Next Article in Journal
Association of the Heart Rate Variability Response to Active Standing with the Severity of Calcific Aortic Valve Disease: Novel Insights of a Neurocardiovascular Pathology
Next Article in Special Issue
Effect of Dipeptidyl Peptidase-4 Inhibitors vs. Metformin on Major Cardiovascular Events Using Spontaneous Reporting System and Real-World Database Study
Previous Article in Journal
Postoperative Outcomes and Reinterventions Following Fenestrated/Branched Endovascular Aortic Repair in Post-Dissection and Complex Degenerative Abdominal and Thoraco-Abdominal Aortic Aneurysms
Previous Article in Special Issue
Novel Method for Early Prediction of Clinically Significant Drug–Drug Interactions with a Machine Learning Algorithm Based on Risk Matrix Analysis in the NICU
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Efficacy and Safety of Empagliflozin in Type 2 Diabetes Mellitus Saudi Patients as Add-On to Antidiabetic Therapy: A Prospective, Open-Label, Observational Study

by
Fahad M. Althobaiti
1,2,3,
Safaa M. Alsanosi
1,4,*,
Alaa H. Falemban
1,3,
Abdullah R. Alzahrani
1,3,
Salma A. Fataha
2,
Sara O. Salih
2,
Ali M. Alrumaih
5,
Khalid N. Alotaibi
5,
Hazim M. Althobaiti
6,
Saeed S. Al-Ghamdi
1,3 and
Nahla Ayoub
1,3,7,*
1
Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University (UQU), Makkah 21955, Saudi Arabia
2
Department of Endocrine and Diabetes, Armed Forces Hospital, Southern Region, Khamis Mushait 61961, Saudi Arabia
3
Saudi Toxicology Society, Umm Al-Qura University (UQU), Makkah 21955, Saudi Arabia
4
Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK
5
Ministry of Defence, Riyadh 11159, Saudi Arabia
6
King Faisal Medical City, Taif 26514, Saudi Arabia
7
Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Cairo 11566, Egypt
*
Authors to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(16), 4769; https://doi.org/10.3390/jcm11164769
Submission received: 4 July 2022 / Revised: 10 August 2022 / Accepted: 12 August 2022 / Published: 16 August 2022

Abstract

:
The Saudi Food and Drug Authority (SFDA) approved sodium-glucose cotransporter-2 (SGLT2) inhibitors in 2018. The efficacy and safety of empagliflozin (EMPA) have been confirmed in the U.S., Europe, and Japan for patients with type 2 diabetes mellitus (T2DM); however, analogous evidence is lacking for Saudi T2DM patients. Therefore, the current study aimed to assess the efficacy and safety of EMPA in Saudi patients (n = 256) with T2DM. This is a 12-week prospective, open-label, observational study. Adult Saudi patients with T2DM who had not been treated with EMPA before enrolment were eligible. The exclusion criteria included T2DM patients less than 18 years of age, adults with type one diabetes, pregnant women, paediatric population. The results related to efficacy included a significant decrease in haemoglobin A1c (HbA1c) (adjusted mean difference −0.93% [95% confidence interval (CI) −0.32, −1.54]), significant improvements in fasting plasma glucose (FPG) (−2.28 mmol/L [95% CI −2.81, −1.75]), and a reduction in body weight (−0.874 kg [95% CI −4.36, −6.10]) following the administration of 25 mg of EMPA once daily as an add-on to ongoing antidiabetic therapy after 12 weeks. The primary safety endpoints were the change in the mean blood pressure (BP) values, which indicated significantly reduced systolic and diastolic BP (−3.85 mmHg [95% CI −6.81, −0.88] and −0.06 mmHg [95% CI −0.81, −0.88], respectively) and pulse rate (−1.18 [95% CI −0.79, −3.15]). In addition, kidney function was improved, with a significant reduction in the urine albumin/creatinine ratio (UACR) (−1.76 mg/g [95% CI −1.07, −34.25]) and a significant increase in the estimated glomerular filtration rate (eGFR) (3.54 mL/min/1.73 m2 [95% CI 2.78, 9.87]). Furthermore, EMPA reduced aminotransferases (ALT) in a pattern (reduction in ALT > AST). The adjusted mean difference in the change in ALT was −2.36 U/L [95% CI −1.031, −3.69], while it was −1.26 U/L [95% CI −0.3811, −2.357] for AST and −1.98 U/L [95% CI −0.44, −3.49] for GGT. Moreover, in the EMPA group, serum high-density lipoprotein (HDL) significantly increased (0.29 mmol/L [95% CI 0.74, 0.15]), whereas a nonsignificant increase was seen in low-density lipoprotein (LDL) (0.01 mmol/L [95% CI 0.19, 0.18]) along with a significant reduction in plasma triglyceride (TG) levels (−0.43 mmol/L [95% CI −0.31, −1.17]). Empagliflozin once daily is an efficacious and tolerable strategy for treating Saudi patients with insufficiently controlled T2DM as an add-on to ongoing antidiabetic therapy.

