. The relative risk
reduction of the primary endpoint was consistent with dapagliflozin between patients
with diabetes (HR 0.64, 95% CI 0.52–0.79) and patients without
diabetes (HR 0.50, 0.35–0.72). Comparable results were seen for the renal
secondary endpoint (0.57 [0.45–0.73] vs 0.51 [0.34–0.75]),
cardiovascular death or hospitalisation for heart failure (0.70 [0.53–0.92]
vs 0.79 [0.40–1.55]) and all-cause mortality (0.74 [0.56–0.98] vs
0.52 [0.29–0.93]).
The 3 SGLT2 inhibitors empagliflozin (EMPA-REG OUTCOME), canagliflozin (CANVAS
programme and CREDENCE trial) and dapagliflozin (DECLARE-TIMI 58) with a total of
38
723 study participants resulted in the meta-analysis by Neuen et al. [123] resulted
in a significant risk reduction for dialysis, kidney transplantation or mortality
due to renal failure (RR 0.67, 95% CI 0.52–0.86, p=0.0019).
SGLT2 inhibitors also reduced the risk of end-stage renal failure (RR 0.65,
95% CI 0.53–0.81, p<0.0001) and acute renal failure (RR
0.75, 95% CI 0.66–0.85, p<0.0001) across all trials. There
was a clear advantage of all 3 SGLT2 inhibitors across all eGFR subgroups and also
independent of the degree of albuminuria at baseline. A recent meta-analysis of 11
trials involving 93 502 patients showed similar beneficial effects of SGLT2
inhibitors in older people with type 2 diabetes (>65 years) on MACE (HR
0.90; 95% CI 0.83–0.98), hospitalisation for heart failure (HR 0.62;
95% CI 0.51–0.76) and composite renal endpoint (HR 0.57; 95%
CI 0.43–0.77) [124]. In the meta-analysis by Bae et al. [125] of 17 trials
involving 87 263 patients, SGLT2 inhibitors significantly reduced renal risks such
as microalbuminuria (OR 0.64; 95% CI 0.41–0.93), macroalbuminuria
(OR 0.48; 95% CI 0.24–0.72), worsening renal function (OR 0.65;
95% CI 0.44–0.91) and end-stage renal failure (OR 0.65; 95%
CI 0.46–0.98) compared with placebo. In the most comprehensive meta-analysis
of 736 trials with a total of 421 346 patients, SGLT inhibitors led to robust
significant reductions in all-cause and cardiovascular mortality, non-fatal
myocardial infarctions, and renal failure, but also, as expected, increased genital
infections. SGLT2 inhibitors had less robust evidence on weight reduction. Weak or
no evidence was found for positive effects of SGLT1 inhibitors on amputations,
retinopathy or loss of sight, neuropathic pain, and health-related quality of life.
The absolute benefit of SGLT2 inhibitors was found across a broad spectrum in
patients with low and high cardiovascular and renal outcomes [126].
Empagliflozin
The effects of SGLT-2 inhibitor therapy on clinical endpoints were investigated for
empagliflozin in a large RCT published in 2015 (EMPA-REG OUTCOME study [127]).
Patients with type 2 diabetes and already manifested cardiovascular diseases showed
fewer cardiovascular events (10.5 vs. 12.1%; HR 0.86; 95% CI
0.74–0.99; p<0.04 for superiority) during an observation period of
3.1 years on average with empagliflozin compared to placebo. There was no difference
in the rate of myocardial infarction and stroke, but a significantly lower event
rate for cardiovascular mortality (3.7 vs. 4.1%; HR 0.62; 95% CI
0.49–0.77; HR 0.49-p<0.001); for all-cause mortality (5.7 vs.
8.3%; HR 0.68; 95% CI 0.57–0.82; p<0.001) and
hospitalization for heart failure (2.7 vs. 4.1%; HR 0.65; 95% CI
0.50–0.85; p=0.002). The risk of cardiovascular events was greater
when cardiovascular risk factors were less well controlled at baseline. However, the
cardioprotective effect of empagliflozin was significantly associated, independent
of the degree of risk factor control [128]. Analysis of recurrent events (including
outcome of coronary events, hospitalisation for heart failure, hospitalisation for
other reasons) and cardiovascular mortality showed significant reductions with
empagliflozin compared to placebo [129].
Further analyses of the EMPA-REG OUTCOME study [130] showed that empagliflozin slows
the development and progression of nephropathy in patients with an eGFR initial
of≥30 ml/min: beginning or progression of nephropathy with
empagliflozin compared to standard therapy (12.7 vs. 18.8%; HR 0.61;
95% CI 0.53–0.70; p<0.001).
The post-hoc renal endpoint (doubling of S-creatinine, renal replacement therapy,
or
death from kidney disease) was significantly lower for empagliflozin compared to
placebo (HR 0.54; 95% CI 0.40–0.75; p<0.001). In an analysis
of the short-term and long-term effects (164 weeks) of empagliflozin on albumin
excretion, a significant reduction of 22% on average in the microalbuminuria
group and 29% in the macroalbuminuria cohort was observed [131],
irrespective of the level of initial albuminuria. Based on 1738 participants in the
EMPA-REG-OUTCOME trial with a history of coronary artery bypass grafting at
baseline, empagliflozin reduced the risk of all-cause mortality by 43%,
cardiovascular mortality by 48%, hospitalisation rate for heart failure by
50% and nephropathy (onset or worsening) by 35% [132].
