Abstract
Elderly patients with diabetes are at high risk of polypharmacy because of multiple coexisting diseases and syndromes. Polypharmacy increases the risk of drug–drug and drug–disease interactions in these patients, who may already have age-related sensory and cognitive deficits; such deficits may delay timely communication of early symptoms of adverse drug events. Several glucagon-like peptide-1 receptor agonists (GLP-1 RAs) have been approved for diabetes: liraglutide, exenatide, lixisenatide, dulagluatide, semaglutide, and albiglutide. Some are also approved for treatment of obesity. The current review of literature along with clinical case discussion provides evidence supporting GLP-1 RAs as diabetes medications for polypharmacy reduction in older diabetes patients because of their multiple pleiotropic effects on comorbidities (e.g. hyperlipidemia, hypertension, and fatty liver) and syndromes (e.g. osteoporosis and sleep apnea) that commonly co-occur with diabetes. Using one medication (in this case, GLP-1 RAs) to address multiple conditions may help reduce costs, medication burden, adverse drug events, and medication nonadherence.
Keywords: Diabetes mellitus, Glucagon-like peptide-1 receptor agonists, Geriatric, Comorbidities, polypharmacy, Osteoporosis, Parkinson’s disease, Non-alcoholic fatty liver disease, Sleep apnea, Alzheimer’s
Key points
Glucagon-like peptide-1 (GLP-1) receptor agonists (RAs) have multiple pleiotropic effects that can help reduce polypharmacy in older diabetes patients with co-occurring chronic diseases (e.g. fatty liver disease and hypertension) and syndromes (such as sleep apnea and Parkinson’s disease).
GLP-1 RAs in older patients with diabetes may manifest salutary effects in systems other than the endocrine system.
The multisystemic effects of GLP-1 RAs warrant more rigorous studies to harness their positive effects while minimizing the risk of negative effects.
Introduction
Medications for type 2 diabetes with low risk of hypoglycemia are especially important in the setting of multiple coexisting chronic diseases (multimorbidity) and advanced age. The risks of hypoglycemia and adverse drug events (ADEs) are amplified in the older adults by myriad diabetic complications: chronic kidney disease affecting drug clearance, stroke and peripheral neuropathy affecting manual dexterity, physical ability and cognition, and diabetic retinopathy affecting proper dosing of insulin.1,2 Higher rates of complications and mortality are associated with both longer duration of disease and greater age, as well as coexistence of multiple chronic diseases.3 When diabetes coexist with multiple chronic diseases such as Alzheimer’s and Parkinson’s disease in an older adults, the risk of serious hypoglycemia is further heightened by exposure to multiple medications (polypharmacy), age-related change in drug metabolism and sensory function, and suboptimal adherence to diet and medications. Hypoglycemic events place older adults with diabetes at greater risk for falls, fractures, depression, cardiac arrhythmias and other cardiac events, dementia, and reduced quality of life.4,5 One strategy to reduce hypoglycemic events, polypharmacy, ADEs, cost, and treatment nonadherence in older diabetic patients is to use one medication, such as glucagon-like peptide-1 receptor agonists (GLP-1 RAs), to address multiple conditions and chronic diseases that are commonly associated with diabetes.
Why is it important to favor a strategy of treating many conditions with one therapeutic agent? Many older adults with diabetes present with concomitant multiple conditions and chronic diseases that complicate their treatment, with use of multiple medications with potential for ADEs and burdensome lifestyle changes. A 2011 study found that about two-thirds of Medicare beneficiaries had two or more chronic conditions, including diabetes.6 In addition to the known cardiovascular complications of diabetes (including coronary artery, peripheral artery, and cerebrovascular diseases), diabetes has been associated with nonalcoholic fatty liver disease (NAFLD),7 cognitive impairment and dementia,5 mood disorders, and pain.8 Older adults in general are at higher risk for sleep apnea9,10 and bone loss and fractures.11 This review presents evidence and illustrative cases on the effects of glucagon-like peptide-1 receptor agonists (GLP-1 RAs) on diabetes-associated comorbidities (e.g. NAFLD and hypertension) and geriatric syndromes (e.g. Parkinson’s and sleep apnea) beyond the endocrine system. GLP-1 RAs may allow clinicians, especially geriatricians and primary care physicians, caring for clinically complex diabetes patients, to address multiple symptoms and conditions with one medication—GLP1RAs. Such a ‘one-stop shop’ approach can lead to reduction of medication burden, adverse drug events, hypoglycemic episodes, medication costs, and treatment nonadherence.
The mechanism of action of GLP-1 RAs in diabetes has been well documented in both human and animal studies.12–14 Briefly, the gastrointestinal tract releases GLP-1 when food is ingested. GLP-1 receptor agonists stimulate GLP-1 receptors expressed on pancreatic islet beta cells, but also those expressed in cells in multiple systems, thus potentially explaining the other beneficial effects of GLP-1 RAs,12,13 as discussed below and presented in Table 1.14–54 The use of GLP-1 RAs is associated primarily with dose-related gastrointestinal side effects (nausea, diarrhea, vomiting), and is not indicated in patients at risk for pancreatitis or kidney disease.55 Despite these contraindications, a recent meta-analysis (n = 4330) found GLP-1 receptor agonists superior to alternative incretin-based therapy, dipeptidyl peptidase-4 (DPP-4) inhibitors, in reducing hemoglobin A1c (HbA1c).56
Table 1.
First author (year) | Evidence type | Sample size | Relevant findings | Limitations | Level of evidence* | Clinicaltrials.gov number |
---|---|---|---|---|---|---|
Cardiovascular | ||||||
Sun (2015)14 | Systematic review and meta-analysis | 35 trials with 13 treatments (n = 14,340) | GLP-1 RAs were associated with modest reductions in LDL-C, total cholesterol, and triglycerides but no significant improvement in HDL-C | Further evidence needed to indicate cardiovascular outcomes | 1a | |
Robinson (2013)15 | Systematic review and meta-analysis | 32 trials at least 12 weeks long | Liraglutide or exenatide reduced SBP −1.79 mmHg (95%CI: −2.94, −0.64) versus placebo and −2.39 mmHg (95%CI: −3.35, −1.42) versus active control; DBP reductions were not statistically significant | Shares some studies with Sun (2015)14 | 1a | |
Sun (2015)16 | Systematic review and meta-analysis | 60 trials; (n = 22,890 for SBP; n = 18,795 for DBP) | Bayesian network meta-analysis indicated that exenatide, liraglutide, and albiglutide were ranked as most beneficial among 14 treatments in terms of effect on SBP and DBP | 1a | ||
Marso (2016)17 | LEADER clinical trial | 9340 with established CVD or risk factors | Over a median duration of 3.8 years, liraglutide significantly reduced death from CV causes, nonfatal MI, nonfatal stroke (HR 0.87, 95% CI 0.78–0.97) | 1b | NCT 01179048 | |
Marso (2016)18 | SUSTAIN-6 clinical trial | 3297 with established CVD, CHF or CKD | Over a median duration of 2.1 years, semaglutide significantly reduced death from CV causes, nonfatal MI, nonfatal stroke (HR 0.74, 95% CI 0.58–0.95) | 1b | NCT 01720446 | |
Simó (2015)19 | RCT of exenatide vs glimepiride | 1029 | Over 36 months, those who took metformin plus exenatide (n = 515) had improved HDL and triglycerides. Significantly fewer needed additional hypertensive (20.4 vs 26.4 %; p = 0.026) or lipid-lowering medication (8.4 vs 12.8 %; p = 0.025) | Open-label trial; those not maintaining glycemic control dropped out | 1b | NCT 00359762 |
Armstrong (2016)20 | Double-blind, randomized, placebo-controlled trial | 18 patients | Liraglutide reduced cholesterol-LDL (–0.7 vs. +0.05mmol/l; p < 0.01), ALT (-54 vs. −4.0IU/l; p < 0.01) and serum leptin, adiponectin, and CCL-2 (all p < 0.05) | Small sample size | 1b | NCT 01237119 |
Blackman (2016)21 | Double-blind RCT | 359 nondiabetic obese patients with OSA | Patients took either 3.0 mg liraglutide (n = 180) or placebo (n = 179). Liraglutide reduced SBP compared with placebo but no significant effect on DBP | 2.23% of subjects dropped out during trial | 1b | |
Zhao (2013)22 | Review of preclinical and five clinical studies | 234 patients with MI, heart failure or CAD | Patients improved (LVEF scores or other measures of coronary strength) after hospital infusion of GLP-1 or long-term infusion | Small sample size (studies of 10, 12, 20, 20 and 172 patients) | 2a | |
Katout (2014)23 | Systematic meta-analysis and meta regression study of 33 trials | 12,469 | 12–56 weeks of GLP-1 therapy was associated with a weighted mean difference (WMD) in SBP of −2.22 mmHg (95CI: −2.97 to −1.47); and WMD in DBP of −0.47 (95%CI: −1.20 to −0.25) using a random effect model. Meta-analysis showed a SBP reduction of −1.56 mmHg (95%CI: − 2.78, −1.35) | 2a | ||
Tanaka (2011)24 | Review | Exenatide reduced mean SBP by −3.8 mmHg and DBP −2.3 mmHg vs placebo. Liraglutide also reduced mean SBP (–5.7 mmHg) and DBP (–3.7 mmHg) in obese patients over a 3-week period. Pooled data from 6 trials showed antihypertensive effects of exenatide lasted for 6 months. An open labelled study showed exenatide reduced SBP and DBP for up to 3.5 years vs lifestyle modification alone | Some reviewed studies were open labelled | 2a | ||
Liu (2012)25 | Rat and human experiment | 10 human renal artery specimens | In 10 human renal artery specimens from normotensive and hypertensive patients, EX-4 resulted in nitric oxide production SHR aortic endothelial cells and improved endothelial function in renal arteries from hypertensive patients | Effects of sitagliptin on resistant arteries were not studied | 5 | |
Liver | ||||||
Carbone (2015)26 | Meta-analysis using random effects model | 136 with NAFLD and T2DM | Treatment with GLP-1 agonists or DPP-4 inhibitors (24-48 weeks) significantly decreased serum ALT, a marker for liver inflammation. Studies with imaging showed decreased steatosis, inflammation, and fibrosis | Potential confounding in 36 of 136 participants (19.4%) who also took metformin; lack of controls or tissue samples | 1a | |