1. Introduction

The growing burden of type 2 diabetes mellites (T2DM) is a crucial issue in health care worldwide. T2DM continues to increase in prevalence and incidence and is a significant cause of human suffering and death. Despite sizable investments in clinical care, research, and public health interventions, there appears to be no signal of reduction in the rate of disease increase [1]. According to the World Health Organisation (WHO), Saudi Arabia has the second highest diabetes prevalence of all Middle Eastern countries (7th in the world), with an estimated population of seven million individuals living with diabetes and more than three million with prediabetes [2,3,4]. Moreover, by 2030, this number is expected to more than double [5].
EMPA was approved by the U.S. Food and Drug Administration (FDA) in 2014 as an adjunct to diet and exercise to improve glycaemic control in adults with T2DM [6]. EMPA is a selective sodium-glucose cotransporter 2 (SGLT2) inhibitor. It is characterised by its unique mechanism as a hypoglycaemic agent. Specifically, it depends on enhancing glycosuria away from insulin independence. This unique mechanism enables EMPA to achieve controllable hypoglycaemic action [7]. Studies conducted in the USA, Canada, UK, and Japan have recommended using EMPA alone or together with other anti-diabetic agents as a cost-effective oral treatment for T2DM once daily [8]. Many clinical trials have reported the antihyperglycemic effect of EMPA due to a reduction in haemoglobin A1c (HbA1c) and fasting plasma glucose (FPG) levels [9,10,11,12]. In addition, EMPA is reported to reduce body weight, waist circumference, and body fat index in patients with T2DM [13,14].
In 2016, the U.S. FDA approved EMPA to reduce the risk of cardiovascular death in adult patients with T2DM and cardiovascular disease. Many studies have documented the relative reductions in the risk of cardiovascular death and hospitalisation with EMPA versus placebo [7,15,16,17,18]. In addition, EMPA causes significant natriuresis [19], rapid reductions in pulmonary artery pressure, and reduced LV volumes in patients with heart failure and reduced ejection fraction (HFrEF) [20,21,22]. The cellular mechanism by which EMPA improves cardiovascular outcomes is its ability to stimulate erythropoiesis via an early increase in erythropoietin production in people with T2DM [23]. Early administration of EMPA may attenuate changes in extracellular water and intracellular water (ICW) in patients with acute myocardial infarction [24].
EMPA slows the progressive decline in kidney function in patients with HFrEF, with or without diabetes [25,26]. The short- and long-term benefits of EMPA on urinary albumin excretion have also been shown [27]. In addition, the haemodynamic effects of EMPA associated with lower glomerular pressure may contribute to the long-term preservation of renal function. [28]. Furthermore, EMPA improved glycaemic control in renal transplant recipients with post-transplantation diabetes mellitus (PTDM) compared with a placebo [29]. Sattar et al. proved that EMPA reduces liver enzymes ALT and AST in patients with T2DM in a pattern consistent with a reduction in liver fat, particularly when ALT levels are high [30,31,32].
Several studies have reported the safety and efficacy of EMPA for T2DM [11,33,34,35]. EMPA was the first in class to not only demonstrate safe SGLT2 inhibition but also cardio- and reno-protective effects in an adequately powered cardiovascular outcome trial [36]. However, EMPA was associated with an increased risk of hypoglycaemia and genital and urinary tract infections [37].
The Saudi Food and Drug Authority (SFDA) approved SGLT2 inhibitors in 2018 [38]. The current SFDA-approved drugs in this class include canagliflozin, dapagliflozin, and EMPA. Based on the literature, the efficacy and safety of empagliflozin have been confirmed in the U.S., Europe, and Japan for patients with T2DM; however, analogous evidence is lacking for Saudi T2DM patients. Therefore, we assessed the efficacy and safety of empagliflozin as an add-on to ongoing antidiabetic therapy in Saudi adult patients with T2DM in the Armed Forces Hospital, Southern Region between June and December 2021.

2. Materials and Methods

2.1. Study Design and Patients

This was a prospective, open-label, observational clinical study conducted at the Endocrine and Diabetes Centre of the Armed Forces Hospital, Southern Region (Saudi Arabia) between June and December 2021. The study protocol was approved by the Armed Forces Hospital, Southern Region Research Ethics Committee (Number: AFHSRMREC/2021/PHARMACY/503). All participants provided signed and dated informed consent prior to screening.
Saudi adult participants aged ≥18 to <80 years were screened to determine whether they met the inclusion criteria: Adult Saudi patients with T2DM in the Endocrine and Diabetes Centre of the Armed Forces Hospital, Southern Region in Saudi Arabia. Patients who had not been treated with EMPA before enrolment were eligible. The exclusion criteria included T2DM patients less than 18 years of age, adults with type one diabetes, and pregnant women. The study population had no cardiovascular or renal disease.
This study was designed to assess the safety and efficacy of EMPA in Saudi adult patients with T2DM. Efficacy and safety analyses were based on a comparison between variables before treatment (baseline group) and in the patients treated with EMPA (25 mg/once daily) for 12 weeks (EMPA group). Efficacy measurements included changes from baseline in HbA1c levels, fasting plasma glucose (FPG) and body weight at week 12. Safety assessments included changes in BP (SBP and DBP), pulse rate, kidney markers (urine albumin/creatinine ratio (UACR) and estimated glomerular filtration rate (eGFR)), liver markers (AST, ALT, and GGT), and serum lipids (LDL-c, HDL-c, and TG) at week 12 [39].

