This site is intended for healthcare professionals only

Journal of
Diabetes Nursing

Issue:

Share this article

GLP-1 receptor agonists in type 2 diabetes: An underused asset?

David Morris
As our understanding of the incretin hormone system has increased, a number of drugs targeting this system have been developed. The realisation of this potential has developed rapidly, and glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are now a standard feature in management guidelines for type 2 diabetes. This article reviews the operation of the incretin system and the mechanism by which GLP-1 RAs act to provide benefit in type 2 diabetes. The availability and indications for use of the GLP-1 RAs, and their clinical benefits and disadvantages, are summarised. The position of GLP-1 RAs in the management of type 2 diabetes is discussed pragmatically, with reference to various key guidelines.

Just over 50 years ago, experimental evidence demonstrated that an oral glucose load elicits a greater insulin response than an intravenous glucose load (Elrick et al, 1964). Subsequently, it was demonstrated that this effect was generated by hormones located in the small intestine secreted in response to oral intake of food (Creutzfeldt and Ebert, 1985). The so-called “incretin effect” is, in fact, mediated principally by two hormones, glucagon-like peptide-1 (GLP-1) and gastric inhibitory polypeptide (GIP; Drucker and Nauck, 2006).

The incretin hormones secreted in response to food stimulate insulin release from pancreatic beta-cells and suppress glucagon release from alpha-cells, both of which reduce postprandial hyperglycaemia. Importantly, the activity of the incretin hormones requires elevated blood glucose levels (i.e. it is glucose-dependent), as well as the presence of food in the gut. In addition, the incretin hormones slow gastric emptying, thereby delaying the absorption of food (and particularly glucose) and reducing appetite (Nauck and Meier, 2016).

Both GLP-1 and GIP are metabolised in vivo by the enzyme dipeptidyl peptidase-4 (DPP-4) within 2 minutes. In people with type 2 diabetes, GLP-1 receptor activity is retained and exogenous GLP-1 administered intravenously has been shown to reduce both fasting and postprandial hyperglycaemia (Drucker and Nauck, 2006). In contrast, GIP receptor activity is much reduced in type 2 diabetes; in fact, the incretin response is diminished in people with type 2 diabetes compared to those without diabetes, probably reflecting beta-cell dysfunction (Nauck et al, 1986; Knop et al, 2007).

The rationale behind using GLP-1 receptor agonists in type 2 diabetes
The short half-life of human GLP-1 in vivo renders it unsuitable for clinical use and has led to the search for GLP-1 receptor agonists (GLP-1 RAs) that mimic the action of endogenous GLP-1 (by binding to and activating the GLP-1 receptor) but which have resistance to breakdown by DPP-4 and thus have more prolonged activity (Meier, 2012). The hope would be that GLP-1 RAs could counter raised glucose levels by stimulating insulin secretion, suppressing glucagon secretion, delaying gastric emptying and inducing satiety (Nauck and Meier, 2016). In individuals with type 2 diabetes, this should translate into improved HbA1c levels and weight reduction.

The fact that GLP-1 RA activity is glucose-dependent offers the advantage of glycaemic control with low risk of hypoglycaemia, as GLP-1 RAs do not stimulate insulin secretion or suppress glucagon secretion when glucose levels are not raised (Sharma et al, 2018).

Currently available GLP-1 RAs
Currently there are six GLP-1 RAs available for clinical use in the UK, all administered by subcutaneous injection to circumvent the problem of peptide degradation by gastric acid. They all share the same mechanism of action but the different molecular structures and formulations lead to varying duration of action and, in turn, to varied dosage regimens. Differences in glycaemic control, weight reduction, cardiovascular effects and side-effect profiles are seen between the drugs in clinical trials.

The first GLP-1 RA that gained approval for use in type 2 diabetes was exenatide, licensed for use in the UK in 2007. Exenatide is a synthetic product reproducing the peptide sequence of exendin-4, which had been isolated from the saliva of the Gila monster, a lizard found in the deserts of Arizona and New Mexico. Exenatide standard-release (Byetta) is licensed for twice-daily use before two main meals (at least 6 hours apart).

Liraglutide (Victoza) was the next GLP-1 RA to be developed, becoming available in 2009. In contrast to exenatide, it is 97% homologous to human GLP-1. Liraglutide has a half-life of around 13 hours, enabling it to be injected once daily.

The first once-weekly GLP-1 RA approved for use in type 2 diabetes was exenatide modified-release (Bydureon). In this formulation the exenatide molecules are enclosed within biodegradable polymeric microspheres that slowly break down in the subcutaneous tissue, releasing exenatide into the circulation in a controlled manner (Fineman et al, 2011).

Lixisenatide (Lyxumia), like exenatide, is a derivative of exendin-4. It has a short duration of action and is licensed to be taken within one hour before the first meal of the day or the evening meal.