The EMPEROR-REDUCED study [133] included 3730 patients (50% with diabetes)
with functional class II, III or IV heart failure and an ejection
fraction≤40% were treated with either empagliflozin
(10 mg/d) or placebo (1:1) in addition to guideline-guided heart
failure therapy. The median duration of the study was 16 months. With empagliflozin,
the primary composite endpoint (cardiovascular death or hospitalisation for
worsening of heart failure) occurred in 19.4% of patients versus
24.7% with placebo. The hazard ratio was 0.75; 95% CI
0.65–0.86; p<0.001). The effect of empagliflozin on the primary
endpoint was independent of whether patients had diabetes or not. The total number
of hospitalisations was lower in the empagliflozin compared with the placebo group
(HR 0.70; 95% CI 0.58–0.85; p<0.001). The annual decline in
eGFR was lower in the empagliflozin vs. placebo group (−0.55 vs.
− 2.28 ml/min./year; p<0.001). The
rate of serious renal complications was also lower with empagliflozin: HR 0.50
(0.32–0.77).
For the SGLT2 inhibitor empagliflozin - as well as for the other gliflozines -
clinically very relevant effects on all-cause mortality as well as on cardiovascular
and renal endpoints in appropriate risk populations have been documented and
confirmed in meta-analyses [134–137].
The underlying mechanisms of cardiac and renal protection of SGLT2 inhibitors are
the
subject of extensive studies [138–142].
In the 2016 benefit assessment by the Federal Joint Committee (Gemeinsamen
Bundesausschuss - G-BA), empagliflozin was certified as having evidence of
considerable additional benefit in patients with type 2 diabetes with manifest
cardiovascular disease in combination therapy with metformin
(https://www.g-ba.de/downloads/39–2612%20694/2016–09–01_AM-RL-XII_Empagliflo
zin_D-214_BAnz.pdf). Accordingly, this additional benefit was included in the new
edition of the disease management programme for type 2 diabetes in 2017 [143].
Ertugliflozin
The cardiovascular safety of ertugliflozin was investigated in the VERTIS-CV study.
The study design and also the characteristics of the study population at baseline
were similar to those of the EMPA-REG-OUTCOME study, particularly in relation to
pre-existing cardiovascular disease [144]. Approximately 2750 patients were included
in each of the 3 study arms (standard therapy/placebo; 5 mg
ertugliflozin, 15 mg ertugliflozin daily) and were followed for
approximately 3.5 years. MACE was slightly lower in ertugliflozin groups compared
with the placebo group (HR 0.97; 95% CI 0.85–1.11; p<0.001
for non-inferiority). Data on cardiovascular death or hospitalisation for heart
failure (ertugliflozin vs. placebo: 8.1 vs. 9.1% (HR 0.88; 95% CI
0.75–1.03; p=0.11 for superiority), the analyses for cardiovascular
death (HR 0.92; 95% CI 0.77–1.11), death from renal causes, renal
replacement therapy or doubling of serum creatinine (HR 0.81; CI 0.63–1.04)
were not significant. Amputations were reported in 2% with ertugliflozin
(5 mg) therapy and in 1.6% with 15 mg dose. The amputation
rate with placebo was also 1.6% [145]. In a post-hoc analysis of the VERTIS
MET [146] and VERTIS SU [147] trials, ertugliflozin reduced eGFR in the first 6
weeks but returned to baseline after 104 weeks and therefore resulted in
preservation of renal function. The eGFR was slightly higher at both ertugliflozin
doses (5 and 15 mg) than in patients who did not receive ertugliflozin.
Ertugliflozin significantly reduced albumin excretion rates by 30 and 38% in
people who had albuminuria at baseline (21%) [148]. Another analysis of the
VERTIS-CV trial showed that at a mean follow-up of 3.5 years, the exploratory
composite endpoint (time to doubling of serum creatinine, dialysis (kidney
transplantation or renal death) was significantly reduced with ertugliflozin
compared with placebo (HR 0.66; 95% CI 0.50–0.88). Renal function
and albumin excretion rates were stabilised [149].
In the VERTIS programme, a number of studies with ertugliflozin were published that
analysed combination therapies with metformin, metformin plus sitagliptin, insulin
or sulfonylureas, which were recently summarised in a review [150].
Ertugliflozin is only approved in Germany in a fixed combination with sitagliptin
(VERTIS-Factorial study). According to the decision of the G-BA of 01.11.2018, there
is no additional benefit of this fixed combination. The G-BA also certified no
additional benefit for the combination of linagliptin and empagliflozin (BAnz AT
24.12.2019 B3).
Canagliflozin
Recent outcome RCT data on canagliflozin [96] (CANVAS programme) show a significant
reduction in composite endpoint (cardiovascular death, non-fatal myocardial
infarction and stroke) with canagliflozin compared with placebo of 14% (HR
0.86; 95% CI 0.75–0.97), decrease in hospitalisation rate due to
heart failure of 33% (HR 0.67; 95% CI 0.52–0.87) and renal
outcome data with a reduction in the progression of albuminuria by 27% (HR
0.73; 95% CI 0.67–0.79) and composite endpoint (40%
reduction in eGFR, renal replacement therapy, renal death) by 40% (HR 0.60;
95% CI 0.47–0.77) [100]. Another large RCT (CREDENCE trial) was
conducted with canagliflozin in relation to a primary combined renal endpoint [104].
Patients already had renal insufficiency at randomisation, significant proteinuria
and had to be already treated with an ACE inhibitor or AT blocker. Canagliflozin
(100 mg per day) was shown to significantly reduce the relative risk of the
composite endpoint (dialysis, transplantation or sustained
eGFR<15 ml/min), doubling of serum creatinine, death from
renal or cardiovascular causes (HR 0.70, 95% CI 0.59–0.82;
p=0.00 001).
In the recently published post hoc analysis of the CANVAS programme and the CREDENCE
trial, canagliflozin was not associated with a reduction in myocardial infarction
in
the study populations [151].
Canagliflozin is currently not available on the German market despite positive
patient-relevant endpoints.