2.2. Patient Demographics at Baseline

Sample size calculations were based on a previous study of empagliflozin with insulin, which suggested that empagliflozin would result in an HbA1c reduction of ~0.5% versus placebo after 12 weeks of treatment, and a standard deviation (SD) of 1.0% [33]. The sample size was 256 patients (113 males and 234 females) with the following characteristics: mean age 58.9 years; males 58.2 and females 59.4. Of 256 patients, 234 (91%) had been diagnosed with T2DM longer than five years, and 22 (9%) had been diagnosed for one to five years. Participating patients had insufficient glycaemic control at baseline, with HbA1c levels ≥7% in 251 patients (97.6%). Further, 167 patients (65%) had HbA1c levels ≥9 despite receiving insulin (156, 64%) or OHA (93, 36%) (Table 1).

2.3. Data Analysis

SPSS (Version 27.0, IBM, Armonk, NY, USA) was employed for statistical analysis using the t-test for two independent samples to determine the rate of change in the means of the two samples, standard deviations, and the level of confidence, which is estimated at 0.05.
Confidence domain: If the level of significance required by researchers is 5%, then the confidence level should be 95%. Thus, the confidence interval contains the possible values of the statistical parameter, which, when subjected to a statistical test using the same sample, will not be rejected. The level of statistical significance for all samples studied was less than 5%, which means that all rates of change in the mean were within the confidence range.

3. Results

3.1. Efficacy

The primary efficacy endpoints were changes in HbA1c, FPG, and body weight from baseline at week 12 (EMPA group). The mean value of HbA1c decreased from 9.77 ± 1.76 to 8.85 ±4.83 with a change of −0.93 (−0.32, −1.54) at a rate of (−0.106) within the confidence interval estimated CI at 95%. In addition, 181 participants (72%) experienced a reduction in HbA1c ≥ 0.5% from baseline. Further, the number of participants with HbA1c ≥ 9% decreased from 167 (65%) to 77 (30%), corresponding to an increase in the proportion of individuals with HbA1c levels ≥8% to <9%, ≥7% to <8%, and <7.0% of (80 to 82), (4 to 76), and (5 to 21), respectively. For participants on insulin and OHA (metformin) therapies (156, 64%), there was a reduction in mean HbA1c % from 9.94 ± 1.84 to 8.66 ±1.47, a change of −1.28 (−1.03, −1.53) at a rate −0.147, accompanied by a reduction in insulin units from 9.94 ± 1.84 to 8.67 ± 1.47 (change −1.27). Meanwhile, for those on OHA only, the reduction in HBA1c % was from 9.46 ± 1.56 to 8.31 ± 1.26 with a change of −1.11 (−0.799, −1.42).
The mean value of FPG reduced from 11.22 ± 4.79 to 8.95 ±3.37, a change of −2.28 (−2.81, −1.75) at a rate of (−0.25) and a 95% CI. A total of 63 participants had FPG levels <7.8 at baseline increased to 115 at week 12. Individuals with FPG levels from 7.8 to <11.0 (80 to 86), while the number with FPG levels ≥11 decreased from 113 to 55. Mean body weight decreased from 89.99 ± 18.09 to 88.03 ± 18.47 with a change of −0.874 (−4.36, −6.10) at a rate of −0.02 (Table 2 and Table 3 and Figure 1).

3.2. Safety

Safety endpoints included changes in SBP, pulse rate, and kidney and liver function status at week 12. SBP decreased from 142.6 ± 19.45 to 138.8 ± 20.23, a change of −3.85 (−6.81, −0.88) at a rate of −0.03. There was a slight reduction in DBP from 79.6 ± 20.32 to 79.4 ± 21.14, a change of −0.06 (−0.81, −0.88) at a rate of −0.001. Moreover, there was a reduction in pulse rate from 85.98 ± 11.33 to 84.80 ± 13.52, a change of −1.18 (−0.79, −3.15) at a rate of −0.01 (Table 2 and Table 3 and Figure 2).
It is apparent from Table 2 and Table 3 that kidney function status improved, with a reduction in UACR from 20.39 ± 43.72 to 17.12 ± 40.05 at week 12, a change of −1.76 (−1.07, −34.25) at a rate of −0.067. The number of participants with UACR levels ≥300 decreased from 75 to 8, accompanied by an increase in the number of patients with UACR levels of 30 to <300 and <30 (from 51 to 67 and 126 to 177 patients, respectively). Furthermore, estimated eGFR also increased from 51.12 ± 120.45 to 72.51 ± 22.80, a change of 3.54 (2.78, 9.87) at a rate of 0.418. The number of participants with normal eGFR (≥90) increased from 49 to 55, along with a reduction in the number of participants with mild eGFR (60 to < 90; from 120 to 116 patients) and moderate-to-severe eGFR (30 to < 60; from 86 to 84 patients). For one participant with severely decreased eGFR (<30), no change was observed (Figure 3).
The results shown in Table 2 and Table 3 show the rates of change in hepatic function status between the baseline and EMPA groups. AST decreased from 22.92 ± 8.10 to 21.65 ± 6.38, a change of −1.26 (−0.3011, −2.227) at a rate of −0.06. ALT decreased from 25.96 ± 8.09 to 23.91 ± 11.71, a change of −2.36 (−1.03, −3.69) at a rate (−0.029). GGT decreased from 30.29 ± 25.15 to 27.12 ± 18.32, a change of −4.31 (−2.33, −6.28) at a rate of −0.159. Interestingly, lipid profiles improved as well (Table 2 and Table 3). HDL increased significantly from 1.69 ± 3.59 to 1.98 ± 0.22, a change of 0.29 (0.74, 0.15) at a rate of 0.171, while LDL increased non-significantly from 2.543 ± 0.93 to 2.544 ±1.50, a change of 0.005 (0.192, 0.18) at a very small rate of 0.0004. Additionally, TG levels decreased from 2.11 ± 5.90 to 1.66 ± 0.20, a change of −0.43 (−0.31, −1.17) at a rate of −0.271 (Figure 4).