Armstrong (2016)20 | Double-blind, randomised, placebo-controlled trial | 18 patients | Liraglutide increased hepatic insulin sensitivity (–9.36 vs. −2.54% vs low-dose insulin; p < 0.05); decreased hepatic de novo lipogenesis in vivo (–1.26 vs. +1.30%; p < 0.05); and decreased lipogenesis in primary human hepatocytes (24.6% vs. untreated controls; p < 0.01) | 1b | NCT 01237119 | |
Klonoff (2008)27 | Placebo controlled trial | 217 patients | Patients (151 with 3.5 years exenatide) with elevated serum ALT at baseline (n = 116) had reduced ALT (–10.4 ± 1.5 IU/L; p < 0.0001) and 41% achieved normal ALT | 1b | ||
Brain | ||||||
Parkinson’s disease | ||||||
Aviles-Olmos (2014)28 | Single blind trial | 44 ‘moderate’ PD patients (20 treated, 24 controls) | At 12 months post-treatment, exenatide treated patients scored 5.6 points higher in MDS-UPDRS motor scale (p = 0.002) and 5.3 points higher on the Mattis DRS-2 (p = 0.006). Previously lowered LED doses were increased by 24 months | 3 patients (1 in treatment group, 2 controls) required DBS during the post-treatment period; analysis was adjusted for these patients. Timed tasks, depression, sleep scores and QOL scores not statistically different | 1b | |
Aviles-Olmos (2013)29 | Single blind trial | 45 ‘moderate’ PD patients | Treated patients improved 2.7 points in MDS-UPDRS at 12 months; untreated patients declined 2.2 points (p = 0.037). Differences persisted to 14 months | Treated patients were slightly older and had slightly shorter disease course than controls. Should be considered ‘proof of concept’ because of sample size; LED reduced in 5 treated patients due to increase in LID | 1b | NCT 01174810 |
Athauda (2017)30, 31 | Phase 2 double blind placebo control trial | 60 ‘mid-stage’ PD patients | Significant improvements in severity of PD motor symptoms persisted after 48 weeks of treatment and 12-week wash-out period | Secondary outcome measures did not reach significance. Cautions to consider the results ‘proof of concept’ rather than ‘proof of efficacy’ | 1b | |
Woert and Mueller (1971)32 | Clinical study | 24 PD patients treated with levodopa | Impaired insulin response and abnormal GTT found in PD patients treated with levodopa that could not be accounted for by age, diet or disease state | 3b | ||
Athauda and Foltynie (2017)33 | Invited review | Need to determine long term safety and efficacy in terms of motor skills; determine mechanism of GLP-1 agonists to determine whether it treats the disease or symptoms | 5 | |||
Li (2016)34 | Review article | Incretins (GLP-1 and GIP) have shown positive results in clinical trials. Similar compounds have been tested in preclinical studies (GIP agonists, DPP-4 inhibitors, OXM, dual GLP-1/GIP receptor agonists, and triple GLP-1/GIP/glucagon receptor agonists | Neurodegenerative diseases = AD, PD, HD, ALS. | 5 | ||
Alzheimer’s Disease | ||||||
Li (2014)35 | Review and meta-analysis | Cross-sectional and longitudinal studies | 7 of 10 major epidemiological studies (n = 2998 stroke patients) reported that those with T2DM have a 2–3 times higher risk of post-stroke dementia | None studied relationship of prediabetes and risk of dementia; different values may exist between studies of T2DM, vascular dementia, AD | 1a | |
Geji (2016)36 | Placebo-controlled double-blinded study | 38 | GLP-1 RAs prevented the decline of brain glucose consumption but had no effect on fibrillary amyloid accumulation or cognition. | Small sample size; 6 month study | 1b | NCT 01469351 |
Craft (2012)37 | randomized, double-blind, placebo-controlled clinical pilot trial | 104 with MCI (64) or mild-to-moderate AD (40) | After 4 months’ treatment, memory improvements in treated group persisted for a further 2 months | 1b | ||
Lerche (2008)38 | Randomized double-blinded placebo-controlled crossover study | 10 healthy men | The cerebral metabolic rate of glucose was reduced by 12–18%
with GLP-1 infusion but the difference was not
significant. GLP-1 reduces BBB transport of glucose in general in the brain and helps to maintain cerebral glucose balance using PET scan and may also have neuroprotective effects linked to both peripheral and cerebral glucose metabolism |
Small sample size | 1b | |
Watson (2005)39 | Placebo-controlled, double-blind, parallel-group pilot study | 30 subjects with mild AD or amnestic MCI | Subjects were randomized to a 6-month course of rosiglitazone (4 mg daily; n = 20) or placebo (n = 10). Memory and attention approved at 6 months in treated subjects | 1b | ||
Bae and Song (2017)40 | Review | Argues for use of GLP-1 RAs against ‘type 3 diabetes’ (AD) | 5 | |||
Bak (2011)41 | Expert opinion | Convincing amount of evidence has shown a beneficial effect of GLP-1 RA treatment on cognitive function | Expert opinion | 5 | ||
Depression/mood disorders | ||||||
Mansur (2017)42 | Open label trial of liraglujtide (4-week pilot) | 19 with MDD or BD | The TMTB (trail making test B) executive function increased significantly from baseline to week 4, as did the DSST (executive function, speed of processing, attention) and RAVLT (learning memory/ acquisition) | Small sample size; open-label design; lack of placebo group or other hypothetical agents; short duration of the study; low statistical power | 2b | |
Grant (2011)43 | Matched groups on exenatide or insulin for poorly controlled T2DM | 71 patients with exenatide, 67 with insulin | Scores of treatment satisfaction, well-being and depression improved for those on exenatide, but not those on insulin (p < 0.05) | 2b | ||
Sleep apnea | ||||||
Amin (2015)44 | Individual RCT | 27 (18 treated, 9 controls) | Overall AHI for treated group decreased from 50 ± 32 to 38 ± 30 events/hour (p = 0.002). In 2.7% of the treated group, AHI declined by 44%, but 30% showed no response to treatment. Controls had no change in AHI | Small sample size, short duration | 1b | |
Blackman (2016)21 | Double-blind RCT | 359 nondiabetic obese patients with OSA | Patients took 3.0 mg liraglutide (n = 180) or placebo (n = 179). Most reduction in mean AHI occurred in first 12 weeks. At 32 weeks, AHI decreased significantly in the liraglutide group vs placebo (12.2 ± 1.8 versus 6.1 ± 2 events /hour). Liraglutide reduced SBP vs with placebo but no significant effect on DBP | 2.23% of subjects dropped out during trial | 1b | |
Matsumoto (2016)45 | Individual cohort study | 96 OSA patients | In patients with varying levels of OSA (43 had 3 months’ CPAP), fasting GLP-1 was significantly higher in those with severe OSA (n = 30), and was positively correlated with AHI | Small sample size; all patients Asian; no control for CPAP group | 2b | |
Reutrakul (2017)46 | Cross sectional study | 71 | Increasing OSA severity was associated with lower GLP1 response to glucose challenge after adjusting for sex, BMI and glycemic status | Small sample size, cross-sectional study | 4 | |
Bone | ||||||
Su (2015)47 | Meta-analysis of 16 RCTs | 11,206 patients | Liraglutide was associated with a significantly lower risk of incident bone fractures (MH-OR = 0.38, 95% CI 0.17–0.87), but exenatide treatment was associated with a doubled risk (MH-OR = 2.09, 95% CI 1.03–4.21) | 1a | ||
Mabilleau (2014)48 | 7 RCTs | 4255 patients in 7 RCTs | Pooled MH-OR for GLP-1 receptor agonists was 0.75 (95% CI 0.28–2.02, p = 0.569) in trials versus DPP-4 | Studies with a follow up <52 weeks had higher MH-OR, but difference was not significant | 1a | NCT 00935532 |
Li (2015)49 | Parallel, randomized, placebo-controlled trial | 62 patients newly diagnosed with T2DM | 24-week treatment with exenatide, insulin or pioglitazone improved glucose control, but had no impact on bone turnover markers or BMD | 1b | ||
Iepsen (2015)50 | RCT | 37 healthy obese women on weight loss diet | Total BMC loss was four times greater in the control group vs the liraglutide group; the latter also had 16% greater bone formation after weight loss and at 52-week follow up | 1b | NCT 02094183 | |
Driessen (2015)51 | Population-based cohort study | 216,816 | GLP-1 RA use not associated with a decreased risk of bone fracture vs. users of other antihyperglycemic drugs | 2b | ||
Driessen (2015)52 | Case control study | 229,114 cases (with fracture), 229,114 controls | Current GLP-1 RA use was not associated with a decreased risk of fracture [adjusted (adj.) OR 1.16; 95%CI 0.83–1.63]. Osteoporotic fracture risk was also not associated with current GLP-1 RA use (adj. OR 0.78; 95%CI 0.44–1.39) | Average GLP-1 RA use was short (36 weeks), perhaps limiting ability to detect association | 4 | |
Skin | ||||||
Faurschou (2014)53 | Case report | 1 59-year old male | Moderate and stable plaque psoriasis of 15-year duration improved immediately and at 3 months with liraglutide titrated to 1.8 mg over 5 weeks | 4 | ||
Hogan (2011)54 | Case series (n = 2) | 2 male patients 48 and 49 years | 6 weeks of liraglutide significantly reduced PASI scores from 13.2 to 10.8 and from 4.8 to 3.8. | 4 |
AHI, apnea hypopnea index; ALS, amyotrophic lateral sclerosis; ALT, alanine transaminase; aMCI, amnestic mild cognitive impairment; ARIT, apomorphine induced rotation test; BBB, blood brain barrier; BD bipolar disorder; BMC, bone mineral content; BMD, bone mineral density; CAD, coronary artery disease; CHF, congestive heart failure; CI (confidence interval); CKD, chronic kidney disease; CPAP, continuous positive airway pressure therapy; CV, cardiovascular; CVD, cardiovascular disease; DBP, diastolic blood pressure; DBS, Deep Brain Stimulation; DPP-4, dipeptidyl peptidase-4; EX-4, extendin 4; GIP, glucose-dependent insulinotropic polypeptide; GLP-1 RAs, glucagon-like peptide-1 receptor agonists; HD, Huntington’s disease, HR, hazard ratio; LED, Levodopa equivalent dose; LID, L-dopa-induced dyskinesia; LVEF, left ventricular ejection fraction; Mattis DRS-2, Mattis Dementia Rating scale; MDD, major depressive disorder; MDS-UPDRS, Movement Disorders Society Unified Parkinson’s Disease Rating Scale; MH-OR, Mantel-Haenszel odds ratio; MI, myocardial infarction; NAFLD; Nonalcoholic fatty liver disease; OXM, oxyntomodulin; PASI, Psoriasis Area and Severity Index; PD, Parkinson’s Disease; QOL, quality of life; RA, receptor agonist; RCT, randomized controlled trial; SBP, systolic blood pressure; WMD, weighted mean difference.
Levels of Evidence for Therapy/Prevention/Etiology/Harm:54
Systematic reviews (with homogeneity) of RCTs.
Individual RCTs (with narrow confidence interval).