Sotagliflozin
Sotagliflozin is a dual SGLT1 and SGLT2 inhibitor. Two large studies have been
published so far for the treatment of type 2 diabetes. In the SOLOIST-WHF trial,
people with type 2 diabetes and decompensated heart failure were studied with
sotagliflozin (n=608) or placebo (n=614) for a median of 9 months.
Mean ejection fraction (EF) was 35% and baseline heart failure therapy was
the same in both groups. There was a significant reduction in the composite primary
endpoint (cardiovascular death and hospitalisation or acute hospitalisation for
heart failure) with sotagliflozin compared with placebo: hazard ratio (HR) 0.67,
95% CI 0.52–0.85, p<0.001). As the study had to be
discontinued due to COVID-19 and a lack of financial support, the calculated event
rates were not achieved, so that the data of this study are not sufficiently robust
overall [152].
In the randomised controlled SCORED trial [153], 10 584 patients with type 2 diabetes
and renal insufficiency (eGFR 25–60 ml/min.) and
cardiovascular risk factors were randomised 1:1 (sotagliflozin:placebo). The median
follow-up was 16 months. The primary endpoint was changed during the study to a
composite endpoint (all-cause cardiovascular mortality, hospitalisation for or acute
care for heart failure). The primary endpoint was significantly lower with
sotagliflozin compared to placebo: hazard ratio 0.74; 95% CI
0.63–0.88; p<0.001). This study also had to be stopped early for
financial reasons. Sotagliflozin is currently only approved for combination therapy
with insulin in people with type 1 diabetes.
GLP-1 receptor agonists (RAs)
GLP-1-RAs are antidiabetic drugs for the subcutaneous or oral therapy of type 2
diabetes. They can on average lower plasma glucose more than classic oral
antidiabetics and also have blood pressure-lowering (slight), weight-reducing [154]
and specific cardio- and renal protective (see below) effects. If the individual
therapeutic objective is not achieved, GLP-1-RAs are useful combination partners to
metformin, other OADs (except DPP-4 inhibitors) and/or basal insulin.
GLP-1-RAs themselves have a low hypoglycaemic risk.
Human GLP-1-RAs
Available in Germany: dulaglutide, liraglutide, semaglutide
Not available in Germany: albiglutide
Exendin-based GLP-1 RAs
Approved in Germany: exenatide, lixisenatide (only in fixed combination with insulin
glargine).
Not available in Germany: efpeglenatide
Dulaglutide
In the AWARD trial programme, dulaglutide was shown to be effective in lowering blood
glucose and weight, and for a low incidence of hypoglycaemia when used as
monotherapy and in combination with prandial and basal insulin. Patients with
various degrees of chronic renal insufficiency were also included [155]. The
multi-centre (371 study centres in 24 countries), randomized, double-blind
placebo-controlled study on the cardiorenal effects of dulaglutide therapy (REWIND
study; 1.5 mg s.c. weekly) was recently published [156, 157].
Included were 9901 patients with type 2 diabetes (mean age 66 years, average HbA1c
55,2 mmol/mol; 7,2%). This study differs from the previously
published studies on the cardiovascular and renal outcome under GLP-1-RA in the
following important points: Longer observational period (mean 5.4 years),
69% of the study participants had cardiovascular risk factors, but no
clinically manifested cardiovascular pre-illnesses and the ratio between women and
men was fairly balanced (46% women). Compared to placebo, dulaglutide was
able to reduce the mean HbA1c baseline value of 7.2% over the entire study
(HbA1c: –0.46% for dulaglutide,+0.16% for placebo;
body weight: –2.95 kg dulaglutide, –1.49 kg
placebo). In addition, dulaglutide showed a reduction of the secondary combined
microvascular endpoint (HR 0.87; 95% CI 0.79–0.95), with this
reduction predominantly affecting the renal outcome (HR 0.85; 95% CI
0.77–0.93; p=0.0004). The primary endpoint 3P-MACE was significantly
lower with dulaglutide (HR 0.88; 95% CI 0.79–0.99; p=0.026),
as was the risk of non-fatal stroke (HR 0.76; 95% CI 0.61–0.95;
p=0.017). No risk reductions were found for the following endpoints:
non-fatal and fatal myocardial infarction, fatal stroke, cardiovascular death,
all-cause mortality, and hospitalization for heart failure. Compared to placebo,
dulaglutide did not show any differences with regard to relevant side effects:
Cancer (pancreatic, medullary thyroid carcinoma, other thyroid carcinomas), acute
pancreatitis or pancreatic enzyme elevations, liver diseases, cardiac arrhythmias
and hypoglycaemic rate.
In an explorative analysis of the REWIND data [157] renal outcome data concerning
dulaglutide, a significant risk reduction for the summarized renal endpoint (new
macroalbuminuria, eGFR reduction of≥30% or chronic renal replacement
therapy; HR 0.85; 95% CI 0.77–0.93; p=0.0004) was determined
with the clearest effect with respect to the macroalbuminuria component (HR 0.77;
95% CI 0.68–0.87; p<0.0001).
In a post-hoc analysis of the REWIND trial, the incidence of MACE (cardiovascular
death, non-fatal myocardial infarction or non-fatal stroke) or non-cardiovascular
death was 35.8 per 1000 person-years in the dulaglutide group and 40.3 per 1000
person years in the placebo group (HR 0.90, 95% CI 0.82–0.98,
p=0.020). The incidence data on more complex MACE (MACE plus heart failure,
unstable angina or revascularisation) were more impressive: dulaglutide vs. placebo
67.1 vs. 74.7 per 1000 person years: HR 0.93 (95% CI 0.87–0.99)
p=0.023 [158]. In the G-BA decision of 16.07.2020, dulaglutide was assigned
an indication for a small additional benefit in people with type 2 diabetes in whom
diet and exercise and treatment with insulin (with or without another antidiabetic
drug) do not sufficiently control blood glucose, both in patients without renal
insufficiency and in patients with moderate or severe renal insufficiency (CKD
stages 3 and 4).