Further once-weekly GLP-1 RAs based on modified human GLP-1 have followed: dulaglutide (Trulicity), albiglutide (Eperzan; now discontinued) and semaglutide (Ozempic).

Conventional wisdom would dictate that administering peptides orally is unfeasible because of gastric acid degradation. An oral formulation of semaglutide, however, is  in the advanced stages of development. The semaglutide is taken once daily after an overnight fast, following which no food or medication should be taken for 30 minutes. The European Medicines Agency has recently recommended a market authorisation for oral semaglutide for use in type 2 diabetes.

The principle mechanism of action of the short-acting exendin-4 derivatives, exenatide and lixisenatide, is to delay gastric emptying and thus lower postprandial glucose levels (Nauck and Meier, 2019). In contrast, the longer-acting GLP-1 RAs exert their effects mainly by stimulating insulin release and inhibiting glucagon secretion, which impacts on both postprandial and fasting glucose levels.

Indications and contraindications
GLP-1 RAs are indicated for the treatment of type 2 diabetes in combination with other glucose-lowering medications, including insulin, when these together with diet and exercise do not provide adequate glycaemic control. Liraglutide, dulaglutide and semaglutide are indicated as monotherapy in situations where metformin is poorly tolerated or contraindicated. All currently licensed GLP-1 RAs are given by subcutaneous injection into the abdomen, thigh or upper arm, rotating the injection sites from one injection to the next.

Combination therapy
When adding a GLP-1 RA to an insulin secretagogue (a sulfonylurea or meglitinide), consideration should be given to lowering the dose of the latter medications to reduce the risk of hypoglycaemia. If a DPP-4 inhibitor is being taken, it should be discontinued on commencement of a GLP-1 RA as the combination does not provide additional glycaemic control (American Diabetes Association, 2018).

Pioglitazone and sodium–glucose cotransporter 2 (SGLT2) inhibitors are reasonable combination therapies with GLP-1 RAs. If a GLP-1RA is added to a basal insulin then, unless HbA1c is markedly raised, it is prudent to reduce the dose of insulin by as much as 20–25%, although this can be re-uptitrated later as necessary (Nauck and Meier, 2019).

Contraindications
Contraindications (these vary slightly between EU and US labels) and reasons to avoid using GLP-1 RAs are summarised in Box 1.

GLP-1 RAs are licensed for use in varying stages of renal impairment depending on the agent (Table 1). There are no specific requirements regarding the use of GLP-1 RAs in hepatic impairment, other than liraglutide, which is not recommended in severe hepatic impairment (Child–Pugh score >9).

No dose adjustment is necessary with regard to weight, BMI or, in most cases, age (a lower starting dose of dulaglutide 0.75 mg once weekly is recommended in those aged 75 years or more; and caution in increasing the dose of immediate-release exenatide from 5 to 10 µg twice daily is advised in the elderly).

Dose regimens and administration
The properties and dose regimens of the GLP-1 RAs are summarised in Table 2 (Lyseng-Willimason, 2019; Nauck and Meier, 2019; Romera et al, 2019). It can be seen that the exendin-4-derived GLP-1 RAs are short-acting (exenatide once-weekly having a longer duration of action because of its microsphere formulation), whilst the modified human GLP-1 derivatives are long-acting.

Selection of a GLP-1 RA for an individual may be influenced by frequency of dosing, ease of dose titration and pen-related factors such as ease of administration, including needle arrangements to minimise the fear of injection. Some of these factors are summarised in Table 3 (Nauck and Meier, 2019; Romera et al, 2019).

Fixed-dose combination products
GLP-1 RAs are also available in combination with a basal insulin in a fixed dose ratio for a once-daily injection. A basal insulin has greatest impact on fasting glucose levels whilst the GLP-1 RA has an effect on postprandial glucose levels (as well as fasting glucose in the case of a long-acting GLP-1 RA; Kenny and Hall, 2015). The GLP-1 RA mitigates weight gain induced by insulin and does not add to hypoglycaemic burden, and the combination is more effective in lowering HbA1c than either component separately (Gough et al, 2014).

Two products are currently available. IDegLira (Xultophy) is formulated with a ratio of 1 unit of insulin degludec to 0.036 mg of liraglutide. The multiple-use, disposable pen can accommodate a top dose of 50 units of degludec plus 1.8 mg of liraglutide once daily, given at any time. A starting dose of 10 “dose steps” (equivalent to 10 units of degludec and 0.36 mg of liraglutide) can be added to oral antidiabetes medications; alternatively, if the individual was previously receiving a basal insulin, the starting dose would be 16 dose steps.

The second combination product is IGlarLixi (Suliqua, formerly LixiLan), a combination of insulin glargine and lixisenatide. This is available as two differing concentrations: glargine 100 units/mL plus lixisenatide 33 µg/mL, or glargine 100 units/mL plus lixisenatide 50 µg/mL. The dose can be titrated up to allow a maximum of 60 units of glargine plus 20 µg of lixisenatide, to be administered an hour before the same main meal once daily.