4. Discussion

The present study was designed to determine the efficacy and safety of EMPA for Saudi T2DM patients. In this 12-week study, once-daily add-on administration of EMPA (25 mg) resulted in significant decreases in HbA1c, FPG, and body weight for Saudi participants with insufficiently controlled T2DM on insulin or OHA. The meaningful improvement in HbA1c was seen in the participants with HbA1c ≥ 9.0, representing 65% of individuals at baseline group. Furthermore, after 12 weeks of treatment, a decrease in HbA1c ≥ 0.5% from baseline was recorded in 239 (93%) of the participants receiving 25 mg of EMPA. These findings are in line with those of another study that found that 12-week treatment with EMPA monotherapy resulted in similar reductions in HbA1c, FPG, and body weight compared to a placebo in drug-free patients with T2DM [40]. Reductions in HbA1c, FPG, and body weight were also consistent with those reported from 12-week studies on other SGLT2 inhibitors [33,41,42]. EMPA as an added therapy to insulin resulted in a significant reduction in insulin units, consistent with the results reported by [43]. Control of blood glucose levels is important in diabetic patients but often associated with weight gain [44]. The potential for a reduction in body weight is a notable feature of SGLT2 inhibitors [45,46] and may make them useful agents to combine with other antidiabetic therapies to reduce glucose levels and facilitate weight loss or mitigate any weight gain associated with improved glycaemic control [13]. Caloric loss through urinary glucose excretion may be an important contributor to this effect [22].
The reduction in BP observed in the current study is consistent with a reduction in SBP reported with a 25 mg dose of EMPA [47]. It is conceivable that EMPA stimulates osmotic diuresis through increased glycosuria rather than natriuresis, which may play a role in the potential antihypertensive effects of EMPA [48] and support its mechanism as an inhibitor of the renin–angiotensin system [49]. In addition, in Black individuals with T2DM, EMPA reduced BP although the full antihypertensive effect took ≥ 6 months to be fully realised [50]. Moreover, the cardioprotective mechanism of SGLT2 is reported [51]. The hypotheses on SGLT2 mechanisms of action have changed: from simple glycosuric drugs, with consequent glucose lowering, erythropoiesis enhancing, and ketogenesis stimulating, to intracellular sodium-lowering molecules. This provides their consequent cardioprotective effect, which justifies its significant reduction in CV events, especially in populations at higher risk [51]. Hyperglycaemia is a well-established cause of endothelial dysfunction (ED) in the pathophysiology of diabetic complications This abnormal vascular phenotype represents an important risk factor for the genesis of any complication of diabetes, contributing to the pathogenesis of not only macrovascular disease but also microvascular damage. Gliflozins have cardiovascular protective mechanisms of SGLT2 inhibition in patients T2DM and their impact on endothelial function [52].
The present study demonstrated an improvement in kidney function, reduced UCAR, and improved eGFR. Diabetes-associated kidney disease is the most common cause of end-stage renal disease in most countries. Diabetic kidney disease, which develops in approximately 40% of patients with T2DM, further increases the risk of cardio-vascular-related morbidity and mortality [53]. Reported kidney protection with EMPA supports our results [25,54]. It has also been shown that EMPA has a beneficial effect on key efficacy outcomes and slows the rate of kidney function decline in patients with and without chronic kidney disease CKD, regardless of the severity of kidney impairment at baseline [25]. The renal-protective effects of EMPA are likely due to a combination of several mechanisms, including EMPA-associated body weight and BP reductions, diuresis, a shift in substrate utilisation, and activation of tubuloglomerular feedback [53]. In addition, haemodynamic effects of EMPA, associated with a reduction in intraglomerular pressure, may contribute to long-term preservation of kidney function [28]. The fact that SGLT2 inhibition is also associated with small decreases in eGFR over the first 3–4 weeks’ treatment suggests that reductions in intraglomerular pressure associated with EMPA may further contribute to the UCAR-lowering effects [55].
Mechanistic insights suggest that ectopic liver fat is probably part of the pathogenic process in diabetes, contributing to hepatic insulin resistance, excess gluconeogenesis, and higher fasting glucose levels [56]. Furthermore, hepatic steatosis due to non-alcoholic fatty liver disease (NAFLD) leads to and is often clinically suspected by increased levels of aminotransferases, with levels of alanine aminotransferase (ALT) exceeding those of aspartate aminotransferase (AST). Elevated ALT levels (typically >40–50 U/l) are common in individuals with type 2 diabetes and for a given serum ALT, and those with type 2 diabetes have more liver fat compared with BMI-, age-, and sex-matched individuals without diabetes [30]. The mechanisms by which empagliflozin might reduce aminotransferases or liver fat are unclear. Data from animal models support a direct effect of empagliflozin on reducing liver fat and improving hepatic glucose handling. In db/db mice, glucose uptake in the liver and kidneys has been reported to be higher in mice treated with empagliflozin than in controls [57]
In this study, EMPA was found to elicit reductions in aminotransferases ALT, AST, and GGT in Saudi patients with T2DM, and the reduction in ALT was greater than the reduction in AST. These results match those observed in an earlier study [58]. This pattern is consistent with a reduction in liver fat, especially when ALT levels are high [30].
One of the most important clinically relevant findings of the current study was the reduction in plasma TG levels and the increase in HDL-c but not LDL-c levels. These findings are in agreement with [58]. The mechanism behind this finding was also reported. Specifically, EMPA increases serum campesterol, a marker of cholesterol absorption, in patients with T2DM. This increase may be associated with SGLT2 inhibitor-induced increases in HDL cholesterol [59]. Further research is needed to investigate the adverse effects of EMPA on Saudi population with T2DM, which have not yet been documented.