Liraglutide
In a randomised trial in obese patients, liraglutide (3 mg/d)
resulted in greater weight loss than placebo in all intensively treated patients
compared to physical activity alone: 8 weeks after a low-calorie diet resulted in
a
weight loss of 13.1 kg. At the end of the study (after one year), weight
loss with increased physical activity was − 4.1 kg
(95% CI − 7.8 to − 0.4; p=0.03); in
the liraglutide group − 6.8 kg (95% CI
− 10.4 to − 3.1; p<0.001); in the
combination physical activity plus liraglutide − 9.5 kg
(95% CI − 13.1 to − 5.9; p<0.001).
The combination therapy also resulted in a 3.9% reduction in body fat mass,
which was approximately 2-fold higher than in the physical activity group
(−1.7%; 95% CI − 3.2 to
− 0.2; p=0.02) and in the liraglutide group alone
(−1.9%; 95% CI − 3.3 to
− 0.5; p=0.009) [159]. For the GLP-1 receptor agonist (RA)
liraglutide, the RCT (LEADER trial) showed positive effects on clinically-relevant
endpoints [160]. The median follow-up of the 9340 patients was 3.8 years. The
composite primary endpoint (first event for cardiovascular death, non-fatal
myocardial infarction, non-fatal stroke) was significantly lower with liraglutide
compared with placebo (13 vs. 14.9%; HR 0.87; 95% CI
0.78–0.97; p<0.001 for non-inferiority and p=0.01 for
superiority). Fewer patients died from cardiovascular causes (4.7 vs. 6.0%;
HR 0.78; 95% CI 0.66–0.93; p=0.007). All-cause mortality was
also lower with liraglutide (8.2 vs. 9.6%; HR 0.85; 95% CI
0.74–0.97; p=0.02). Thus, for the first time, a positive effect on
patient-relevant outcomes could also be demonstrated for a GLP-1 RA in an RCT.
A sub-analysis of the LEADER study population showed that 72% of patients had
vascular disease at baseline. 23% of this subpopulation had polyvascular
disease and 77% had monovascular disease. Liraglutide led to a reduction in
MACE at 54-month follow-up: in polyvascular disease (HR 0.82; 95% CI
0.66–1.02) and in monovascular disease (HR 0.82; 95% CI
0.71–0.95) compared with placebo. No positive effects of liraglutide were
found in patients without vascular complications [161]. The analysis by Marso et al.
[162], which demonstrated a reduction in myocardial infarctions with liraglutide in
patients at high vascular risk, points in the same direction. In the meta-analysis
published by Duan et al. in 2019 [163], patients in the liraglutide group compared
with controls were found to have lower risks of: MACE (RR 0.89, 95% CI
0.82–0.96, p=0.002), acute myocardial infarction (RR=0.85,
95% CI 0.74–0.99, p=0.036), all-cause mortality (RR 0.84,
95% CI 0.74–0.96, p=0.009) and cardiovascular death (RR
0.77, 95% CI 0.65–0.91, p=0.002). However, the incidence of
stroke was not reduced in the liraglutide group (RR 0.86, 95% CI
0.70–1.04, p=0.124).
In the analysis of secondary renal endpoints in the LEADER study, liraglutide was
associated with a lower rate of development and progression of the renal composite
endpoint (HR 0.78; 95% CI 0.67–0.92; p=0.003) and
persistence of macroalbuminuria (HR 0.74; 95% CI 0.60–0.91;
p=0.004) compared with placebo [164].
In its decision of 17.01.2019 (BAnz AT 22.03.2019 B5), the G-BA granted liraglutide
an added benefit and included it in the structured treatment programmes for type 2
diabetes.
The meta-analysis by Kristensen et al. [165] showed a significant reduction in MACE
of 12% (HR 0.88; 95% CI 0.82–0.94; p<0.0001) with
GLP-1-RA. The hazard ratios were 0.88 (95% CI 0.81–0.96;
p=0.003) for death from cardiovascular events, 0.84 (95% CI
0.76–0.93; p<0.0001) for fatal and non-fatal stroke, and 0.91
(95% CI 0.84–1.00; p=0.043) for non-fatal and fatal
myocardial infarction. GLP- 1-RA resulted in a 12% reduction in all-cause
mortality (HR 0.88; 95% CI 0.83–0.95; p=0.001) and a
9% reduction in hospitalisation for heart failure (HR 0.91; 95% CI
0.83–0.99; p=0.028). The composite renal endpoint (development of
new macroalbuminuria, reduction in eGFR, progression to ESRD) decreased by
17% (HR 0.83; 95% CI 0.78–0.89; p<0.0001), mainly
due to the reduction in albuminuria. No increased risk of hypoglycaemia,
pancreatitis or pancreatic cancer was reported with GLP-1 RA.
The very detailed and critical meta-analysis by Liu et al. [166] also came to a
comparable conclusion. All-cause mortality was slightly lower under GLP-1 RAs
compared to control therapies: OR 0.89 (95%-KI 0.80–0.98).
The association of GLP-2 RAs with renal events under real-world conditions was
analysed in a large Scandinavian study [167]. 38 731 users of GLP-1 RAs (liraglutide
92.5%, exenatide 6.2%, lixisenatide 0.7%, dulaglutide
0.6%) were studied 1:1 in a propensity-matched control group taking DPP-4
inhibitors. The primary composite endpoint (renal replacement therapy, renal-related
death and hospitalisation for renal complications) was significantly lower with
GLP-1-RA than with DPP-4 inhibitor therapy: HR 0.76 (95% CI
0.68–0.85). In particular, renal replacement therapy (HR 0.73, 95%
CI 0.62–0.87) and hospitalisation rates (HR 0.73, 95% CI
0.65–0.83) were significantly lower with GLP-1-RA [167].