Side-effects and other issues
The most frequent side-effects from using GLP-1 RAs are gastrointestinal. Individuals need to be warned of the possibility of nausea and vomiting but should also be reassured that these are usually mild to moderate in intensity and dissipate with time (Raccah, 2017). The incidence of nausea is around 20%, with a higher rate for the short-acting GLP-1 RAs (Bettge et al, 2017), which is associated with increased discontinuation. Diarrhoea can also be experienced, and in this case incidence is higher for the long-acting rather than short-acting GLP-1 RAs (Bettge et al, 2017).

Acute pancreatitis has been observed with GLP-1 RA use and product licenses retain a warning on this. Users should also be informed of the symptoms of acute pancreatitis and to stop the GLP-1 RA should they experience severe upper abdominal pain, and to seek medical advice. If pancreatitis is confirmed then treatment should not be restarted. Reassuringly, the incidence of pancreatitis did not appear to be increased in the cardiovascular outcome trials with GLP-1 RAs (see later), and recent meta-analyses did not find evidence of an increased risk of acute pancreatitis or pancreatic cancer, although an increased risk of gallbladder events was identified (Monami et al, 2017; Storgaard et al, 2017; Bethel et al, 2018).

An association between GLP-1 RA use and medullary thyroid cancer had been suggested from animal studies. However, a meta-analysis of once-weekly GLP-1 RAs showed no increased risk in comparison with other antidiabetes drugs (Bethel et al, 2018).

Injection site reactions are rare with GLP-1 RAs but do appear to be significantly more frequent with the microsphere formulation of once-weekly exenatide (Blevins et al, 2011).

In situations of hypovolaemia, such as severe gastrointestinal disturbance, GLP-1 RAs have been linked to acute kidney injury, and measures to counter dehydration and temporary cessation of treatment may be required (Filippatos et al, 2014).

In the cardiovascular outcome trial of semaglutide (SUSTAIN-6), a significant increased risk in the prespecified retinal outcome (a composite of need for photocoagulation, need for intravitreal agents, vitreous haemorrhage or new-onset blindness) was observed (hazard ratio [HR], 1.76), principally in subjects with pre-existing diabetic retinopathy who were using insulin (Marso et al, 2016a). It has been suggested that this was linked to a rapid improvement in glycaemic control, a phenomenon that has been noted in type 1 diabetes and pregnancy studies. Accordingly, caution is advised when using semaglutide in people with diabetic retinopathy.

Clinical benefits
The GLP-1 RAs are effective glucose-lowering agents, achieving HbA1c reductions in the order of 11 mmol/mol (1.0%) compared with placebo in practice (Waldrop et al, 2018), although there is some variability in efficacy within the class. Greater HbA1c reductions are seen with higher starting HbA1c levels (Bihan et al, 2016). The longer-acting GLP-1 RAs appear to be more effective than the shorter-acting agents, although not all clinical trials had the same design. The evidence indicates that semaglutide is the most powerful agent, ahead of liraglutide and dulaglutide, followed by exenatide once-weekly and then exenatide twice-daily and lixisenatide (Htike et al, 2017; Davies et al, 2018).

As indicated previously, a useful property of GLP-1 RAs is their low propensity to induce hypoglycaemia. The risk of hypoglycaemia is lower than with insulin or sulfonylureas (Levin et al, 2017). If a GLP-1 RA is added to insulin or an insulin secretagogue (sulfonylureas and meglitinides), consideration should be given to reducing the dose of the latter agents to minimise the risk of hypoglycaemia.

An important clinical benefit offered by GLP-1 RAs in type 2 diabetes is that of weight loss associated with reduced appetite (Sun et al, 2015; Bihan et al, 2016). This reaches a plateau after 6 months or so of treatment and varies between members of the class from around 1.5 kg to 6.0 kg, the highest reductions being found with semaglutide (Htike et al, 2017; Andreadis et al, 2018; Nauck and Meier, 2019). In fact, there is a relatively high degree of variation in weight loss at the individual level (anything from zero to over 20 kg), much greater than the variation seen in glycaemic control (Nauck and Meier, 2019). The weight loss observed with GLP-1 RA therapy contrasts with the weight gain which can be seen with insulin, pioglitazone and most sulfonylureas.

A small reduction in systolic blood pressure (around 2–5 mmHg) is associated with GLP-1 RA treatment, although this is accompanied by a small increase in pulse rate of 2–5 beats/min (Holman et al, 2017; Nauck et al, 2017). A slightly beneficial effect on lipid profile is also apparent, with a small reduction in LDL-cholesterol and triglyceride levels.