All or none RCTs.
Systematic reviews (with homogeneity) of cohort studies.
Individual cohort study or low quality RCTs (e.g. <80% follow-up).
‘Outcomes’ Research; ecological studies.
Systematic review (with homogeneity) of case-control studies.
Individual case-control study.
Case-series (and poor quality cohort and case-control studies).
Expert opinion without explicit critical appraisal, or based on physiology, bench research or ‘first principles’.
Methodology of evidence review
The primary aim of the current review was to characterize the secondary effects of GLP-1 RAs that may be relevant to clinically complex older adults with diabetes and multimorbidity, in whom polypharmacy is a concern.57,58 In September 2017, a PubMed search was conducted for relevant terms, including glucagon-like peptide-1 receptor agonists, exenatide, liraglutide, lixisenatide, albiglutide, dulaglutide, and semaglutide, and suspected secondary effects (e.g. exenatide + cardiovascular); a search of cited and related articles was done in PubMed and Google Scholar. Sources were examined until no new human studies were found. The relevance of each reference was discussed until consensus was obtained. Animal studies were included when human studies were missing. Expert opinion was included when deemed relevant by the authors. A summary of results (Table 1) was developed using previous guides.54,57 Drugs were evaluated based on the effect on a secondary system (i.e. blood lipids, hypertension, liver, brain, etc.) and effects refined as indicated. Levels of Evidence for Therapy/Prevention/Etiology/Harm: 1a, Systematic reviews (with homogeneity) of randomized controlled trials (RCTs); 1b: Individual RCTs (with narrow confidence interval); 1c: All-or-none RCTs; 2a: Systematic reviews (with homogeneity) of cohort studies; 2b: Individual cohort study or low quality RCTs (e.g. <80% follow-up); 2c: ‘Outcomes’ Research; ecological studies; 3a: Systematic review (with homogeneity) of case-control studies; 3b: Individual case-control study; 4: Case-series (and poor quality cohort and case-control studies); and 5: Expert opinion without explicit critical appraisal, or based on physiology, bench research or ‘first principles’.54
Secondary effects and mechanism of action of GLP-1RAs on comorbidities: evidence from clinical and basic science research
Effects on the cardiovascular system
The effect of GLP-1 receptor agonists on triglyceride, low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C) levels has been evaluated in multiple studies. In the meta-analysis by Sun and colleagues, 35 trials were analyzed for the effects of exenatide, liraglutide, or taspoglutide on lipid profile.14 GLP-1 agonist therapy was associated with modest decreases in LDL-C, triglycerides, and total cholesterol, but no significant effect on HDL-C. Compared with placebo, the GLP-1 agonist group demonstrated a reduction in LDL-C from −0.08 mmol/L to −0.16 mmol/L.14 Exenatide, liraglutide, and taspoglutide treatment led to a decrease in total cholesterol, while liraglutide and taspoglutide led to a reduction in triglyceride levels.14 Other studies showed a reduction in one or more lipid levels with exenatide or liraglutide.19,20
Hypertension, found in 30–70% of those with type 2 diabetes, is a well-known risk factor for both cardiovascular and cerebrovascular disease.24 The mechanisms for the antihypertensive effects of GLP-1 RAs include a reduction in the risk of left ventricular hypertrophy, an increase in sodium excretion and increased atrial natriuretic peptide (ANP) secretion, leading to reduced blood pressure (BP).22 Indeed, GLP-1 receptor-deficit mice secreted no ANP when given GLP-1 RAs, indicating a putative BP-regulating pathway via a gut-heart GLP-1 receptor-dependent and ANP-dependent axis.22 In humans, Zhao found strong evidence from clinical and preclinical studies that GLP-1 RA treatment can benefit patients with myocardial ischemia and heart failure.22 Sun and colleagues, in a second systematic review and network meta-analysis of 26,654 patients, found exenatide to significantly reduce systolic blood pressure (SBP) compared with insulin (–4.86 mmHg 95%CI: −8.33, −1.40) or sulphonylurea (–3.00 mmHg 95% CI: −5.84,–1.35). In the same review, albiglutide reduced BP compared with placebo but not when compared with other treatments. Exenatide also reduced diastolic blood pressure (DBP) significantly (–0.9 mmHg 95%CI: −1.68, −0.11), as did sulphonylurea (–1.60 mmHg 95%CI: −2.86, −0.35). Dulaglutide produced no significant effect.16 A similar meta-analysis of 32 trials found that liraglutide or exenatide reduced SBP −1.79 mmHg (95%CI: −2.94, −0.64) versus placebo and −2.39 mmHg (95%CI: −3.35, −1.42) versus active control; DBP reductions were not statistically significant.15 Liraglutide also reduced mean SBP (–5.7 mmHg) and DBP (–3.7 mmHg) in obese patients over a 3-week period.24
Long-term studies indicate a sustained effect of GLP-1 RAs on BP. For example, in a systematic meta-analysis and meta-regression study of 33 trials covering 12–56 weeks (n = 12,469), Katout and colleagues found a weighted mean difference (WMD) in SBP reduction of −2.22 mmHg (95%CI: −2.97 to −1.47), and WMD in DBP reduction of −0.47 mmHg (95%CI: −1.20 to −0.25) in the GLP-1 RAs group versus control.23 The greatest BP-lowering effect was found with exenatide, which reduced mean SBP by −3.8 mmHg at 5–10 mcg and DBP −2.3 mmHg at 10 mcg versus placebo. Data from six large trials showed that the antihypertensive effects of exenatide lasted 6 months, with greatest reduction in patients with SBP greater than 150 mmHg. In another open labeled study, exenatide treatment for up to 3.5 years reduced SBP and DBP in diabetic patients, in comparison with lifestyle modification alone.24
The effect of GLP-1 RAs on cardiovascular health has been studied in a number of large trials. Four trials of the cardiovascular effects of GLP-1 RAs in patients with diabetes and cardiovascular risk factors have been published: ELIXA (lixisenatide), LEADER (liraglutide), SUSTAIN-6 (semaglutide), and EXSCEL (exenatide extended release).17,18,59,60 Of these, LEADER and SUSTAIN-6 showed significant reductions in the primary outcome and reduction in cardiovascular death. Liraglutide reduced the primary outcome (a combination of first occurrence of death from cardiovascular causes, nonfatal myocardial infarction, including silent, or nonfatal stroke) 13% [hazard ratio (HR) 0.87, 95% CI 0.78, 0.97] and reduced cardiovascular death 22% over a median follow up of 3.8 years.17 Semaglutide, over a median follow up of 2.1 years, reduced the same primary outcome 26% (HR 0.74, 95% CI 0.58, 0.95) but did not significantly affect cardiovascular death.18 A meta-analysis of seven trials, including LEADER and SUSTAIN-6, concurs that liraglutide and semaglutide both offer protection from adverse cardiovascular events, but only liraglutide reduced cardiovascular mortality.61 One review recommended that GLP-1 RAs could replace metformin as a first-line therapy in those with type 2 diabetes with high cardiovascular risk factors or those who are intolerant to metformin.62 The PIONEER 6 trial, to determine the cardiovascular safety of semaglutide in type 2 diabetes patients with high cardiovascular risk, is currently ongoing and has enrolled 3183 patients in 21 countries (NCT02692716).63
Effects in the liver
NAFLD is common in patients with type 2 diabetes.7 Indeed, a 2015 systematic review in JAMA estimates 66% of adults over 50 who are overweight and have diabetes are also likely to have the NAFLD subtype, nonalcoholic steatohepatitis with advanced fibrosis.64 NAFLD and diabetes together worsen hepatic function and hasten development of diabetes complications.7 Mechanistic evidence from several animal studies indicate that treatment of diabetes with GLP-1 RAs affects hepatic function both directly and indirectly. Treatment of mice with exenatide for 60 days significantly decreased hepatic lipid content.65 In mice fed a high fat/fructose diet, the liraglutide group demonstrated a significant reduction in hepatic lipid accumulation as well as significant improvements in insulin sensitivity and glucose tolerance, lower serum triglyceride and cholesterol levels.66
Multiple clinical trials and meta-analyses show improved hepatic function in NAFLD as measured by transaminase levels, biopsy, and images. In studies of patients with both type 2 diabetes and NAFLD (diagnosed by imaging or biopsy), GLP-1 receptor agonists (liraglutide and/or exenatide) and a DPP-4 inhibitor (sitagliptin) lowered serum alanine aminotransferase (ALT) levels by a mean of 14.1 IU/L. Other studies of GLP-1 RAs using ultrasound or proton magnetic resonance spectroscopy assessment of fat content or liver biopsy showed a 42% median relative reduction in intrahepatocellular lipid in imaging, and significant histological improvement in the biopsy group.26 Other RCTs found similar positive results: an average reduction of ALT of −54 IU/L in the liraglutide group versus −4.0 IU/L in the placebo group, with p < 0.01 and a significant decrease in de novo lipogenesis in the liraglutide group (−1.26% versus +1.30%; p < 0.05);31 and a mean ALT reduction over 3 years in 217 diabetes patients values, with mean ALT change of −10.4 +/– 1.5 IU/L; (p < 0.0001) in a 3-year clinical trial of exenatide therapy.27
Effects in the brain
GLP-1 receptors are present in many areas of the brain, including neurons, glia, and astrocytes. Studies show an abundance of GLP-1 receptors in the central nervous system (CNS), especially in areas of the brain critical for memory, learning, and locomotion.67–69 These include the pyramidal neurons of the hippocampus and the cerebral cortex. In addition, activated glial cells express GLP-1 receptors under stress, suggesting a potential role for GLP-1 RAs in modulating the inflammatory response.67–70 Han and colleagues found that GLP-1 RAs and GLP-1 receptors are produced by the hippocampus, first as a post-translational product of preglucagon, acting as a growth factor in the brain.71 GLP-1 is neurotrophic via its protection of neurons against glutamate-mediated apoptosis and oxidative injury. Indeed, both mice and rats overexpressing hippocampal GLP-1 receptors experienced substantial neurotic growth, learned faster, and had better memory.70–80 In addition, administration of liraglutide significantly protected against amyloid beta 25–35-induced reduction in spatial cognition in rats.71–74 Similar anti-amyloid effects were demonstrated with lixisenatide.72
Parkinson’s disease
The evidence from basic science research, mostly in animal models,80,82–85 led to human studies and clinical trials of GLP-1 RAs,28–32 especially since GLP-1 agonists/analogs have been shown to cross the blood-brain-barrier (BBB) and stimulate the GLP-1 receptor in the brain.37,81 Lerche and colleagues found that GLP-1 RAs modulate transport of glucose through the BBB and help maintain cerebral glucose balance, as shown by PET scans. This function may have neuroprotective effects via peripheral and cerebral glucose metabolic pathways that may benefit Parkinson’s Disease (PD) patients.37 A 12-month trial of exenatide in 44 ‘moderate’ PD patients (20 treated, 24 controls) found that the treated patients improved by 2.7 points in Movement Disorders Society Unified Parkinson’s Disease Rating Scale (MDS-UPDRS) scores, while untreated patients declined 2.2 points (p = 0.037); these differences lasted for 14 months.29 In an open-labelled RCT of 45 patients with PD, a mean improvement of 1.8 points in the Mattis dementia rating scale, was observed at 24 months in patients treated with exenatide, compared with a mean deterioration of 3.5 in the control group.28 Similarly, in a single center, double-blind, randomized placebo-controlled 60-week trial with 62 participants,30 the exenatide group had MDS-UPDRS scores 3.5 points lower than the placebo group. This improvement in severity of PD motor systems persisted for 48 weeks of treatment and through the 2-week wash-out period.20 These results are considered ‘proof of concept’ rather than ‘proof of efficacy,’ and larger and longer RCT studies are needed to confirm these findings.86