Semaglutide
Semaglutide s. c.
Semaglutide 1×weekly s. c. showed a greater HbA1c reduction
(−0.4%) and weight loss (−2.5 kg) compared to other
GLP-1 RAs [168].
In the STEP-1 study with semaglutide (1×weekly s. c.), a mean weight loss of
− 14.9% was observed in the observation period of 68 weeks
compared to placebo of only − 2.4%. The difference in weight
loss of − 12.4% was highly significant. More patients in the
semaglutide group than in the placebo group achieved weight losses
of≥5% (86.4 vs. 31.5%),≥10% (69.1 vs.
12.0%) and≥15% (50.5 vs. 4.9%), all of which were
highly significant with a p=0.001 [169]. The STEP 3 and STEP 4 trials showed
similar favourable effects of semaglutide on weight progression [170, 171].
In the SUSTAIN-6 trial, cardiovascular benefit was demonstrated by significant
reduction in the primary endpoint 3P-MACE compared to the control group. In patients
with a high cardiovascular risk, there was a significant risk reduction (HR 0.74;
95% CI 0.58–0.95) for the primary endpoint (cardiovascular death,
non-fatal myocardial infarction or non-fatal stroke) in the semaglutide group
compared to placebo [172]. In the recently published post-hoc analysis of the
SUSTAIN-6 study, semaglutide 1×weekly s.c. vs. placebo was found to reduce
the risk of MACE in all study participants regardless of sex, age or cardiovascular
risk profile at baseline [173].
Oral semaglutide
In the PIONEER-6 trial of oral semaglutide 1×daily (n=3183 patients,
84.7%>50 years with cardiovascular or chronic renal complications;
mean observation time 15.9 months) the following results were found: MACE was found
in 3.8% in the oral semaglutide and 4.8% in the placebo group (HR
0.79; 95% CI 0.57–1.11; p<0.001 for non-inferiority);
cardiovascular death (HR 0.49; 95% CI 0.27–0.92); non-fatal
myocardial infarction (HR 1.18; 95% CI 0.73–1.90); non-fatal stroke
(HR 0.74; 95% CI 0.35–1.57); all-cause mortality (HR 0.51;
95% CI 0.31–0.84) [174]. In the meta-analysis published in 2020,
oral semaglutide was shown to reduce the risk of all-cause mortality (OR 0.58;
95% CI 0.37–0.92) and cardiovascular mortality (OR 0.55; 95%
CI 0.31–0.98) compared with placebo. However, it showed a neutral effect
with regard to myocardial infarction, stroke and severe hypoglycaemia [175].
In a combined post-hoc analysis of the two cardiovascular outcome trials SUSTAIN 6
and PIONEER 6, the effect of semaglutide was analysed in patients with a continuum
of initial cardiovascular risk. Thereby, semaglutide showed a significant absolute
and relative risk reduction of MACE (cardiovascular death, non-fatal myocardial
infarction, non-fatal stroke) across the spectrum of cardiovascular risk compared
to
comparator therapies. This was also found for the individual components of MACE
[176].
However, in the recent re-analysis of the SUSTAIN 6 and PIONEER 6 studies [177], the
authors placed the analyses in a broader context to the results of the other studies
SUSTAIN 1–5 and PIONEER 1–5, 7–8. The hazard ratio for MACE
was 0.85 with a wide confidence interval (95% CI: 0.55–1.33) because
of the low event rates in most studies.
Treatment with GLP-1 RAs or SGLT2 inhibitors was associated with significantly lower
all-cause mortality compared with DPP-4 inhibitors or other antidiabetic drugs or
no
therapy in the meta-analysis by Zheng SL et al. (HR 0.88; 95% CI
0.81–0.94 and/or HR 0.80; 95% CI 0.71–0.89,
respectively). Similar data were also found for cardiovascular mortality as well as
myocardial infarction and heart failure compared to the control groups [178].
In the meta-analysis of the GLP-1 RAs exenatide, liraglutide, lixisenatide,
albiglutide, dulaglutide and semaglutide published in 2017, there was a significant
reduction in the incidence of nephropathy compared with other antidiabetic drugs (OR
0.74; 95% CI 0.60–0.92; p=0.005) [179]. Mann et
al.’s [180] post-hoc analysis of the SUSTAIN 1–7 trials showed that
semaglutide initially led to a reduction in eGFR in normal and mildly impaired renal
function (in the SUSTAIN 6 trial with 1.0 mg semaglutide). From week 30
onwards, there was no difference in eGFR between the semaglutide vs. placebo groups
in the SUSTAIN 1–5 and SUSTAIN 7 trials and at week 104 for SUSTAIN 6. In
the SUSTAIN 1–6 trials, albuminuria decreased in patients with
microalbuminuria and macroalbuminuria. In patients with normoalbuminuria, there was
no difference in albuminuria from the beginning to the end of the study.
Semaglutide and G-BA
In a detailed statement by the German Diabetes Society (DDG), the German Society of
Cardiology (DGK), the German Society for Atherosclerosis Research (DGAF), the German
Ophthalmological Society (DOG), the Retinological Society (RG), the Professional
Association of Ophthalmologists (BVA), the Research Group Diabetes e.V. at Helmholtz
Zentrum München, and the Federal Association of Registered Diabetologists
(BVND) on the dossier assessment (A20–93, version 1. 0, status 28.1.2021) of
the Institute for Quality and Efficiency in Health Care (IQWiG) on the benefit
assessment of semaglutide in the form of a subcutaneous application as well as in
an
oral dosage form for the treatment of patients with type 2 diabetes mellitus, the
experts of the professional societies came to the conclusion that the negative
assessment of semaglutide (oral and s. c.) by IQWiG is unjustified
[www.deutsche-diabetes-gesellschaft.de/politik/stellungnahmen/].