Cardiovascular benefits
Cardiovascular outcome trials have been performed on all the available GLP-1 RAs except twice-daily exenatide. The participants in these double-blind, randomised, placebo-controlled trials comprised individuals with type 2 diabetes who had pre-existing cardiovascular disease or cardiovascular risk factors. The primary outcome in all was major adverse cardiac events (MACE): a combination of non-fatal myocardial infarction (MI), non-fatal stroke and cardiovascular death.

The first GLP-1 RA to show significant benefit in cardiovascular outcomes was liraglutide in the LEADER trial (Marso et al, 2016b), with an absolute risk reduction (ARR) in MACE of 1.9% over 3.8 years compared with placebo, and a significant relative risk reduction of 13% (HR, 0.87). The HR for all-cause mortality was also significantly reduced versus placebo (HR, 0.87).

In SUSTAIN-6, semaglutide was also shown to provide cardiovascular benefit, with an ARR of 2.3% over 2.1 years and an HR of 0.74 for MACE compared with placebo (Marso et al, 2016a). This was driven mainly by a reduction in risk of stroke.

The EXSCEL study compared exenatide modified-release with placebo (Holman et al, 2017). Although the HR for MACE was 0.91 in favour of exenatide, this just missed statistical significance. There was a significant reduction in all-cause mortality. For the other exendin-4 derivative, lixisenatide, cardiovascular safety but not benefit was demonstrated in the ELIXA trial (Pfeffer et al, 2015).

Cardiovascular benefits (significant improvements in MACE) were subsequently identified in the trials of dulaglutide (REWIND; Gerstein et al, 2019), principally driven by a reduction in non-fatal stroke, and albiglutide (HARMONY; Hernandez et al, 2018), largely driven by reductions in fatal and non-fatal MI.

The PIONEER-6 trial of oral semaglutide showed a strong trend towards cardiovascular benefit but, as with the exenatide trial, this did not reach the level of significance (Husain et al, 2019). A summary of the GLP-1 RA cardiovascular outcome trials can be found in Table 4.

In addition to the individual studies above, systematic reviews of the cardiovascular safety of the GLP-1 RAs as a class indicate that they reduce the risk of cardiovascular events, with an overall 10% reduction in MACE, a 13% reduction in cardiovascular death and a 12% reduction in all-cause mortality (Bethel et al, 2018; Jia et al, 2018; Kristensen et al, 2019).

Thus, it would appear that the longer-acting (rather than short-acting) GLP-1 RAs offer benefit in those with established cardiovascular disease, possibly favouring the agents with a close analogy to the human GLP-1 molecular structure. The strongest evidence lies with semaglutide, liraglutide, dulaglutide and albiglutide, followed by exenatide once-weekly (although these results did not reach significance), with lixisenatide being neutral in terms of cardiovascular outcomes. Unlike certain SGLT2 inhibitors, the GLP-1 RAs do not appear to confer benefits in terms of heart failure.

The mechanisms behind these cardiovascular benefits are probably multifactorial (Nauck et al, 2017; Boyle et al, 2018). Reduced blood pressure, weight loss, lower LDL-cholesterol concentrations and lower blood glucose levels are all conventional risk factors that are positively affected by GLP-1 RAs. Other factors that might retard the progress of atherosclerosis are reduced low-grade inflammation and improved plaque stability.

Renal benefits
In the cardiovascular outcome trials of the GLP-1 RAs, renal effects were a secondary outcome, although the criteria were not uniform between different studies. In the LEADER trial, liraglutide demonstrated a 22% relative risk reduction compared to placebo in the prespecified secondary renal composite outcome of new-onset macroalbuminuria, doubling of serum creatinine, end-stage renal disease or death due to renal disease (Mann et al, 2017). In SUSTAIN-6 (Marso et al, 2016a), semaglutide reduced the same secondary renal outcome versus placebo (relative risk reduction, 36%). In both studies, the most significant factor contributing to the improved renal outcome was reduced progression of albuminuria.

Similarly, reduction in progression to macroalbuminuria was found with exenatide once-weekly, lixisenatide, albiglutide and dulaglutide (Pfeffer et al, 2015; Holman et al, 2017; Hernandez et al, 2018; Gerstein et al, 2019). A recent meta-analysis confirmed that treatment with GLP-1 RAs was associated with a reduction in risk of adverse kidney outcomes and that this was principally due to a reduction in urinary albumin excretion (Kristensen et al, 2019). Thus, it can be seen that a beneficial effect on development of proteinuria is a consistent feature of the GLP-1 RAs, although the response in terms of estimated glomerular filtration rate (eGFR) and incidence of end-stage renal failure is less clear.

Clinical guidelines
NICE (2015) guidance on GLP-1 RAs is summarised in Box 2. In light of subsequent evidence, this guidance, in the opinion of the author, now appears quite restrictive and is behind current practice.

More recent guidelines from SIGN (2017) on glycaemic control in type 2 diabetes acknowledge that GLP-1 RAs are an alternative to insulin initiation and recognise their benefit in people with established cardiovascular disease (Box 3).