Alzheimer’s disease
Impairment of short-term or long-term memory and learning is a key symptom of Alzheimer’s disease (AD), the cause of 60–80% of all cases of dementia in older adults.87,88 Thus, there is an urgent need for more effective medications to treat AD and other neurodegenerative diseases.89,90 GLP-1 RAs in particular have shown promise as therapy for AD and other causes of dementia.87,89,90 The hallmark of AD is the accumulation of amyloid plaques in the hippocampus and cerebral cortex,72,88 and the effects of GLP-1 RAs on reducing amyloid load and neuroinflammation are documented.78,79 Type 2 diabetes and AD share several clinical and pathologic characteristics, including the role of insulin signaling in neuroprotection, neuronal stem cell activation, and general cell growth.39,90 GLP-1 RAs help normalize insulin signaling,91 and GLP-1 receptors are located widely throughout the brain.92–94
In a 26-week, placebo-controlled double-blinded intervention (n = 38) in AD patients with long-standing diabetes, the use of liraglutide prevented the expected decline of cerebral metabolic rate for glucose uptake (using FDG-PET) in the parieto-temporal frontal and posterior cingulate cortices.35 Clinical studies on GLP-1 RAs provide evidence of promising neuroprotective, and even regenerative, properties that may be utilized in patients with AD or PD.28–32,35–38 Given further clinical evidence, they have the potential to be the therapeutic agent of choice for diabetic patients with AD.28,40,89,90 For example, in clinical trials, exenatide-treated patients showed a clear improvement in cognitive score, suggesting a beneficial effect on cognition and memory; this benefit was still visible 1 year after the trials.28 Optimism for the benefit of GLP-1 RAs in AD has recently been dampened, however. A recent Cochrane review found no relationship between the use of diabetes medications and cognitive decline during a follow-up period of 40–60 months,95 and a recent review of clinical treatment of diabetes in the elderly argues for a holistic approach that incorporates caregiver input, screening for geriatric syndromes, functional medicine, and periodic re-evaluation of targets and strategies.96
Depression/mood disorders
Mood disorders (major depressive and anxiety disorders) are common coexisting chronic diseases in diabetic patients, which increase morbidity, disability, and mortality in these patients.8 GLP-1 receptor agonists have been found to affect areas of the brain associated with emotional regulation.97 In a meta-analysis, McIntyre and colleagues found improvement in mood and cognition with GLP-1 RAs, regardless of psychiatric diagnoses.98 More recently, Billings and colleagues found that those taking weekly GLP1-RAs (exenatide, dulaglutide, or semaglutide) had greater health-related quality of life (i.e. improved treatment satisfaction and willingness to continue treatment) over those taking daily GLP-1 RAs; weekly semaglutide was associated with the greatest treatment satisfaction.99 The cognitive effects of GLP-1 RAs are especially important in patients with diabetes and psychiatric comorbidities, given the high prevalence of cognitive impairment and antipsychotic related metabolic syndrome in this population.100 In a case-controlled study of diabetic patients (n = 138), Grant and colleagues found that exenatide-treated patients had significantly reduced Hospital Anxiety and Depression Scale (HADS) scores at 6 months when compared with the insulin-treated group, indicating greater treatment satisfaction and higher wellbeing.42 Another open labelled trial with 19 patients showed a significant increase from baseline to week 4 in scores for the Trail Making Test B (executive function), in Digit Symbol Substitution Test (DSST; executive function, speed of processing, attention) and in the Rey Auditory Verbal Learning Test (RAVLT; learning memory)/acquisition), in response to liraglutide, especially in patients with comorbid bipolar disorder (BD) or major depressive disorder (MDD) and cognitive impairment.41
The mechanisms for the above findings have not been well described but initial studies provide some insight. Sharma and colleagues found that liraglutide reversed the behavioral depression and metabolic abnormalities (weight gain, increased blood lipids) associated with long-term atypical antipsychotic treatment in rats.100 In another experimental rat model, Anderberg and colleagues found that liraglutide reversed the antipsychotic-induced metabolic abnormalities in rats, and that chronic injections of exenatide were neither anxiogenic nor anxiolytic, but caused a reduction in depression-like behavior.97 The presence of GLP-1 receptor in the dorsal raphe (DR), a major area of serotonergic neurons, suggests a potential role for GLP-1 RAs in emotional processing and mood regulation. Activation of the DR-GLP-1 receptor alone is sufficient to increase anxiety-like behavior − an effect similar to findings in the initial use of selective serotonin reuptake inhibitors.97 In summary, use of GLP-1 RAs in older multimorbid diabetes patients with depression and bipolar and other psychiatric disorders may help improve mood and cognition, while reducing the metabolic side effects of antipsychotics such as obesity, hyperlipidemia, and hypertension.100
However, those with nondementia psychoses are likely to be unaffected, or any effect is not clinically relevant. A recent study from Denmark found no cognitive improvement from exenatide in obese patients with clinically stable schizophrenia who were also receiving antipsychotic treatment. The authors noted, however, that 3 months may not have been enough time for cognitive effects to manifest. These subjects were primarily young and middle-aged adults, and none were over 65 years (exenatide group: n = 23, age 37.1 ± 10.6, range 19–65 years; placebo group: n = 22, age 34.5 ± 10.1, range 19–56 years).101
Pain
Pain syndromes of multifactorial etiologies are highly prevalent, but potentially treatable, causes of excess disability in diabetes patients.8 GLP-1 RAs may be useful in pain management. Gong and colleagues, in a rat model experiment, found GLP-1 receptors expressed in spinal microglia and upregulated after peripheral nerve injury.102 When GLP-1 RAs stimulate spinal microglia GLP-1 receptors, beta-endorphins are released and pain diminishes. Exenatide and GLP-1 have been associated with a decrease in pain hypersensitivity via its antinocioceptive effect, an effect mediated by activation of GLP-1 receptors. This effect can be blocked by naloxone, suggesting a modulatory effect on opioidergic pathways.102,103
Sleep apnea
Obstructive sleep apnea (OSA) symptoms include snoring, apneas during sleep, and excessive daytime somnolence.9,10 Untreated, OSA has been linked to hypertension, stroke, congestive heart failure, type 2 diabetes, and depression. OSA is common in mid and later life especially in older obese men.9,10 In an RCT of 30 OSA subjects, Amin and colleagues found that the overall apnea-hypopnea index (AHI) for patients treated with liraglutide decreased from 50+ or −32 to 38+ or −30 (p = 0.002). In 70% of the treated group, AHI declined by 44% or 20+ or −12 events per hour, while 30% showed no response to treatment. AHI was 32.6+ or −21 versus 33.2+ or −21 at follow up. Body mass index (BMI) did not change from baseline in either group.43 In another double blinded RCT (n = 359), Blackman and colleagues found that the greatest reduction in AHI occurred in the first 12 weeks of treatment and minimally thereafter. In the liraglutide group, AHI was significantly reduced after 32 weeks compared with placebo, independent of age, baseline BMI or OSA severity. Fewer subjects in the liraglutide group met the criteria for OSA diagnosis after treatment.21 A cross-sectional study by Reutrakyl and colleagues (n = 70) found that increasing OSA was associated with a lower GLP-1 response to a glucose challenge after adjusting for sex, BMI, and glycemic status.45 In an individual cohort study, Matsumoto and colleagues (n = 96) found higher fasting GLP-1 levels in a severe OSA group, and concluded that this was a compensatory mechanism to increase insulin levels to prevent the development of diabetes, since hypoxia leads to pancreatic damage.44 The benefit of GLP-1 RAs may therefore increase as the severity of OSA increases.
Effects in bone
Evidence, mostly from basic science research (largely in rodents and in vitro studies), supports the salutary effects of GLP-1 RAs on bone mass and strength reported in human studies; however, the effect on fracture prevention is less clear. Overall, research shows the presence of GLP-1 receptors in bone tissue, especially in osteoblasts, osteocytes, and osteoclasts.104–109 The thyroid parafollicular cells, known as C-cells, make a peptide hormone called calcitonin that controls calcium homeostasis. A rise in serum calcium levels stimulates calcitonin secretion from C-cells, which, in turn, inhibits osteoclast-induced bone resorption. The C-cells express GLP-1 receptors; C-cell activation suppresses bone-resorption activity and osteoclast proliferation.104–106 The localization of GLP-1 receptors in bone and C-cells has led to various mechanistic studies explaining the anti-resorptive and bone-building effects of GLP-1 RAs. These mechanisms include an increase in the activity of osteoblast in response to GLP1R agonist; an increase in the activity of C-cells, which leads to increased levels of calcitonin − a potent inhibitor of osteoclast-induced bone resorption; and improvement in collagen synthesis, bone blood supply, and bone architecture.106–109
Reports of the effect of GLP-1 agonist on bone integrity in humans are mixed, ranging from no difference between GLP-1 agonist and other antidiabetic medications,48,50 to significant reduction in fractures with liraglutide and a higher rate of fractures with exenatide.46 The meta-analysis by Mabileau and colleagues analyzed the fracture rate of seven RCTs, and found that GLP-1 receptor agonists (specifically, exenatide in three studies, liraglutide in four) did not significantly reduce incidents of bone fracture in comparison with other antidiabetic medications.47 Another meta-analysis by Su and colleagues of 16 RCTs using GLP-1 agonists showed that patients receiving liraglutide had a significantly decreased risk of bone fracture, whereas those receiving exenatide had an increased risk of bone fracture.46 The bone-protective effect of liraglutide was further supported by Iepsen and colleagues in their RCT of 17 obese women randomized to liraglutide or placebo; liraglutide increased bone formation by 16% and prevented the bone loss associated with weight loss.49 A more recent Bayesean network meta-analysis of many of the same studies found that exenatide was the safest GLP-1 RA to prescribe to reduce fracture risk, followed by dulaglutide, then liraglutide, albiglutide, lixisenatide, and finally semaglutide.110 Other controlled trials and cohort studies have also shown no significant difference between the GLP-1 agonist group and the control group using other anti-glycemic drugs.50,51 However, these studies failed to separate out the effect of different GLP-1 agonist medications (liraglutide versus exenatide versus others). Some of the inconsistency between trials may reflect differences among patients in dose and duration of GLP-1 RAs, type and number of coprescribed medications, severity of diabetes, adherence to diabetes medications, and comorbidity burden. Additional studies are needed to clarify the effect of different GLP-1 receptor agonists on bone formation.