5. Conclusions

This is the first study reporting the efficacy and safety of EMPA as an add-on to antidiabetic therapy (insulin or oral hypoglycaemic agent) in Saudi patients with T2DM.

Author Contributions

Conceptualization, F.M.A. and N.A.; methodology, S.A.F., A.M.A., K.N.A. and H.M.A.; software, F.M.A. and N.A.; validation, A.R.A., S.S.A.-G. and N.A.; formal analysis, S.M.A. and S.O.S.; investigation, A.H.F.; resources, F.M.A.; data curation, F.M.A. and N.A.; writing—original draft preparation, F.M.A., S.M.A. and A.H.F.; writing—review and editing, N.A. and S.M.A.; visualization, A.R.A.; supervision, S.S.A.-G.; project administration, N.A.; funding acquisition, N.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by Ethics Committee of the Armed Forces Hospital, Southern Region Research (protocol code AFHSRMREC/2021/PHARMACY/503, 30 June 2021).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data that support the findings of this study are openly available from the corresponding author upon reasonable request.

Acknowledgments

The authors would like to thank the Deanship of Scientific Research at Umm Al-Qura University for supporting this work by Grant Code: (22UQU4331100DSR01).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Khan, M.A.B.; Hashim, M.J.; King, J.K.; Govender, R.D.; Mustafa, H.; Al Kaabi, J. Epidemiology of Type 2 Diabetes-Global Burden of Disease and Forecasted Trends. J. Epidemiol. Glob. Health 2020, 10, 107–111. [Google Scholar] [CrossRef]
  2. Meo, S.A. Prevalence and future prediction of type 2 diabetes mellitus in the Kingdom of Saudi Arabia: A systematic review of published studies. JPMA. J. Pak. Med. Assoc. 2017, 66, 722–725. [Google Scholar]
  3. Al Dawish, A.A.; Robert, A.; Braham, M.; Musallam, R.A.; Al Hayek, M.A.; Nasser, A.H. Type 2 Diabetes Mellitus in Saudi Arabia: Major Challenges and Possible Solutions. Curr. Diabetes Rev. 2017, 133, 59–64. [Google Scholar]
  4. Alotaibi, A.; Perry, L.; Gholizadeh, L.; Al-Ganmi, A. Incidence and prevalence rates of diabetes mellitus in Saudi Arabia: An overview. J. Epidemiol. Glob. Health 2017, 7, 211–218. [Google Scholar] [CrossRef]
  5. Robert, A.A.; Al Awad, A.D.; Al Dawish, M.A. Current Status of Knowledge and Awareness of Diabetes Mellitus in Saudi Arabia. Curr. Diabetes Rev. 2021, 15, e101220186818. [Google Scholar] [CrossRef]
  6. FDA. US Food and Drug Adminstration: FDA Approves Jardiance (empagliflozin) to Treat Type 2 Diabetes. Available online: https://www.fda-approves-jardiance-empagliflozin-type-2-diabetes-4064.html (accessed on 1 August 2014).
  7. Pan, D.; Xu, L.; Chen, P.; Jiang, H.; Shi, D.; Guo, M. Empagliflozin in Patients With Heart Failure: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Front. Cardiovasc. Med. 2021, 8, 683281. [Google Scholar] [CrossRef]
  8. Frampton, J.E. Empagliflozin: A Review in Type 2 Diabetes. Drugs 2018, 78, 1037–1048. [Google Scholar] [CrossRef]
  9. Ridgefield, C. Jardiance (Empagliflozin) Tablets [Prescribing Information]; Boehringer Ingelheim Pharmaceuticals: Ingelheim am Rhein, Germany, 2014. [Google Scholar]
  10. Fala, L. Jardiance (Empagliflozin), an SGLT2 Inhibitor, Receives FDA Approval for the Treatment of Patients with Type 2 Diabetes. Am. Health Drug Benefits 2015, 8, 92–95. [Google Scholar]