ADA/EASD guidelines
Both NICE and SIGN advice places GLP-1 RAs as a third- or fourth-line treatment to improve glycaemic control in type 2 diabetes. The American Diabetes Association and European Association for the Study of Diabetes (ADA/EASD) consensus report took the advice further, recommending GLP-1 RAs as a possible second-line treatment for glycaemic control in type 2 diabetes depending on individual circumstances (Davies et al, 2018). Specifically, in people with established atherosclerotic cardiovascular disease (ASCVD), a GLP-1 RA with proven CV benefit is recommended, an alternative choice being an SGLT2 inhibitor. If HbA1c is already at target, the guidance suggested to consider switching across to a suitable GLP-1 RA to provide cardiovascular benefit.

More recently, the ADA/EASD consensus report was updated, strengthening the recommendations for use of GLP-1 RA and SGLT2 inhibitor therapy, notably in recommending them in those at high risk of cardiovascular disease, chronic kidney disease and/or heart failure (Buse et al, 2020). These drugs are now recommended for consideration in these situations regardless of whether the individual’s HbA1c is at target level. GLP-1 RAs with proven cardiovascular benefit (liraglutide, semaglutide and dulaglutide) are recommended in preference to SGLT2 inhibitors if ASCVD is the key pathology because of their positive impact on MACE.

GLP-1 RAs may be considered as an option for individuals with type 2 diabetes who have risk factors for, but not established, ASCVD (risk factors defined as age 55 years or older with >50% stenosis of coronary, carotid or peripheral arteries, left ventricular hypertrophy, eGFR <60 mL/min/1.73 m2 or albuminuria). The strongest evidence to support this use of GLP-1 RAs in primary prevention of ASCVD came from the dulaglutide cardiovascular outcome trial, in which nearly 70% of participants had this high risk profile but not established ASCVD (Gerstein et al, 2019).

If chronic kidney disease or heart failure predominates, and if an SGLT2 inhibitor (the first-choice therapy) is contraindicated or not tolerated, or if eGFR is less than adequate for SGLT2 inhibitor use, then a GLP-1 RA becomes the agent of choice to improve glycaemic control.

GLP-1 RAs are also an option to add to metformin if there is a compelling need to lose weight or avoid hypoglycaemia.

The ADA/EASD guideline advocates GLP-1 RAs to be the first-choice injectable therapy (ahead of a basal insulin) to maximise glycaemic lowering. The caveat to this recommendation is that, for individuals with a very high HbA1c (>97 mmol/mol [11.0%]), evidence of osmotic symptoms (polyuria, polydipsia, weight loss) or if type 1 diabetes is a possibility, insulin is recommended.

Discussion: When to use GLP-1 RAs in the real world
Lifestyle change focused on diet, exercise and weight loss are the cornerstone in managing type 2 diabetes. Almost all guidelines recommend metformin as the first pharmacological treatment; however, the advice begins to diverge after this.

The key principle in choosing treatment is individualisation of therapy. So alongside effectiveness, safety, tolerability, ease of use, cost of treatment and need for monitoring, patient factors including age, comorbidities (notably cardiovascular and renal status), duration of diabetes, other medications and, importantly, the individuals’ circumstances (occupation and domestic situation) and preferences need to be taken into account.

Advantages
GLP-1 RAs exert a significant glucose-lowering effect (generally greater than is seen with oral agents) and are effective in both the early and late stages of type 2 diabetes (Nauck and Meier, 2019). The longer-acting GLP-1 RAs hold the advantage in facilitating HbA1c reduction; they act to lower both postprandial and fasting glucose levels, whereas shorter-acting agents exert their effect mainly on postprandial glucose levels alone (Nauck and Meier, 2019). Semaglutide appears to have the greatest HbA1c-lowering effect, followed by liraglutide and dulaglutide, and then exenatide once-weekly. Oral semaglutide has similar efficacy to liraglutide in terms of glycaemic lowering (Pratley et al, 2019).

Important features of GLP-1 RAs are the promotion of weight loss and a low risk of hypoglycaemia, which makes them a useful option for managing hyperglycaemia in obese individuals with type 2 diabetes and in situations where hypoglycaemia must be avoided (Davies et al, 2018). Semaglutide (both subcutaneous and oral preparations) appears to offer the greatest weight loss (Nauck and Meier, 2019; Hussain et al, 2019).

There is now good evidence that several long-acting GLP-1 RAs (liraglutide, dulaglutide and semaglutide) provide cardiovascular protection (Table 4), and hence these agents (as do SGLT2 inhibitors) become a logical add-on therapy to metformin for glycaemic lowering in those with established cardiovascular disease (Buse et al, 2020).

Many individuals with type 2 diabetes suffer from renal impairment, and it is therefore of practical importance that GLP-1 RAs are licensed to be used in varying degrees of chronic kidney disease (Table 1). As a group, the GLP-1 RAs appear to reduce the progression of albuminuria, so they are a logical choice (behind SGLT2 inhibitors) for glycaemic control in diabetic nephropathy (Buse et al, 2020).