Effects on skin
The evidence for GLP-1 RAs on skin diseases is still very preliminary, with antioxidant, anti-inflammatory, and neuromodulatory mechanisms likely responsible for the effects. In a case report by Faurschou and colleagues, psoriasis was found to clear after liraglutide was prescribed for the patient’s type 2 diabetes.52 In another case, the patient was taken off liraglutide and psoriasis returned; two subsequent patients showed lower Psoriasis Area and Severity Index (PASI) scores with treatment.53 In all four cases, PASI scores lowered immediately. Another report showed plaque clearance with sitagliptin.111 Glycemic control was not associated with plaque clearance, suggesting another mechanism, perhaps anti-inflammatory or the operation of T-cells.52,53,112
Contraindications
Important clinically relevant adverse effects (nausea, vomiting, and diarrhea) warrant mention.113–116 These adverse effects are extensions of the pharmacological effects of the GLP-1 RAs, especially when they are started at higher doses in frail elders with dementia and multiple coexisting morbidities. Indeed, the side effects of nausea (about 50%, mostly mild and transient) and vomiting (about 10%) were the reasons most commonly reported for discontinuing GLP-1 RAs, which occurs in about 5% of patients. Rarely, the gastrointestinal side effects can lead to dehydration and acute kidney injury.55 The severity of gastrointestinal side effects depends on both the dose of the GLP-1 RAs and the coprescribed drug, such as metformin. Lowering the dose, using long-acting GLP-1 RAs, and avoiding prodiarrhea/emetic drugs (e.g. metformin or high-dose cholinesterase inhibitors) can reduce the incidence of side effects.116
In a systematic analysis of 32 clinical trials with GLP-1 RAs, Bettge and colleagues found a significant relationship between the risk of nausea and diarrhea and the dose of long-acting GLP-1 RAs; however, this relationship was not significant for vomiting. They also found that concomitant use of metformin significantly increased the odds of nausea (p = 0.04) and vomiting (p = 0.0009).116 Among all GLP-1 RAs, lixisenatide treatment resulted in the least nausea and vomiting, and semaglutide the most, while all long-acting GLP-1 RAs had lower rates of nausea and vomiting, but higher rate of diarrhea, than short-acting ones.116–118 Minor side effects, such as injection site reactions, headache, and nasopharyngitis, rarely led to drug discontinuation and were usually transient. Because of the pancreatic and thyroid tissue effects of GLP-1 RAs, reports of adverse effects from animal studies and database analyses have raised concerns about the potential for increased risk pancreatitis, pancreatic cancer, and thyroid cancer in GLP-1 RA users, but no cause and effect associations have been seen in any of these studies.113–116 Most recently, once-weekly semaglutide has been associated with an increased risk of diabetic retinopathy complications.119 Nevertheless, patients on GLP-1 RAs should be monitored for these side effects if the clinical scenario fits. Although GLP-1 RAs do not cause hypoglycemia, all clinicians should reduce or discontinue concomitant sulphonylurea or insulin to avoid hypoglycemia.
Illustrative cases
Case #1
A 76-year-old woman was seen regularly by her primary care physician (PCP) for type 2 diabetes, hypertension, hypertriglyceridemia, and vascular dementia/AD. The duration of her conditions were unclear except that she had been on antihypertensive, antilipid and antidiabetes medications for at least 6 years. Her medications included donepezil 10 mg daily, hydrochlorothiazide 25 mg daily, lisinopril 20 mg daily, sitagliptin 100 mg daily, metformin 1000 mg twice daily, pravastatin 10 mg daily, gemfibrozil 600 mg twice daily, omega-3-fatty acid 2000 mg twice daily, aspirin 81 mg daily, and insulin glargine 50–55 units subcutaneous injection daily. Diabetes remained uncontrolled, with HbA1c levels at 12–13 and BMI at 29 kg/m2. Because of prior episodes of recurrent hypoglycemia in the setting of dementia [Mini-Mental State Examination (MMSE) = 25/30], the patient’s caregiver and clinician agreed to a noninsulin approach for glycemic control. Thus, the patient was started on weekly injections of 2 mg exenatide, changed 5 months later to daily 1.2 mg liraglutide injections. Eight months thereafter, hypoglycemia episodes abated, HbA1c levels fell to 5.3%, BMI to 23 kg/m2, triglycerides from 849 mg/dl to 250 mg/dl, total cholesterol from 202 mg/dl to 111 mg/dl, thyroid stimulating hormone (TSH) from 1.65 mIU/l to 0.80 mIU/l (normal range 0.45–4.70 mIU/l), and SBPs averaged in the 100s mmHg. Because of the aforementioned numbers, indicating tight control of blood pressure, diabetes, and lipids, all medications were stopped except vitamin D3 and aspirin. The patient’s family stopped donepezil because of concerns about additional weight loss and low appetite. After 3 months of cessation, lisinopril, and liraglutide were restarted due to an increase in HbA1c levels to 8% and SBP into the 140s mmHg. MMSE done at the 1-year follow-up visit was 21. Because the patient had nausea on a full GLP-1 RA dose, the patient’s caregiver decided on her own, against the physician’s instruction for daily dosing, to reduce daily liraglutide to three times per week. The nausea abated. The HbA1c level was 7.3% at the 4-month follow up.
Case #2
A 73-year-old man was being followed by his PCP for type 2 diabetes, AD and vascular dementia, low energy, OSA on a continuous positive airway pressure (CPAP) machine, obesity (BMI 43 kg/m2), uncontrolled hypertension (SBP in the 170s mmHg), benign prostatic hyperplasia, hypothyroidism, depression, hyperlipidemia, and seasonal allergic rhinitis. The patient had received a diagnosis of dementia 2 years prior to his first PCP visit. He had been on medications for the rest of his medical conditions for at least 5 years prior to his visit, but the patient and his spouse were unsure of the onset and duration of his conditions. The patient received a once-a-day dose of the following medications: amlodipine 5 mg, losartan 100 mg, hydrochlorothiazide 25 mg, levothyroxine 25 µg, sitagliptin 100 mg, bupropion SR 100 mg, finasteride 5 mg, rivastigmine patch 9.5 mg, simvastatin 20 mg, aspirin 81 mg, and venlafaxine 37.5 mg. Fexofenadine 60 mg was twice daily. Diabetes control was adequate for age, with multiple HbA1c results ranging between 7% and 8%. The patient’s BMI remained at 43 kg/m2 with worsening of his mood (Patient Health Questionnaire-2, PHQ-2), energy, and OSA. MMSE was 22. His Geriatric Depression Scale (GDS) score was zero, but his PHQ-2 screen positive for depression. His SBP remained in the 170s mmHg despite three drugs. His tests showed total cholesterol was 222 mg/dl, LDL was 147 mg/dl, and TSH was 4.28 mIU/l. After a discussion with the patient and his spouse about using GLP-1 RAs for diabetes and weight, the patient was started on subcutaneous exenatide 2 mg per week. His sitagliptin was stopped. Simvastatin was changed to pravastatin, which is hydrophilic and thus has lower cognitive side effects. His venlafaxine was increased to 75 mg daily to further help his depression as well as his mild anxiety symptoms. About 6 months thereafter, the patient’s average SBP remained in the 130s mmHg, his BMI remained around 39 kg/m2, and his HbA1c levels ranged from 7% to 8%. His MMSE was 17, despite the family reporting an improvement in patient’s conversational skills, attention, and memory for people names. His repeat blood tests showed LDL was 103 mg/dl, total cholesterol was 180 mg/dl, and TSH was1.91 mIU/l. Both his GDS and PHQ-2 depression screens were negative. The patient reported being more energetic, sleeping better, and feeling less depressed, prompting discontinuation of his bupropion (which, according to his spouse, worsened his sleep). The patient’s family stopped fexofenadine because of abatement of the allergic rhinitis symptoms and concerns about the patient taking too many pills. Sitagliptin was stopped due to overlapping mechanism of action (incretin-based therapeutics) with GLP-1 RAs. The patient continued to tolerate the exenatide with no reports of nausea, vomiting, diarrhea, or abdominal pain (known side effects of exenatide). After up to 15 months of periodic follow-up, the patient remained nondepressed and free of gastrointestinal side effects.
The improvement in HbA1c, weight, lipid profile, and blood pressure in the cases described above (especially in our first patient) is consistent with recent findings from the SUSTAIN-5 Trial, a 30-week multicountry randomized double-blind, placebo-controlled trial of weekly semaglutide 0.5 or 1.0 mg among 397 diabetes patients on stable basal insulin dose.120 However, unlike the subjects in the SUSTAIN-5 trial, basal insulin was discontinued in our elderly dementia patient, because of recurrent hypoglycemia episodes.
Clinical implications
A key approach from our clinical experience to reducing the risk of GLP-1 RA-related gastrointestinal side effects is by starting at a low dose (e.g. at 25% of recommended starting dose in frail elders) and by slow titration over months instead of weeks. Another approach is switching to a different GLP-1 RA, as was done for Case #1 above, as the minor pharmacokinetic and dynamic differences between GL-1 RAs may alter an individual patient’s risk of adverse effects. The preponderance and variety of these GLP-1 RAs studies (both human and animal, and in vitro) would seem to indicate the need for larger, more rigorous, studies. In particular, more understanding is required of how genetic differences in drug metabolism and GLP-RA effects on renal functions among patients alter the secondary effects of GLP-1 RAs and the mechanisms behind these differences.121–123 For example, recent evidence supports the renoprotective effect (via reduced proteinuria and increased natriuresis) of GLP-1RAs: a slowing of progression of diabetic kidney disease, and a better preservation of renal functions in diabetes patients with CKD.122,123 Future studies are needed to understand the roles of genomics and metabolomics in determining the person-to-person differences in response to GLP-1RAs. Data from such studies and others have the potential to inform diabetes clinical practice guidelines vis-à-vis the use of GLP-1RAs.
One limitation of this review is that many of the studies lack the ‘gold standard’ rigor of RCTs. The number and variety of effects do, however, appear to indicate a trend. RCTs or database analysis of a large number of patients may be the next step before recommending prescribing changes.