Nevertheless, with the decision of the Federal Joint Committee of 15.04.2021, no
additional benefit was granted to semaglutide (BAnz AT 02.06.2021 B5).
Albiglutide
Safety and cardiorenal outcome data have been published for albiglutide [181, 182].
Cardiovascular outcome data on albiglutide (HARMONY outcomes trial [183]) were
analysed and published in 2018. At that time, albiglutide had already been withdrawn
from the market worldwide (July 2017). The HARMONY trial enrolled and randomised
9463 patients (albiglutide 30–50 mg, n=4731; placebo
n=4732). The median observation period was only 1.6 years. There was no
evidence of a difference in major adverse events between the two study arms. In the
3P-MACE, a significant risk reduction with albiglutide (HR 0.78; 95% CI
0.68–0.90; non-inferiority p=0.0001, superiority p=0.0006)
was already evident after this short study duration.
In a recent publication, the authors reported that albiglutide was able to completely
replace prandial insulin in 54% of study participants in patients with type
2 diabetes on baseline bolus insulin therapy, with concomitant improvement in
metabolic control, reduction in hypoglycaemia and body weight [184].
Exendin-based GLP-1 RAs
Exenatide
In the EXSCEL study 14 752 patients (73.1% with cardiovascular disease) were
treated at a mean of 3,2 years with 2.0 mg exenatide once a week. Patients
with or without cardiovascular disease showed no significant difference in the
incidence of MACE between those who received exenatide or a placebo. Critical for
the evaluation of the effects in the EXSCEL study is the very high dropout rate of
over 40%. Compared to the control group, there were no differences in
cardiovascular mortality, non-fatal or fatal myocardial infarction or stroke,
hospitalization for heart failure and incidence of acute pancreatitis, pancreatic
carcinoma, medullary thyroid carcinoma or other serious side effects [185].
In the EXSCEL study, the benefits of exenatide, namely risk reduction in all-cause
mortality (−14%) and first hospitalisation for heart failure
(−11%), could only be seen in study participants who did not have
heart failure at baseline [186]. The risk reduction for all-cause mortality was
confirmed in a recent meta-analysis [187]. The combination of exenatide
(1×weekly) plus dapagliflozin resulted in a significant reduction in HbA1c
(−1.7 vs. − 1.29%) compared to exenatide plus
placebo; dapagliflozin plus placebo decreased HbA1c by
− 1.06% over the same 104-week period. There were also
clinically-relevant positive changes for fasting glucose, 2-h postprandial glucose,
body weight and systolic blood pressure. Severe hypoglycaemia was not observed in
any of the treatment arms [188].
In the meta-analysis by Bethel et al. [189], the 4 large RCTs ELIXA (lixisenatide),
LEADER (liraglutide), EXSCEL (exenatide 1×weekly) and SUSTAIN 6
(semaglutide) were evaluated. Compared with placebo, GLP-1 RAs showed a significant
risk reduction (HR 0.90; 95% CI 0.82–0.99; p=0.033) in the
primary endpoint (cardiovascular mortality, non-fatal myocardial infarction,
non-fatal stroke), a relative risk reduction (RRR) of 13% for cardiovascular
mortality (HR 0.87; 95% CI 0.79–0.96; p=0.007) and for
all-cause mortality of 12% (HR 0.88; 95% CI 0.81–0.95;
p=0.002). However, the statistical heterogeneity between the studies was
large. No significant reductions were found by GLP-1 RAs for non-fatal or fatal
myocardial infarction, stroke, hospitalisation for unstable angina or heart
failure.
Exenatide 1×weekly resulted in a significant reduction in albumin excretion
of 26 rel.% (95% CI − 39.5 to − 10)
compared with a comparison group. Compared with oral antidiabetics, the reduction
in
albuminuria was − 29.6% (95% CI
− 47.6 to − 5.3); with insulin therapy, the value
was − 23.8 rel.% (95% CI − 41.8 to
− 0.2) [190].
Lixisenatide
After this GLP-1 RA showed only non-inferior effects on cardiovascular endpoints in
the ELIXA study [191] and was thus inferior to other GLP-1 RAs, the combination of
insulin glargine with lixisenatide (iGlarLixi) was then investigated [192]. In a
meta-analysis, 8 studies (study duration: 24–30 weeks) with 3538
participants were evaluated. In this analysis, iGlarLixi was superior to therapy
with combination insulin: − 0.50%- units (95% CI
− 0.93 to − 0.06), basal bolus therapy
− 0.35% (−0.89 to+0.13) and basal plus
therapy − 0.68% (−1.18 to − 0.17).
When compared with combi-insulin therapy, there were fewer symptomatic
hypoglycaemias and less weight gain. Analyses of cardiovascular or renal endpoints
were not reported.
Efpeglenatide
Efpeglenatide is an exendin-based GLP-1 RA that has recently been studied in large
RCTs (multicentre and international) in 4076 patients with type 2 diabetes and a
history of cardiovascular disease or renal insufficiency (eGFR 25.0 to
59.9 ml/min) plus another cardiovascular risk factor. Patients were
randomised 1:1:1 (efpeglenatide 4 mg: efpeglenatide 6 mg: placebo)
and analysed after a median observation period of 1.8 years. The primary endpoint
was MACE. This was found in 7.0% with efpeglenatide and 9.2% with
placebo: HR 0.73; 95% CI 0.58–0.92; p<0.001 for
non-inferiority; p=0.007 for superiority. The composite renal endpoint
(reduction in eGFR or macroalbuminuria) was found in 13% in the
efpeglenatide group and 18.4% in the placebo group: HR 0.68; 95% CI
0.57–0.79; p<0.001) [193].