  11. Patorno, E.; Pawar, A.; Wexler, J.; Glynn, J.; Bessette, G.; Paik, J.M.; Najafzadeh, M.; Brodovicz, K.G.; Deruaz-Luyet, A.; Schneeweiss, S. Effectiveness and safety of empagliflozin in routine care patients: Results from the EMPagliflozin compaRative effectIveness and SafEty (EMPRISE) study. Diabetes Obes. Metab. 2021, 24, 442–454. [Google Scholar] [CrossRef]
  12. Liakos, A.; Karagiannis, T.; Athanasiadou, E.; Sarigianni, M.; Mainou, M.; Papatheodorou, K.; Bekiari, E.; Tsapas, A. Efficacy and safety of empagliflozin for type 2 diabetes: A systematic review and meta-analysis. Diabetes Obes. Metab. 2014, 16, 984–993. [Google Scholar] [CrossRef]
  13. Neeland, I.J.; McGuire, D.K.; Chilton, R.; Crowe, S.; Lund, S.S.; Woerle, H.J.; Broedl, U.C.; Johansen, O.E. Empagliflozin reduces body weight and indices of adipose distribution in patients with type 2 diabetes mellitus. Diab. Vasc. Dis. Res. 2016, 13, 119–126. [Google Scholar] [CrossRef]
  14. Cherney, D.Z.I.; Cooper, M.E.; Tikkanen, I.; Pfarr, E.; Johansen, O.E.; Woerle, H.J.; Broedl, U.C.; Lund, S.S. Pooled analysis of Phase III trials indicate contrasting influences of renal function on blood pressure, body weight, and HbA1c reductions with empagliflozin. Kidney Int. 2018, 93, 231–244. [Google Scholar] [CrossRef]
  15. Fitchett, D.; Inzucchi, S.E.; Cannon, C.P.; McGuire, D.K.; Scirica, B.M.; Johansen, O.E.; Sambevski, S.; Kaspers, S.; Pfarr, E.; George, J.T.; et al. Empagliflozin Reduced Mortality and Hospitalization for Heart Failure Across the Spectrum of Cardiovascular Risk in the EMPA-REG OUTCOME Trial. Circulation 2019, 139, 1384–1395. [Google Scholar] [CrossRef]
  16. Anker, S.D.; Butler, J.; Filippatos, G.; Khan, M.S.; Marx, N.; Lam, C.S.P.; Schnaidt, S.; Ofstad, A.P.; Brueckmann, M.; Jamal, W.; et al. Effect of Empagliflozin on Cardiovascular and Renal Outcomes in Patients With Heart Failure by Baseline Diabetes Status: Results From the EMPEROR-Reduced Trial. Circulation 2021, 143, 337–349. [Google Scholar] [CrossRef]
  17. Packer, M.; Anker, S.D.; Butler, J.; Filippatos, G.; Pocock, S.J.; Carson, P.; Januzzi, J.; Verma, S.; Tsutsui, H.; Brueckmann, M.; et al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N. Engl. J. Med. 2020, 383, 1413–1424. [Google Scholar] [CrossRef]
  18. Patorno, E.; Pawar, A.; Franklin, J.M.; Najafzadeh, M.; Deruaz-Luyet, A.; Brodovicz, K.G.; Sambevski, S.; Bessette, L.G.; Santiago Ortiz, A.J.; Kulldorff, M.; et al. Empagliflozin and the Risk of Heart Failure Hospitalization in Routine Clinical Care. Circulation 2019, 139, 2822–2830. [Google Scholar] [CrossRef]
  19. Griffin, M.; Rao, V.S.; Ivey-Miranda, J.; Fleming, J.; Mahoney, D.; Maulion, C.; Suda, N.; Siwakoti, K.; Ahmad, T.; Jacoby, D.; et al. Empagliflozin in Heart Failure: Diuretic and Cardiorenal Effects. Circulation 2020, 142, 1028–1039. [Google Scholar] [CrossRef]
  20. Lee, M.M.Y.; Brooksbank, K.J.M.; Wetherall, K.; Mangion, K.; Roditi, G.; Campbell, R.T.; Berry, C.; Chong, V.; Coyle, L.; Docherty, K.F.; et al. Effect of Empagliflozin on Left Ventricular Volumes in Patients With Type 2 Diabetes, or Prediabetes, and Heart Failure With Reduced Ejection Fraction (SUGAR-DM-HF). Circulation 2021, 143, 516–525. [Google Scholar] [CrossRef]
  21. Abraham, W.T.; Lindenfeld, J.; Ponikowski, P.; Agostoni, P.; Butler, J.; Desai, A.S.; Filippatos, G.; Gniot, J.; Fu, M.; Gullestad, L.; et al. Effect of empagliflozin on exercise ability and symptoms in heart failure patients with reduced and preserved ejection fraction, with and without type 2 diabetes. Eur. Heart J. 2021, 42, 700–710. [Google Scholar] [CrossRef]