Conclusion
GLP-1 RAs have multiple pleiotropic activities that may help reduce polypharmacy in elderly diabetes patients living with other serious comorbidities. The cases and evidence presented here support the potential for GLP-1 RAs to reduce the incidence or symptoms of those conditions (i.e. PD, osteoporosis, NAFLD, lipoprotein disorders, and hypertension), among other common comorbidities in older adults with diabetes. The effectiveness of GLP-1 RAs in patients with diabetes with impairments in systems other than the endocrine system − cardiovascular, liver, brain, bone, and skin − may reflect the multisystemic role of GLP-1 receptor functions. Inasmuch as the effects of GLP-1 RAs (both positive and negative) are multisystemic, they warrant more rigorous studies on how to harness the positive effects while minimizing the risk of negative effects. The usefulness of GLP-1 RAs in reducing polypharmacy and medication costs in patients with diabetes merits further study in a large randomized, controlled, clinically comparative trial.
Footnotes
Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article.
Conflict of interest statement: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Ethical standards: The UTMB Institutional Review Board (IRB) has deemed this study exempt from IRB review.
ORCID iD: Mukaila A. Raji https://orcid.org/0000-0002-7460-7281
Contributor Information
Olusola F. Onoviran, Department of Internal Medicine, University of Texas Medical Branch, Galveston, USA
Dongming Li, Department of Internal Medicine, University of Texas Medical Branch, Galveston, USA.
Sarah Toombs Smith, Department of Internal Medicine, University of Texas Medical Branch, Galveston, USA; Sealy Center on Aging, University of Texas Medical Branch, Galveston, USA.
Mukaila A. Raji, Division of Geriatric Medicine, Department of Internal Medicine, The University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0177, USA.
References
- 1. World Health Organization. Global report on diabetes. World Health Organization, 2016. http://apps.who.int/iris/bitstream/handle/10665/204871/9789241565257_eng.pdf;jsessionid=3C9A02BAAB6F463CFEEA1DAD53C23D6B?sequence=1 (accessed 3 December 2018). [Google Scholar]
- 2. Centers for Disease Control and Prevention. National diabetes statistics report, 2017. Atlanta, GA: Centers for Disease Control and Prevention, U.S. Department of Health and Human Services, 2017. Available at https://www.cdc.gov/diabetes/pdfs/data/statistics/national-diabetes-statistics-report.pdf. Accessed December 3, 2018. [Google Scholar]
- 3. Huang ES, Laiteerapong N, Liu JY, et al. Rates of complications and mortality in older patients with diabetes mellitus: the diabetes and aging study. JAMA Intern Med 2014; 174: 251–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Bansal N, Dhaliwal R, Weinstock RS. Management of diabetes in the elderly. Med Clin N Am 2015; 99: 351–377. [DOI] [PubMed] [Google Scholar]
- 5. Huang ES. Management of diabetes mellitus in older people with comorbidities. BMJ 2016; 353: i2200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Lochner KA, Goodman RA, Posner S, et al. Multiple chronic conditions among Medicare beneficiaries: state-level variations in prevalence, utilization, and cost, 2011. Medicare Medicaid Res Rev 2013; 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Hazlehurst JM, Woods C, Marjot T, et al. Non-alcoholic fatty liver disease and diabetes. Metabolism 2016; 65: 1096–1108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Beard HA, Markides K, Al Ghatrif M, et al. Trends in diabetes medications use and prevalence of geriatric syndromes in older Mexican Americans from 1993/1994 to 2004/2005. Ann Pharmacother 2010; 44: 1376–1383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Lévy P, Kohler M, McNicholas WT, et al. Obstructive sleep apnoea syndrome. Nat Rev Dis Primers 2015; 1: 15015. [DOI] [PubMed] [Google Scholar]
- 10. McEvoy RD, Antic NA, Heeley E, et al. CPAP for prevention of cardiovascular events in obstructive sleep apnea. N Engl J Med 2016; 375: 919–931. [DOI] [PubMed] [Google Scholar]
- 11. Alejandro P, Constantinescu F. A review of osteoporosis in the older adult: an update. Rheum Dis Clin North Am 2018; 44: 437–451. [DOI] [PubMed] [Google Scholar]
- 12. Jazayeri A, Rappas M, Brown AJH, et al. Crystal structure of the GLP-1 receptor bound to a peptide agonist. Nature 2017; 546: 254–258. [DOI] [PubMed] [Google Scholar]
- 13. Chatterjee S, Ghosal S, Chatterjee S. Glucagon-like peptide-1 receptor agonists favorably address all components of metabolic syndrome. World J Diabetes 2016; 7: 441–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Sun F, Wu S, Wang J, et al. Effect of glucagon-like peptide-1 receptor agonists on lipid profiles among type 2 diabetes: a systematic review and network meta-analysis. Clin Ther 2015; 37: 225–241.e8. [DOI] [PubMed] [Google Scholar]
- 15. Robinson LE, Holt TA, Rees K, et al. Effects of exenatide and liraglutide on heart rate, blood pressure and body weight: systematic review and meta-analysis. BMJ Open 2013; 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Sun F, Wu S, Guo S, et al. Impact of GLP-1 receptor agonists on blood pressure, heart rate and hypertension among patients with type 2 diabetes: a systematic review and network meta-analysis. Diabetes Res Clin Pract 2015; 110: 26–37. [DOI] [PubMed] [Google Scholar]
- 17. Marso SP, Daniels GH, Brown-Frandsen K, et al. ; LEADER Steering Committee; LEADER Trial Investigators. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2016; 375: 311–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Marso SP, Bain SC, Consoli A, et al. ; SUSTAIN-6 Investigators. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 2016; 375: 1834–1844. [DOI] [PubMed] [Google Scholar]
- 19. Simó R, Guerci B, Schernthaner G, et al. Long-term changes in cardiovascular risk markers during administration of exenatide twice daily or glimepiride: results from the European exenatide study. Cardiovasc Diabetol 2015; 14: 116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Armstrong MJ, Hull D, Guo K, et al. Glucagon-like peptide 1 decreases lipotoxicity in non-alcoholic steatohepatitis. J Hepatol 2016; 64: 399–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Blackman A, Foster GD, Zammit G, et al. Effect of liraglutide 3.0 mg in individuals with obesity and moderate or severe obstructive sleep apnea: the SCALE Sleep Apnea randomized clinical trial. Int J Obes (Lond) 2016; 40: 1310–1319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Zhao TC. Glucagon-like peptide-1 (GLP-1) and protective effects in cardiovascular disease: a new therapeutic approach for myocardial protection. Cardiovasc Diabetol 2013; 12: 90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Katout M, Zhu H, Rutsky J, et al. Effect of GLP-1 mimetics on blood pressure and relationship to weight loss and glycemia lowering: results of a systematic meta-analysis and meta-regression. Am J Hypertens 2014; 27: 130–139. [DOI] [PubMed] [Google Scholar]
- 24. Tanaka T, Nangaku M, Nishiyama A. The role of incretins in salt-sensitive hypertension: the potential use of dipeptidyl peptidase-IV inhibitors. Curr Opin Nephrol Hypertens 2011; 20: 476–481. [DOI] [PubMed] [Google Scholar]
- 25. Liu L, Liu J, Wong WT, et al. Dipeptidyl peptidase 4 inhibitor sitagliptin protects endothelial function in hypertension through a glucagon-like peptide 1-dependent mechanism. Hypertension 2012; 60: 833–841. [DOI] [PubMed] [Google Scholar]
- 26. Carbone LJ, Angus PW, Yeomans ND. Incretin-based therapies for the treatment of non-alcoholic fatty liver disease: a systematic review and meta-analysis. J Gastroenterol Hepatol 2016; 31: 23–31. [DOI] [PubMed] [Google Scholar]
- 27. Klonoff DC, Buse JB, Nielsen LL, et al. Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr Med Res Opin 2008; 24: 275–286. [DOI] [PubMed] [Google Scholar]
- 28. Aviles-Olmos I, Dickson J, Kefalopoulou Z, et al. Motor and cognitive advantages persist 12 months after Exenatide exposure in Parkinson’s disease. J Parkinsons Dis 2014; 4: 337–344. [DOI] [PubMed] [Google Scholar]
- 29. Aviles-Olmos I, Dickson J, Kefalopoulou Z, et al. Exenatide and the treatment of patients with Parkinson’s disease. J Clin Invest 2013; 123, 2730–2736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Athauda D, Maclagan K, Skene SS, et al. Exenatide once weekly versus placebo in Parkinson’s disease: a randomised, double-blind, placebo-controlled trial. Lancet 2017; 390: 1664–1675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Athauda D, Foltynie T. Protective effects of the GLP-1 mimetic exendin-4 in Parkinson’s disease. Neuropharmacology 2018; 136: 260–270. [DOI] [PubMed] [Google Scholar]
- 32. Woert MH, Mueller PS. Glucose, insulin, and free fatty acid metabolism in Parkinson’s disease treated with levodopa. Clin Pharmacol Ther 1971; 12: 360–367. [DOI] [PubMed] [Google Scholar]
- 33. Li Y, Li L, Hölscher C. Incretin-based therapy for type 2 diabetes mellitus is promising for treating neurodegenerative diseases. Rev Neurosci 2016; 27: 689–711. [DOI] [PubMed] [Google Scholar]
- 34. Li J, Shao YH, Gong YP, et al. Diabetes mellitus and dementia - a systematic review and meta-analysis. Eur Rev Med Pharmacol Sci 2014; 18: 1778–1789. [PubMed] [Google Scholar]
- 35. Gejl M, Gjedde A, Egefjord L, et al. In Alzheimer’s disease, 6-month treatment with GLP-1 analog prevents decline of brain glucose metabolism: randomized, placebo-controlled, double-blind clinical trial. Front Aging Neurosci 2016; 8: 108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Craft S, Baker LD, Montine TJ, et al. Intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment: a pilot clinical trial. Arch Neurol 2012; 69: 29–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Lerche S, Brock B, Rungby J, et al. Glucagon-like peptide-1 inhibits blood-brain glucose transfer in humans. Diabetes 2008; 57: 325–331. [DOI] [PubMed] [Google Scholar]