Once-daily and, especially, once-weekly GLP-1 RAs may, because of reduced injection frequency, improve compliance (Romera et al, 2019). This may also be an important factor in those with needle phobia, some of whom may prefer a once-weekly injection with a hidden pre-attached needle (see Table 3).

If a GLP-1 RA is added to a sulfonylurea or a meglitinide (such as repaglinide) then, unless HbA1c is particularly high, it is wise to reduce the dose of the latter to lower the risk of hypoglycaemia. DPP-4 inhibitors are unlikely to have a complementary effect to GLP-1 RAs and, therefore, are recommended to be discontinued (Davies et al, 2018; Lajthia et al, 2019). In contrast, the combination of a GLP-1 RA and an SGLT2 inhibitor to improve glycaemic control looks particularly attractive as both treatments facilitate weight loss and neither induce hypoglycaemia; furthermore, both treatments appear to offer cardiovascular benefits and renoprotection (Castellana et al, 2019; Guo et al, 2020).

The evidence indicates that GLP-1 RAs are at least as effective as a basal insulin in lowering HbA1c in type 2 diabetes (Abd El Aziz et al, 2017; Singh et al, 2017), and this coupled with the benefit of weight reduction and low risk of hypoglycaemia (in contrast to insulin) underpins the ADA/EASD advice to use GLP-1 RAs ahead of insulin (Davies et al, 2018). However, be wary when considering the individual with very high blood glucose levels, osmotic symptoms, a personal or family history of autoimmune disease, or with normal or low BMI – such individuals may well be insulin-deficient, not insulin-resistant, and insulin would be the right choice for them.

For individuals with type 2 diabetes already using a basal insulin, options to further improve glycaemic control include addition of a GLP-1 RA or adding a rapid-acting insulin with meals. There are strong arguments for preferring the former strategy. Compared with a prandial insulin, addition of a GLP-1 RA offers potential weight loss (rather than weight gain), avoidance of hypoglycaemia, less monitoring of blood glucose levels and easier dose titration (Abd El Aziz et al, 2017; Wysham et al, 2017; Davies et al, 2018). When adding a GLP-1 RA to insulin, if HbA1c is <70 mmol/mol (8.5%), it is prudent to reduce the dose of insulin (at least initially) by around 20–25% (Nauck and Meier, 2019). The combination of a GLP-1 RA and basal insulin is highly effective in improving glycaemic control in type 2 diabetes and logical given their complementary actions. Thus, adding a basal insulin to a GLP-1 RA is also a useful strategy (Buse et al, 2020). Disadvantages
On the downside, GLP-1 RAs are an expensive treatment and this can restrict their use, particularly in developing countries, where they may be excluded from the national formulary on the grounds of cost.

Gastrointestinal side-effects, particularly nausea, are common with GLP-1 RAs, although generally these are mild to moderate in intensity and usually decline over time (Nauck and Meier, 2019). As the side-effects are dose-dependent, it may be appropriate to reduce the dose of GLP-1 RA. Depending on the agent, a gradual uptitration of dose can minimise these side-effects, and a once-weekly preparat

REFERENCES:

Abd El Aziz MS, Kahle M, Meier JJ, Nauck MA (2017) A meta-analysis comparing clinical effects of short- or long-acting GLP-1 receptor agonists versus insulin treatment from head-to-head studies in type 2 diabetic patients. Diabetes Obes Metab 19: 216–27
American Diabetes Association (2018) 8. Pharmacologic approaches to glycemic treatment: standards of medical care in diabetes – 2018. Diabetes Care 41(Suppl 1): S73–85
Andreadis P, Karagiannis T, Malandris K et al (2018) Semaglutide for type 2 diabetes mellitus: a systematic review and meta-analysis. Diabetes Obes Metab 20: 2255–63
Bethel MA, Patel RA, Merrill P et al (2018) Cardiovascular outcomes with glucagon-like peptide-1 receptor agonists in patients with type 2 diabetes: a meta-analysis. Lancet Diabetes Endocrinol 6: 105–13
Bettge K, Kahle M, Abd El Aziz MS et al (2017). Occurrence of nausea, vomiting and diarrhoea reported as adverse events in clinical trials studying glucagon-like peptide-1 receptor agonists: a systematic analysis of published clinical trials. Diabetes Obes Metab 19: 336–47
Bihan H, Ng WL, Magliano DJ, Shaw JE (2016) Predictors of efficacy of GLP-1 agonists and DPP-4 inhibitors: a systematic review. Diabetes Res Clin Pract 121: 27–34
Blevins T, Pullman J, Malloy J et al (2011) DURATION-5: exenatide once weekly resulted in greater improvements in glycemic control compared with exenatide twice daily in patients with type 2 diabetes. J Clin Endocrinol Metab 96: 1301–10
Boyle JG, Livingstone R, Petrie JR (2018) Cardiovascular benefits of GLP-1 agonists in type 2 diabetes: a comparative review. Clin Sci (Lond) 132: 1699–709
Brønden A, Naver SV, Knop FK, Christensen M (2015) Albiglutide for treating type 2 diabetes: an evaluation of pharmacokinetics/pharmacodynamics and clinical efficacy. Expert Opin Drug Metab Toxicol 11: 1493–503
Buse JB, Wexler DJ, Tsapas A et al (2020) 2019 update to: Management of hyperglycaemia in type 2 diabetes, 2018. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetologia 63: 221–8
Castellana M, Cignarelli A, Brescia F et al (2019) Efficacy and safety of GLP-1 receptor agonists as add-on to SGLT2 inhibitors in type 2 diabetes mellitus: A meta-analysis. Sci Rep 9: 19351
Creutzfeldt W, Ebert R (1985) New developments in the incretin concept. Diabetologia 28: 565–73
Davies MJ, D’Alessio DA, Fradkin J et al (2018) Management of hyperglycaemia in type 2 diabetes, 2018. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetologia 61: 2461–98
Drucker DJ, Nauck MA (2006) The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 368: 1696–705
Elrick H, Stimmler L, Hlad CJ Jr, Arai Y (1964) Plasma insulin response to oral and intravenous glucose administration. J Clin Endocrinol Metab 24: 1076–82
Filippatos TD, Panagiotopoulou TV, Elisaf MS (2014) Adverse effects of GLP-1 receptor agonists. Rev Diabet Stud 11: 202–30
Fineman M, Flanagan S, Taylor K et al (2011) Pharmacokinetics and pharmacodynamics of exenatide extended-release after single and multiple dosing. Clin Pharmacokinet 50: 65–74
Gerstein HC, Colhoun HM, Dagenais GR et al (2019) Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomised placebo-controlled trial. Lancet 394: 121–30
Gough SC, Bode B, Woo V et al (2014) Efficacy and safety of a fixed-ratio combination of insulin degludec and liraglutide (IDegLira) compared with its components given alone: results of a phase 3, open-label, randomised, 26-week, treat-to-target trial in insulin-naive patients with type 2 diabetes. Lancet Diabetes Endocrinol 2: 885–93
Guo M, Gu J, Teng F et al (2020) The efficacy and safety of combinations of SGLT2 inhibitors and GLP-1 receptor agonists in the treatment of type 2 diabetes or obese adults: a systematic review and meta-analysis. Endocrine 67: 294‑304
Hernandez AF, Green JB, Janmohamed S et al (2018) Albiglutide and cardiovascular outcomes in patients with type 2 diabetes and cardiovascular disease (Harmony Outcomes): a double-blind, randomised placebo-controlled trial. Lancet 392: 1519–29
Holman RR, Bethel MA, Mentz RJ et al (2017) Effects of once-weekly exenatide on cardiovascular outcomes in type 2 diabetes. N Engl J Med 377: 1228–39
Htike ZZ, Zaccardi F, Papamargaritis D et al (2017). Efficacy and safety of glucagon-like peptide-1 receptor agonists in type 2 diabetes: a systematic review and mixed-treatment comparison analysis. Diabetes Obes Metab 19: 524–36
Husain M, Birkenfeld AL, Donsmark M et al (2019) Oral semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 381: 841–51
Jia X, Alam M, Ye Y et al (2018) GLP-1 receptor agonists and cardiovascular disease: a meta-analysis of recent cardiac outcome trials. Cardiovasc Drugs Ther 32: 65–72
Kenny C, Hall G (2015) GLP-1 receptor agonist and basal insulin co-therapy in type 2 diabetes: clinical evidence and practicalities of use. Diabetes & Primary Care 17: 80–5
Knop FK, Vilsbøll T, Højberg PV et al (2007) Reduced incretin effect in type 2 diabetes: cause or consequence of the diabetic state? Diabetes 56: 1951–9
Kristensen SL, Rørth R, Jhund PS et al (2019) Cardiovascular, mortality, and kidney outcomes with GLP-1 receptor agonists in patients with type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet Diabetes Endocrinol 7: 776–85