Combination peptides in the near future
Tirzepatide
Tirzepatide is a dual receptor agonist (RA) based on glucose-dependent insulinotropic
peptide (GIP) and dulaglutide administered 1×weekly. It combines the effects
of both substances in a new molecule [194, 195]. In the recently published results
of the RCT study SURPASS 1, tirzepatide was superior at all doses (5 mg,
n=121; 10 mg, n=121; 15 mg, n=121) compared
with placebo (n=115) at the end of the study (40 weeks): Mean HbA1c
decreased from baseline by 1.87% (20 mmol/mol),
1.89% (21 mmol/mol) and 2.07%
(23 mmol/mol Hb), respectively. There was no increased risk of
hypoglycaemia. With placebo, the value increased by 0.04%
(+0.4 mmol/mol Hb). Tirzepatide resulted in a dose-dependent
weight loss of 7.0 to 9.5 kg [196]. When comparing metabolic effects,
tirzepatide was not inferior to semaglutide, but superior in terms of reduction of
HbA1c and body weight [197]. As the first peptide of a new substance class, another
therapy option will soon be available for the treatment of type 2 diabetes, obesity
and fatty liver [198, 199].
A review discusses the potential advantages - SURPASS studies - of this combined
peptide over dulaglutide [200].
Safety aspects of GLP-1 RAs
Retinopathy remained unchanged among GLP-1-RAs except for semaglutide, which
had a negative effect on changes in the ocular fundus (OR 1.75; 95% CI
1.10–2.78; p=0.018) [179]. Whether this is related to the rapid
optimisation of the metabolism is being discussed [201]. In addition, only patients
with pre-existing retinopathy were affected. A corresponding study was initiated to
clarify the retinopathy risk when using semaglutide (Clinical-Trials.gov number,
NCT03 811 561). However, the meta-analysis by Avgerinos et al. on oral semaglutide
showed no evidence of a higher rate of retinopathy [175].
Pancreatitis and cholecystolithiasis as well as neoplasms: Of 113 studies
included in the analysis by Monami et al., 13 found no data on pancreatitis. No
pancreatitis or pancreatic cancer events were reported in 72 studies. In the
remaining studies (n=28), the incidence of pancreatitis and pancreatic
carcinomas with GLP-1-RAs was comparable with the comparative drugs (pancreatitis
OR
0.93; 95% CI 0.65–1.34; p=0.71; pancreatic carcinomas OR
0.94; 95% CI 0.52–1.70; p=0.84). However, the risk for
gallstones was increased (OR 1.30; 95% CI 1.01–1.68;
p=0.041) [126]. In the comprehensive analysis of RCTs published in 2020 with
incretin-based therapies (SAVOR-TIMI 53 (saxagliptin), EXAMINE (alogliptin), TECOS
(sitagiptin), ELIXA (lixisenatide), and with liraglutide in LEADER and semaglutide
in SUSTAIN-6) no significant risk increase for pancreatitis and pancreatic carcinoma
for GLP-1-RA could be found in contrast to therapies with DPP-4 inhibitors [203].
In
the meta-analysis by Cao et al. there was also no evidence for an increased cancer
risk under therapy with GLP-1 RAs [204]. In the meta-analysis published in 2018 by
Bethel et al. [189], there were no differences in pancreatitis, pancreatic carcinoma
and medullary thyroid carcinoma in patients treated with GLP-1-RA therapy compared
to participants treated with placebo. In addition, the large multinational
population-based cohort study with 1 532 513 patients included in the period from
January 1, 2007 to June 30, 2013, and followed up until June 30, 2014, showed no
association of a higher risk for pancreatitis among incretin-based therapies
compared to OADs [128]. These data are consistent with the results of a
meta-analysis of real-world data, which also found no evidence of a higher risk for
pancreatitis among incretin-based therapies [205]. These data fit with the results
of a further meta-analysis of real-world data, which also found no evidence for a
higher risk of pancreatitis with incretin-based therapies [206].
The rate of cholangiocarcinoma was not increased with incretin-based therapy in a
large recent cohort study [207]. A recent meta-analysis also found no evidence for
a
higher risk of breast neoplasia with GLP-1 RA therapy [208].
Incretin-based therapies and fatty liver
Non-alcoholic fatty liver (NASH) is a risk factor for the manifestation of type 2
diabetes, is commonly present in people with type 2 diabetes and is associated with
higher morbidity and mortality. In a recent study with an observation period of 72
weeks, 380 patients with NASH and fibrosis F2 and F3 were randomised to receive
semaglutide s. c. (0.1 mg; n=80 or 0.2 mg; n=78 or
0.4 mg; n=82) or placebo (n=80). In contrast to placebo,
regression of fatty liver without progression of fibrosis was found with
semaglutide: 40% in the 0.1 mg group, 36% in the
0.2 mg group and 59% in the 0.4 mg group. In the placebo
group, the improvement was only 17% (p<0.001 for semaglutide
0.4 mg vs. placebo). However, neoplasia (benign, malignant or unspecified)
was found in 15% of patients in the semaglutide group and 8% in the
placebo group, with no specific organ manifestations observed [209].
Combination of GLP-1 receptor agonists and SGLT2 inhibitors
Compared with GLP-1 RA monotherapy, HbA1c was 0.61% (95% CI
− 1.09 to − 0.14%, 4 trials) lower, body
weight reduction was (−2.59 kg, − 3.68 to
− 1.51 kg, 3 trials) and systolic blood pressure reduction
was (−4.13 mmHg, − 7.28 to
− 0.99 mmHg, 4 trials) in 7 trials analysed (n=1913
patients). Monotherapy with SGLT2 inhibitors reduced HbA1c by 0.85%,
− 1.19 to − 0.52%, 6 trials) and systolic
blood pressure (−2.66 mmHg, − 5.26 to
− 0.06 mmHg, 6 trials). Body weight was unchanged in 5
analysable studies (−1.46 kg, − 2.94 to
0.03 kg). Combination therapy did not lead to increased severe
hypoglycaemia. Data on clinical endpoints were insufficient [210].