- 38. Watson GS, Cholerton BA, Reger MA, et al. Preserved cognition in patients with early Alzheimer disease and amnestic mild cognitive impairment during treatment with rosiglitazone: a preliminary study. Am J Geriatr Psychiatry 2005; 13: 950–958. [DOI] [PubMed] [Google Scholar]
- 39. Bae CS, Song J. The role of Glucagon-Like Peptide 1 (GLP1) in Type 3 Diabetes: GLP-1 controls insulin resistance, neuroinflammation and neurogenesis in the brain. Int J Mol Sci 2017; 18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Bak AM, Egefjord L, Gejl M, et al. Targeting amyloid-beta by glucagon-like peptide −1 (GLP-1) in Alzheimer’s disease and diabetes. Expert Opin Ther Targets 2011; 15: 1153–1162. [DOI] [PubMed] [Google Scholar]
- 41. Mansur RB, Ahmed J, Cha DS, et al. Liraglutide promotes improvements in objective measures of cognitive dysfunction in individuals with mood disorders: a pilot, open-label study. J Affect Disord 2017; 207: 114–120. [DOI] [PubMed] [Google Scholar]
- 42. Grant P, Lipscomb D, Quin J. Psychological and quality of life changes in patients using GLP-1 analogues. J Diabetes Complications 2011; 25: 244–246. [DOI] [PubMed] [Google Scholar]
- 43. Amin RS, Naong Simakajornboon N, Szczesniak RV. Treatment of obstructive sleep apnea with glucagon like peptide-1 receptor agonist. Am J Respiratory Critical Care Medicine 2015; 191: A4144. [Google Scholar]
- 44. Matsumoto T, Harada N, Azuma M, et al. Plasma incretin levels and dipeptidyl peptidase-4 activity in patients with obstructive sleep apnea. Ann Am Thorac Soc 2016; 13: 1378–1387. [DOI] [PubMed] [Google Scholar]
- 45. Reutrakul S, Sumritsopak R, Saetung S, et al. The relationship between sleep and glucagon-like peptide 1 in patients with abnormal glucose tolerance. J Sleep Res 2017; 26: 756–763. [DOI] [PubMed] [Google Scholar]
- 46. Su B, Sheng H, Zhang M, et al. Risk of bone fractures associated with glucagon-like peptide-1 receptor agonists’ treatment: a meta-analysis of randomized controlled trials. Endocrine 2015; 48: 107–115. [DOI] [PubMed] [Google Scholar]
- 47. Mabilleau G, Mieczkowska A, Chappard D. Use of glucagon-like peptide-1 receptor agonists and bone fractures: a meta-analysis of randomized clinical trials. J Diabetes 2014; 6: 260–266. [DOI] [PubMed] [Google Scholar]
- 48. Li R, Xu W, Luo S, et al. Effect of exenatide, insulin and pioglitazone on bone metabolism in patients with newly diagnosed type 2 diabetes. Acta Diabetol 2015; 52: 1083–1091. [DOI] [PubMed] [Google Scholar]
- 49. Iepsen EW, Lundgren JR, Hartmann B, et al. GLP-1 receptor agonist treatment increases bone formation and prevents bone loss in weight-reduced obese women. J Clin Endocrinol Metab 2015; 100: 2909–2917. [DOI] [PubMed] [Google Scholar]
- 50. Driessen JH, Henry RM, van Onzenoort HA, et al. Bone fracture risk is not associated with the use of glucagon-like peptide-1 receptor agonists: a population-based cohort analysis. Calcif Tissue Int 2015; 97: 104–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Driessen JH, van Onzenoort HA, Starup-Linde J, et al. Use of glucagon-like-peptide 1 receptor agonists and risk of fracture as compared to use of other anti-hyperglycemic drugs. Calcif Tissue Int 2015; 97: 506–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Faurschou A, Knop FK, Thyssen JP, et al. Improvement in psoriasis after treatment with the glucagon-like peptide-1 receptor agonist liraglutide. Acta Diabetol 2014; 51: 147–150. [DOI] [PubMed] [Google Scholar]
- 53. Hogan AE, Tobin AM, Ahern T, et al. Glucagon-like peptide-1 (GLP-1) and the regulation of human invariant natural killer T cells: lessons from obesity, diabetes and psoriasis. Diabetologia 2011; 54: 2745–2754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Oxford Centre for Evidence-Based Medicine. OCEBM Levels of Evidence: Levels of Evidence for Therapy/Prevention/Etiology/Harm, www.cebm.net/?o=1025. (2009, accessed 28 March 2018).
- 55. Prasad-Reddy L, Isaacs D. A clinical review of GLP-1 receptor agonists: efficacy and safety in diabetes and beyond. Drugs Context 2015; 4: 212283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Tran S, Retnakaran R, Zinman B, et al. Efficacy of glucagon-like peptide-1 receptor agonists compared to dipeptidyl peptidase-4 inhibitors for the management of type 2 diabetes: a meta-analysis of randomized clinical trials. Diabetes Obes Metab 2018; 20(Suppl. 1): 68–76. [DOI] [PubMed] [Google Scholar]
- 57. Kaushik V, Smith ST, Mikobi E, et al. Acetylcholinesterase inhibitors: beneficial effects on comorbidities in patients with Alzheimer’s disease. Am J Alzheimers Dis Other Demen 2018; 33: 73–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Raji MA, Kuo YF, Snih SA, et al. Ethnic differences in herb and vitamin/mineral use in the elderly. Ann Pharmacother 2005; 39: 1019–1023. [DOI] [PubMed] [Google Scholar]
- 59. Pfeffer MA, Claggett B, Diaz R, et al. ; ELIXA Investigators. Lixisenatide in patients with type 2 diabetes and acute coronary syndrome. N Engl J Med 2015; 373: 2247–2257. [DOI] [PubMed] [Google Scholar]
- 60. Holman RR, Bethel MA, Mentz RJ, et al. ; EXSCEL Study Group. Effects of once-weekly exenatide on cardiovascular outcomes in type 2 diabetes. N Engl J Med 2017; 377: 1228–1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Kaul S. Mitigating cardiovascular risk in type 2 diabetes with antidiabetes drugs: a review of principal cardiovascular outcome results of EMPA-REG OUTCOME, LEADER, and SUSTAIN-6 trials. Diabetes Care 2017; 40: 821–831. [DOI] [PubMed] [Google Scholar]
- 62. Bahtiyar G, Pujals-Kury J, Sacerdote A. Cardiovascular effects of different GLP-1 receptor agonists in patients with type 2 diabetes. Curr Diab Rep 2018; 18: 92. [DOI] [PubMed] [Google Scholar]
- 63. Bain SC, Mosenzon O, Arechavaleta R, et al. Cardiovascular safety of oral semaglutide in patients with type 2 diabetes: Rationale, design and patient baseline characteristics for the PIONEER 6 trial. Diabetes Obes Metab 2019; 21: 499–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Rinella ME. Nonalcoholic fatty liver disease: a systematic review. JAMA 2015; 313: 2263–2273. [DOI] [PubMed] [Google Scholar]
- 65. Ding X, Saxena NK, Lin S, et al. Exendin-4, a glucagon-like protein-1 (GLP-1) receptor agonist, reverses hepatic steatosis in ob/ob mice. Hepatology 2006; 43: 173–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Mells JE, Fu PP, Sharma S, et al. Glp-1 analog, liraglutide, ameliorates hepatic steatosis and cardiac hypertrophy in C57BL/6J mice fed a Western diet. Am J Physiol Gastrointest Liver Physiol 2012; 302: G225–G235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Hamilton A, Hölscher C. Receptors for the incretin glucagon-like peptide-1 are expressed on neurons in the central nervous system. Neuroreport 2009; 20: 1161–1166. [DOI] [PubMed] [Google Scholar]
- 68. Hölscher C. Drugs developed for treatment of diabetes show protective effects in Alzheimer’s and Parkinson’s diseases. Acta Physiologica Sinica 2014; 66: 497–510. [PubMed] [Google Scholar]
- 69. Duffy AM, Hölscher C. The incretin analogue D-Ala2GIP reduces plaque load, astrogliosis and oxidative stress in an APP/PS1 mouse model of Alzheimer’s disease. Neuroscience 2013; 228: 294–300. [DOI] [PubMed] [Google Scholar]
- 70. Perry T, Haughey NJ, Mattson MP, et al. Protection and reversal of excitotoxic neuronal damage by glucagon-like peptide-1 and exendin–4. J Pharmacol Exp Ther 2002; 302: 881–888. [DOI] [PubMed] [Google Scholar]
- 71. Han WN, Hölscher C, Yuan L, et al. Liraglutide protects against amyloid-β protein-induced impairment of spatial learning and memory in rats. Neurobiol Aging 2013; 34: 576–588. [DOI] [PubMed] [Google Scholar]
- 72. Cai HY, Hölscher C, Yue XH, et al. Lixisenatide rescues spatial memory and synaptic plasticity from amyloid beta protein-induced impairments in rats. Neuroscience 2014; 277: 6–13. [DOI] [PubMed] [Google Scholar]
- 73. Li Y, Bader M, Tamargo I, et al. Liraglutide is neurotrophic and neuroprotective in neuronal cultures and mitigates mild traumatic brain injury in mice. J Neurochem 2015; 135: 1203–1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Sharma M, Jalewa J, Hölscher C. Neuroprotective and antiapoptotic effects of liraglutide on SH-SY5Y cells exposed to methylglyoxal stress. J Neurochem 2014; 128: 459–471. [DOI] [PubMed] [Google Scholar]
- 75. Gault V, Hölscher C. GLP-1 agonists facilitate hippocampal LTP and reverse the impairment of LTP induced by beta-amyloid. Eur J Pharmacol 2008; 587: 112–117. [DOI] [PubMed] [Google Scholar]
- 76. McClean PL, Gault VA, Harriott P, et al. Glucagon-like peptide-1 analogues enhance synaptic plasticity in the brain: a link between diabetes and Alzheimer’s disease. Eur J Pharmacol 2010; 630: 158–162. [DOI] [PubMed] [Google Scholar]
- 77. McClean P, Parthsarathy V, Faivre E, et al. The diabetes drug liraglutide prevents degenerative processes in a mouse model of Alzheimer’s disease. J Neurosci 2011; 31: 6587–6594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Harkavyi A, Abuirmeileh A, Lever R, et al. Glucagon-like peptide 1 receptor stimulation reverses key deficits in distinct rodent models of Parkinson’s disease. J Neuroinflammation 2008; 5: 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. McClean PL, Hölscher C. Liraglutide can reverse memory impairment, synaptic loss and reduce plaque load in aged APP/PS1 mice, a model of Alzheimer’s disease. Neuropharmacology 2014; 76: 57–67. [DOI] [PubMed] [Google Scholar]
- 80. Aksoy D, Solmaz V, Çavuşoğlu T, et al. Neuroprotective effects of exenatide in a rotenone-induced rat model of Parkinson’s disease. Am J Med Sci 2017; 354: 319–324. [DOI] [PubMed] [Google Scholar]