  22. Wanner, C.; Lachin, J.M.; Inzucchi, S.E.; Fitchett, D.; Mattheus, M.; George, J.; Woerle, H.J.; Broedl, U.C.; von Eynatten, M.; Zinman, B.; et al. Empagliflozin and Clinical Outcomes in Patients With Type 2 Diabetes Mellitus, Established Cardiovascular Disease, and Chronic Kidney Disease. Circulation 2018, 137, 119–129. [Google Scholar] [CrossRef]
  23. Mazer, C.D.; Hare, G.M.T.; Connelly, P.W.; Gilbert, R.E.; Shehata, N.; Quan, A.; Teoh, H.; Leiter, L.A.; Zinman, B.; Juni, P.; et al. Effect of Empagliflozin on Erythropoietin Levels, Iron Stores, and Red Blood Cell Morphology in Patients With Type 2 Diabetes Mellitus and Coronary Artery Disease. Circulation 2020, 141, 704–707. [Google Scholar] [CrossRef] [PubMed]
  24. Hoshika, Y.; Kubota, Y.; Mozawa, K.; Tara, S.; Tokita, Y.; Yodogawa, K.; Iwasaki, Y.K.; Yamamoto, T.; Takano, H.; Tsukada, Y.; et al. Effect of Empagliflozin Versus Placebo on Body Fluid Balance in Patients With Acute Myocardial Infarction and Type 2 Diabetes Mellitus: Subgroup Analysis of the EMBODY Trial. J. Card. Fail. 2022, 28, 56–64. [Google Scholar] [CrossRef] [PubMed]
  25. Zannad, F.; Ferreira, J.P.; Pocock, S.J.; Zeller, C.; Anker, S.D.; Butler, J.; Filippatos, G.; Hauske, S.J.; Brueckmann, M.; Pfarr, E.; et al. Cardiac and Kidney Benefits of Empagliflozin in Heart Failure Across the Spectrum of Kidney Function: Insights From EMPEROR-Reduced. Circulation 2021, 143, 310–321. [Google Scholar] [CrossRef] [PubMed]
  26. Wanner, C.; Inzucchi, S.E.; Lachin, J.M.; Fitchett, D.; von Eynatten, M.; Mattheus, M.; Johansen, O.E.; Woerle, H.J.; Broedl, U.C.; Zinman, B.; et al. Empagliflozin and Progression of Kidney Disease in Type 2 Diabetes. N. Engl. J. Med. 2016, 375, 323–334. [Google Scholar] [CrossRef] [PubMed]
  27. Cherney, D.Z.I.; Zinman, B.; Inzucchi, S.E.; Koitka-Weber, A.; Mattheus, M.; von Eynatten, M.; Wanner, C. Effects of empagliflozin on the urinary albumin-to-creatinine ratio in patients with type 2 diabetes and established cardiovascular disease: An exploratory analysis from the EMPA-REG OUTCOME randomised, placebo-controlled trial. Lancet Diabetes Endocrinol. 2017, 5, 610–621. [Google Scholar] [CrossRef]
  28. Wanner, C.; Heerspink, H.J.L.; Zinman, B.; Inzucchi, S.E.; Koitka-Weber, A.; Mattheus, M.; Hantel, S.; Woerle, H.J.; Broedl, U.C.; von Eynatten, M.; et al. Empagliflozin and Kidney Function Decline in Patients with Type 2 Diabetes: A Slope Analysis from the EMPA-REG OUTCOME Trial. J. Am. Soc. Nephrol. 2018, 29, 2755–2769. [Google Scholar] [CrossRef] [PubMed]
  29. Halden, T.A.S.; Kvitne, K.E.; Midtvedt, K.; Rajakumar, L.; Robertsen, I.; Brox, J.; Bollerslev, J.; Hartmann, A.; Asberg, A.; Jenssen, T. Efficacy and Safety of Empagliflozin in Renal Transplant Recipients With Posttransplant Diabetes Mellitus. Diabetes Care 2019, 42, 1067–1074. [Google Scholar] [CrossRef]
  30. Sattar, N.; Fitchett, D.; Hantel, S.; George, J.T.; Zinman, B. Empagliflozin is associated with improvements in liver enzymes potentially consistent with reductions in liver fat: Results from randomised trials including the EMPA-REG OUTCOME(R) trial. Diabetologia 2018, 61, 2155–2163. [Google Scholar] [CrossRef]
  31. Kuchay, M.S.; Krishan, S.; Mishra, S.K.; Farooqui, K.J.; Singh, M.K.; Wasir, J.S.; Bansal, B.; Kaur, P.; Jevalikar, G.; Gill, H.K.; et al. Effect of Empagliflozin on Liver Fat in Patients With Type 2 Diabetes and Nonalcoholic Fatty Liver Disease: A Randomized Controlled Trial (E-LIFT Trial). Diabetes Care 2018, 41, 1801–1808. [Google Scholar] [CrossRef]