Lajthia E, Bucheit JD, Nadpara PA et al (2019) Combination therapy with once-weekly glucagon like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes: a case series. Pharm Pract (Granada) 17: 1588
Levin PA, Nguyen H, Wittbrodt ET, Kim SC (2017) Glucagon-like peptide-1 receptor agonists: a systematic review of comparative effectiveness research. Diabetes Metab Syndr Obes 10: 123–39
Lyseng-Williamson KA (2019) Glucagon-like peptide-1 receptor analogues in type 2 diabetes: their use and differential features. Clin Drug Investig 39: 805–19
Mann JFE, Ørsted DD, Brown-Frandsen K et al (2017) Liraglutide and renal outcomes in type 2 diabetes. N Engl J Med 377: 839–48
Marso SP, Bain SC, Consoli A et al (2016a) Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 375: 1834–44
Marso SP, Daniels GH, Brown-Frandsen K et al (2016b) Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med 375: 311–22
Meier JJ (2012) GLP-1 receptor agonists for individualized treatment of type 2 diabetes mellitus. Nat Rev Endocrinol 8: 728–42
Milne N (2019) How to use GLP-1 receptor agonist therapy safely and effectively. Diabetes & Primary Care 21: 45–6
Monami M, Nreu B, Scatena A et al (2017) Safety issues with glucagon-like peptide-1 receptor agonists (pancreatitis, pancreatic cancer and cholelithiasis): data from randomized controlled trials. Diabetes Obes Metab 19: 1233–41
Nauck MA, Meier JJ (2016) The incretin effect in healthy individuals and those with type 2 diabetes: physiology, pathophysiology, and response to therapeutic interventions. Lancet Diabetes Endocrinol 4: 525–36
Nauck MA, Meier JJ (2019) Management of endocrine disease: are all GLP-1 agonists equal in the treatment of type 2 diabetes? Eur J Endocrinol 181: R211–234
Nauck M, Stöckmann F, Ebert R, Creutzfeldt W (1986) Reduced incretin effect in type 2 (non-insulin-dependent) diabetes. Diabetologia 29: 46–52
Nauck MA, Meier JJ, Cavender MA et al (2017) Cardiovascular actions and clinical outcomes with glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors. Circulation 136: 849–70
NICE (2015) Type 2 diabetes in adults: management [NG28]. Updated August 2019. NICE, London. Available at: www.nice.org.uk/guidance/ng28 (accessed 21.07.20)
Pfeffer MA, Claggett B, Diaz R et al (2015) Lixisenatide in patients with type 2 diabetes and acute coronary syndrome. N Engl J Med 373: 2247–57
Pratley R, Amod A, Hoff ST et al (2019) Oral semaglutide versus subcutaneous liraglutide and placebo in type 2 diabetes (PIONEER 4): a randomised, double-blind, phase 3a trial. Lancet 394: 39–50
Raccah D (2017) Safety and tolerability of glucagon-like peptide-1 receptor agonists: unresolved and emerging issues. Expert Opin Drug Saf 16: 227–36
Romera I, Cebrián-Cuenca A, Álvarez-Guisasola F et al (2019) A review of practical issues on the use of glucagon-like peptide-1 receptor agonists for the management of type 2 diabetes. Diabetes Ther 10: 5–19
Sharma D, Verma S, Vaidya S et al (2018) Recent updates on GLP-1 agonists: current advancements & challenges. Biomed Pharmacother 108: 952–962
SIGN (2017) Pharmacological management of glycaemic control in people with type 2 diabetes [SIGN 154]. SIGN, Edinburgh. Available at: www.sign.ac.uk/sign-116-and-154-diabetes (accessed 21.07.20)
Singh S, Wright EE Jr, Kwan AY et al (2017) Glucagon-like peptide-1 receptor agonists compared with basal insulins for the treatment of type 2 diabetes mellitus: a systematic review and meta-analysis. Diabetes Obes Metab 19: 228–38
Storgaard H, Cold F, Gluud LL et al (2017) Glucagon-like peptide-1 receptor agonists and risk of acute pancreatitis in patients with type 2 diabetes. Diabetes Obes Metab 19: 906–8
Sun F, Chai S, Li L et al (2015) Effects of glucagon-like peptide-1 receptor agonists on weight loss in patients with type 2 diabetes: a systematic review and network meta-analysis. J Diabetes Res 2015: 157201
Waldrop G, Zhong J, Peters M et al (2018) Incretin-based therapy in type 2 diabetes: an evidence based systematic review and meta-analysis. J Diabetes Complications 32: 113–122
Wysham CH, Lin J, Kuritzky L (2017) Safety and efficacy of a glucagon-like peptide-1 receptor agonist added to basal insulin therapy versus basal insulin with or without a rapid-acting insulin in patients with type 2 diabetes: results of a meta-analysis. Postgrad Med 129: 436–45

Related content
;
Free for all UK & Ireland healthcare professionals

Sign up to all DiabetesontheNet journals

 

By clicking ‘Subscribe’, you are agreeing that DiabetesontheNet.com are able to email you periodic newsletters. You may unsubscribe from these at any time. Your info is safe with us and we will never sell or trade your details. For information please review our Privacy Policy.

Are you a healthcare professional? This website is for healthcare professionals only. To continue, please confirm that you are a healthcare professional below.

We use cookies responsibly to ensure that we give you the best experience on our website. If you continue without changing your browser settings, we’ll assume that you are happy to receive all cookies on this website. Read about how we use cookies.