- 81. Kastin AJ, Akerstrom V. Entry of exendin-4 into brain is rapid but may be limited at high doses. Int J Obes Relat Metab Disord 2003; 27: 313–318. [DOI] [PubMed] [Google Scholar]
- 82. Li Y, Perry T, Kindy MS, et al. GLP-1 receptor stimulation preserves primary cortical and dopaminergic neurons in cellular and rodent models of stroke and Parkinsonism. Proc Natl Acad Sci U S A 2009; 106: 1285–1290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Bertilsson G, Patrone C, Zachrisson O, et al. Peptide hormone exendin-4 stimulates subventricular zone neurogenesis in the adult rodent brain and induces recovery in an animal model of Parkinson’s disease. J Neurosci Res 2008; 86: 326–338. [DOI] [PubMed] [Google Scholar]
- 84. Kim S, Moon M, Park S. Exendin-4 protects dopaminergic neurons by inhibition of microglial activation and matrix metalloproteinase-3 expression in an animal model of Parkinson’s disease. J Endocrinol 2009; 202: 431–439. [DOI] [PubMed] [Google Scholar]
- 85. Liu W, Jalewa J, Sharma M, et al. Neuroprotective effects of lixisenatide and liraglutide in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson’s disease. Neuroscience 2015; 303: 42–50. [DOI] [PubMed] [Google Scholar]
- 86. Athauda D, Wyse R, Brundin P, et al. Is exenatide a treatment for Parkinson’s Disease? J Parkinsons Dis 2017; 7: 451–458. [DOI] [PubMed] [Google Scholar]
- 87. Imperial College London. Evaluating liraglutide in Alzheimer’s disease (ELAD) [ClinicalTrials.gov identifier NCT01843075]. US National Institutes of Health, ClinicalTrials.gov, https://clinicaltrials.gov/ct2/show/NCT01843075, (2013, accessed 5 April 2016). [Google Scholar]
- 88. Arlt S. Non-Alzheimer’s disease-related memory impairment and dementia. Dialogues Clin Neurosci 2013; 15: 465–473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Greig NH, Mattson MP, Perry T, et al. New therapeutic strategies and drug candidates for neurodegenerative diseases: p53 and TNF-alpha inhibitors, and GLP-1 receptor agonists. Ann N Y Acad Sci 2004; 1035: 290–315. [DOI] [PubMed] [Google Scholar]
- 90. Calsolaro V, Edison P. Novel GLP-1 (Glucagon-Like Peptide-1) analogues and insulin in the treatment for Alzheimer’s disease and other neurodegenerative diseases. CNS Drugs 2015; 29: 1023–1039. [DOI] [PubMed] [Google Scholar]
- 91. Ji C, Xue GF, Li G, et al. Neuroprotective effects of glucose-dependent insulinotropic polypeptide in Alzheimer’s disease. Rev Neurosci 2016; 27: 61–70. [DOI] [PubMed] [Google Scholar]
- 92. Cork SC, Richards JE, Holt MK, et al. Distribution and characterization of glucagon-like peptide-1 receptor expressing cells in the mouse brain. Mol Metab 2015; 4: 718–731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Abbas T, Faivre E, Holscher C. Impairment of synaptic plasticity and memory formation in GLP-1 receptor KO mice: interaction between type 2 diabetes and Alzheimer’s disease. Behav Brain Res 2009; 205: 265–271. [DOI] [PubMed] [Google Scholar]
- 94. Hamilton A, Patterson S, Porter D, et al. Novel GLP-1 mimetics developed to treat type 2 diabetes promote progenitor cell proliferation in the brain. J Neurosci Res 2011; 89: 481–489. [DOI] [PubMed] [Google Scholar]
- 95. Areosa Sastre A, Vernooij RW, González-Colaço Harmand M, et al. Effect of the treatment of Type 2 diabetes mellitus on the development of cognitive impairment and dementia. Cochrane Database Syst Rev 2017; 6: CD003804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Valencia WM, Botros D, Vera-Nunez M, et al. Diabetes treatment in the elderly: incorporating geriatrics, technology, and functional medicine. Curr Diab Rep 2018; 18: 95. [DOI] [PubMed] [Google Scholar]
- 97. Anderberg RH, Richard JE, Hansson C, et al. GLP-1 is both anxiogenic and antidepressant; divergent effects of acute and chronic GLP-1 on emotionality. Psychoneuroendocrinology 2016; 65: 54–66. [DOI] [PubMed] [Google Scholar]
- 98. McIntyre RS, Powell AM, Kaidanovich-Beilin O, et al. The neuroprotective effects of GLP-1: possible treatments for cognitive deficits in individuals with mood disorders. Behav Brain Res 2013; 237: 164–171. [DOI] [PubMed] [Google Scholar]
- 99. Billings LK, Handelsman Y, Heile M, et al. Health-related quality of life assessments with once-weekly glucagon-like peptide-1 receptor agonists in type 2 diabetes mellitus. J Manag Care Spec Pharm 2018; 24 (9-a Suppl.): S30–S41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Sharma AN, Ligade SS, Sharma JN, et al. GLP-1 receptor agonist liraglutide reverses long-term atypical antipsychotic treatment associated behavioral depression and metabolic abnormalities in rats. Metab Brain Dis 2015; 30: 519–527. [DOI] [PubMed] [Google Scholar]
- 101. Ishøy PL, Fagerlund B, Broberg BV, et al. No cognitive-enhancing effect of GLP-1 receptor agonism in antipsychotic-treated, obese patients with schizophrenia. Acta Psychiatr Scand 2017; 136: 52–62. [DOI] [PubMed] [Google Scholar]
- 102. Gong N, Xiao Q, Zhu B, et al. Activation of spinal glucagon-like peptide-1 receptors specifically suppresses pain hypersensitivity. J Neurosci 2014; 34: 5322–5334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Fan H, Gong N, Li TF, et al. The non-peptide GLP-1 receptor agonist WB4–24 blocks inflammatory nociception by stimulating β-endorphin release from spinal microglia. Br J Pharmacol 2015; 172: 64–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Yamada C, Yamada Y, Tsukiyama K, et al. The murine glucagon-like peptide-1 receptor is essential for control of bone resorption. Endocrinology 2008; 149: 574–579. [DOI] [PubMed] [Google Scholar]
- 105. Vianna AGD, Sanches CP, Barreto FC. Review article: effects of type 2 diabetes therapies on bone metabolism. Diabetol Metab Syndr 2017; 9: 75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Mabilleau G, Pereira M, Chenu C. Novel skeletal effects of glucagon-like peptide-1 (GLP-1) receptor agonists. J Endocrinol 2018; 236: R29–R42. [DOI] [PubMed] [Google Scholar]
- 107. Ma X, Meng J, Jia M, et al. Exendin-4, a glucagon-like peptide-1 receptor agonist, prevents osteopenia by promoting bone formation and suppressing bone resorption in aged ovariectomized rats. J Bone Miner Res 2013; 28: 1641–1652. [DOI] [PubMed] [Google Scholar]
- 108. Gier B, Butler PC, Lai CK, et al. Glucagon like peptide-1 receptor expression in the human thyroid gland. J Clin Endocrinol Metab 2012; 97: 121–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Bjerre Knudsen L, Madsen LW, Andersen S, et al. Glucagon-like peptide-1 receptor agonists activate rodent thyroid C-cells causing calcitonin release and C-cell proliferation. Endocrinology 2010; 151: 1473–1486. [DOI] [PubMed] [Google Scholar]
- 110. Zhang YS, Weng WY, Xie BC, et al. Glucagon-like peptide-1 receptor agonists and fracture risk: a network meta-analysis of randomized clinical trials. Osteoporos Int 2018; 29: 2639–2644. [DOI] [PubMed] [Google Scholar]
- 111. Nishioka T, Tanimoto N, Kumagai C, et al. Sitagliptin, a dipeptidyl peptidase-IV inhibitor, improves psoriasis. Dermatology 2012; 224: 20–21. [DOI] [PubMed] [Google Scholar]
- 112. Lee YS, Jun HS. Anti-inflammatory effects of GLP-1-based therapies beyond glucose control. Mediators Inflamm 2016; 2016: 3094642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Filippatos TD, Panagiotopoulou TV, Elisaf MS. Adverse effects of GLP-1 receptor agonists. Rev Diabet Stud 2014; 11: 202–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Sun F, Chai S, Yu K, et al. Gastrointestinal adverse events of glucagon-like peptide-1 receptor agonists in patients with type 2 diabetes: a systematic review and network meta-analysis. Diabetes Technol Ther 2015; 17: 35–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Buse JB, Rosenstock J, Sesti G, et al. ; LEAD-6 Study Group. Liraglutide once a day versus exenatide twice a day for type 2 diabetes: a 26-week randomised, parallel-group, multinational, open-label trial (LEAD-6). Lancet 2009; 374: 39–47. [DOI] [PubMed] [Google Scholar]
- 116. Bettge K, Kahle M, Abd El, Aziz MS, et al. 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 2017; 19: 336–347. [DOI] [PubMed] [Google Scholar]
- 117. Mishriky BM, Cummings DM, Powell JR, et al. Comparing once-weekly semaglutide to incretin-based therapies in patients with type 2 diabetes: a systematic review and meta-analysis. Diabetes Metab 2019; 45: 102–109. [DOI] [PubMed] [Google Scholar]
- 118. Li X, Qie S, Wang X, et al. The safety and efficacy of once-weekly glucagon-like peptide-1 receptor agonist semaglutide in patients with type 2 diabetes mellitus: a systemic review and meta-analysis. Endocrine 2018; 62: 535–545. [DOI] [PubMed] [Google Scholar]
- 119. Coon SC, Crannage KLC, Guyton JE. Semaglutide once-weekly: improved efficacy with a new safety warning. Expert Rev Clin Pharmacol 2018; 11: 1061–1072. [DOI] [PubMed] [Google Scholar]
- 120. Rodbard HW, Lingvay I, Reed J, et al. Semaglutide added to basal insulin in type 2 diabetes (SUSTAIN 5): a randomized, controlled trial. J Clin Endocrinol Metab 2018; 103: 2291–2301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Goldenberg RM, Steen O. Semaglutide: review and place in therapy for adults with type 2 diabetes. Can J Diabetes 2019; 43(2): 136–145. [DOI] [PubMed] [Google Scholar]
- 122. Mann JFE, Ørsted DD, Brown-Frandsen K, et al. ; LEADER Steering Committee and Investigators. Liraglutide and renal outcomes in type 2 diabetes. N Engl J Med 2017; 377: 839–848. [DOI] [PubMed] [Google Scholar]
- 123. Tuttle KR, Lakshmanan MC, Rayner B, et al. Dulaglutide versus insulin glargine in patients with type 2 diabetes and moderate-to-severe chronic kidney disease (AWARD-7): a multicentre, open-label, randomised trial. Lancet Diabetes Endocrinol 2018; 6: 605–617. [DOI] [PubMed] [Google Scholar]