Insulins
With the manifold possibilities of oral antidiabetic therapy with or without
combination with GLP-1-RAs, insulin therapy can in many cases be postponed to later
stages of the disease. However, a necessary insulin administration should not then
be delayed by years, as can sometimes be observed [211]. Insulin therapy can be
easily combined with other antidiabetics, and the large number of insulins and
injection aids facilitates individualisation of the therapy.
An extensive discussion on new insulins, however, would go far beyond the scope of
this Clinical Practice Guideline but a comprehensive review was recently published
as a contribution to 100 years of insulin [212].
Therefore, the authors have concentrated on a few aspects of new insulin preparations
in the Clinical Practice Guidelines.
Basal insulin analogues
Insulin degludec (n=3818) is not inferior to insulin glargin 100
(n=3819) in the therapy of people with type 2 diabetes and a high risk of
cardiovascular events in terms of MACE. The HbA1c values were identical in both
groups over the observational period of 2 years (7.5±1.2%), but the
fasting plasma glucose values were significantly lower under insulin degludec. The
hazard ratio was 0.91 (95% CI 0.78–1.06) for the primary endpoint
(cardiovascular death, non-fatal myocardial infarction, non-fatal stroke). By
contrast, the rate of severe hypoglycaemia (secondary endpoint) was significantly
lower for insulin degludec (4.9%) than for insulin glargin 100
(6.6%) (hazard ratio 0.60; 95% CI 0.48–0.76;
p<0.001). The rate of severe side effects such as benign and malignant
neoplasia was comparable (DEVOTE study [213]). In the DEVOTE study, it was shown
once again that confirmed severe hypoglycaemia was associated with an increased rate
of all-cause mortality in a period of 15–365 days before the clinical
endpoint [214].
Pharmacokinetic and pharmacodynamic studies have shown that insulin glargin 300 has
a
flatter efficacy profile, lasts slightly longer and has a lower day-to-day
variability than insulin glargin 100. Metabolic control was comparable for both
insulin types, while the rate of nocturnal hypoglycaemia was significantly lower for
insulin glargin 300 than for insulin glargin 100 [215–217].
Biosimilar insulin glargin 100: Pharmacokinetics and -dynamics are comparable
for insulin glargin 100 and biosimilar insulin glargin 100 in people without and
with type 2 diabetes [218, 219]. In the meta-analysis by Yamada et al. [220] there
were no differences between biosimilar insulins and the original insulins in
relation to: HbA1c, fasting plasma glucose, hypoglycaemia, injection site reactions,
insulin antibodies, allergic reactions and mortality.
When comparing different insulin analogues (insulin glargin and insulin degludec)
with human insulin, a large cohort study from Denmark, Finland, Norway, Sweden and
Great Britain found no evidence of an increased carcinoma risk, neither for insulin
glargin nor for insulin degludec compared to human insulin for the 10 examined
carcinomas in a mean observational period of 4.6 years [221].
Combination of long-acting insulin plus GLP-1-RA
The fixed combination of long-acting insulin plus GLP-1-RA or free simultaneous or
consecutive combinations have advantages over intensive insulin therapy with
prandial and basal insulin in terms of therapy adherence, rate of hypoglycaemia,
weight progression and insulin usage. Compared to intensive insulin therapy,
however, gastrointestinal side effects were more frequent with GLP-1-RA
[222–224]. In a recent meta-analysis, the authors concluded that
combinations of basal insulin with long-acting GLP-1-RA were superior to
combinations of basal insulin with short-acting GLP-1-RA in terms of weight
reduction, HbA1c value reduction, lower fasting glucose values and benefits in terms
of gastrointestinal side effects [225].
The first fixed combination approved in Germany is insulin glargine (100
I.U./ml) and lixisenatide (see above).
Fast-acting insulin analogues
Insulin lispro 200 shows potential advantages for a higher concentrated insulin
especially in cases of severe insulin resistance (e. g., obesity), as less
volume has to be injected with the same amount of insulin and economic advantages
for the patient. Compared to insulin lispro 100, insulin lispro 200 showed also
significant improvements in variability of fasting glucose, HbA1c, hypoglycaemic
rate and satisfaction with therapy. At the same time, a reduction of 20%
insulin was possible [226].
Ultra-fast insulin aspart is absorbed by the blood twice as fast and thus has an
approximately 50% higher insulin effect with significantly lower
postprandial blood glucose values, especially in the first 30 min after
injection. The faster onset of action means that glucose is even better
controllable, especially in people with type 1 diabetes and those on insulin pump
therapy [227]. Ultra-fast insulin aspart showed a similar reduction of HbA1c
compared to insulin aspart in people with type 2 diabetes (observation time 26
weeks); the 1-hour postprandial glucose values were significantly lower after
injection of fast insulin aspart, but not 2–4 h after a test meal.
The total rates of severe hypoglycaemia were not different between the two insulins.
However, the relative risk of hypoglycaemia 0–2 h postprandially was
significantly higher with fast insulin aspart (RR 1.60; 95% CI
1.13–2.27) [228]. Ultra-rapid insulin lispro (URLI=Ultra Rapid
Lispro Insulin) showed a 6.4-fold faster onset in the first 15 min after
injection compared to insulin lispro (p<0.0001). The insulin effect of URLi
was 13 min significantly faster and 4.2-fold greater in the first
30 min than that of insulin lispro [229].