  32. Taheri, H.; Malek, M.; Ismail-Beigi, F.; Zamani, F.; Sohrabi, M.; Reza Babaei, M.; Khamseh, M.E. Effect of Empagliflozin on Liver Steatosis and Fibrosis in Patients With Non-Alcoholic Fatty Liver Disease Without Diabetes: A Randomized, Double-Blind, Placebo-Controlled Trial. Adv. Ther. 2020, 37, 4697–4708. [Google Scholar] [CrossRef]
  33. Sone, H.; Kaneko, T.; Shiki, K.; Tachibana, Y.; Pfarr, E.; Lee, J.; Tajima, N. Efficacy and safety of empagliflozin as add-on to insulin in Japanese patients with type 2 diabetes: A randomized, double-blind, placebo-controlled trial. Diabetes Obes. Metab. 2020, 22, 417–426. [Google Scholar] [CrossRef]
  34. Kaku, K.; Chin, R.; Naito, Y.; Iliev, H.; Ikeda, R.; Ochiai, K.; Yasui, A. Safety and effectiveness of empagliflozin in Japanese patients with type 2 diabetes: Interim analysis from a post-marketing surveillance study. Expert Opin. Drug Saf. 2020, 19, 211–221. [Google Scholar] [CrossRef]
  35. Monteiro, P.; Bergenstal, R.M.; Toural, E.; Inzucchi, S.E.; Zinman, B.; Hantel, S.; Kis, S.G.; Kaspers, S.; George, J.T.; Fitchett, D. Efficacy and safety of empagliflozin in older patients in the EMPA-REG OUTCOME(R) trial. Age Ageing 2019, 48, 859–866. [Google Scholar] [CrossRef]
  36. Herat, L.Y.; Matthews, V.B.; Magno, A.L.; Kiuchi, M.G.; Carnagarin, R.; Schlaich, M.P. An evaluation of empagliflozin and it’s applicability to hypertension as a therapeutic option. Expert Opin. Pharmacother. 2020, 21, 1157–1166. [Google Scholar] [CrossRef]
  37. Devi, R.; Mali, G.; Chakraborty, I.; Unnikrishnan, M.K.; Abdulsalim, S. Efficacy and safety of empagliflozin in type 2 diabetes mellitus: A meta-analysis of randomized controlled trials. Postgrad. Med. 2017, 129, 382–392. [Google Scholar] [CrossRef]
  38. SFDA. Saudi vigilance, Saudi Food & Drug Authority (SFDA): The Risk of Rare but Serious Infection in the Genital Area with Sodium Glucose Cotransporter-2 (SGLT2) Inhibitors. 2018. Available online: https://www.sfda.gov.sa/sites/default/files/2021-02/Jardiance%C2%AE%20SAFTEY%20COMM.pdf (accessed on 1 June 2018).
  39. Kadowaki, T.; Haneda, M.; Inagaki, N.; Terauchi, Y.; Taniguchi, A.; Koiwai, K.; Rattunde, H.; Woerle, H.J.; Broedl, U.C. Empagliflozin monotherapy in Japanese patients with type 2 diabetes mellitus: A randomized, 12-week, double-blind, placebo-controlled, phase II trial. Adv. Ther. 2014, 31, 621–638. [Google Scholar] [CrossRef]
  40. Ferrannini, E.; Berk, A.; Hantel, S.; Pinnetti, S.; Hach, T.; Woerle, H.J.; Broedl, U.C. Long-term safety and efficacy of empagliflozin, sitagliptin, and metformin: An active-controlled, parallel-group, randomized, 78-week open-label extension study in patients with type 2 diabetes. Diabetes Care 2013, 36, 4015–4021. [Google Scholar] [CrossRef]
  41. Rosenstock, J.; Aggarwal, N.; Polidori, D.; Zhao, Y.; Arbit, D.; Usiskin, K.; Capuano, G.; Canovatchel, W.; Canagliflozin, D.I.A.S.G. Dose-ranging effects of canagliflozin, a sodium-glucose cotransporter 2 inhibitor, as add-on to metformin in subjects with type 2 diabetes. Diabetes Care 2012, 35, 1232–1238. [Google Scholar] [CrossRef]
  42. Rosenstock, J.; Seman, L.J.; Jelaska, A.; Hantel, S.; Pinnetti, S.; Hach, T.; Woerle, H.J. Efficacy and safety of empagliflozin, a sodium glucose cotransporter 2 (SGLT2) inhibitor, as add-on to metformin in type 2 diabetes with mild hyperglycaemia. Diabetes Obes. Metab. 2013, 15, 1154–1160. [Google Scholar] [CrossRef]
  43. Kishimoto, M.; Yamaoki, K.; Adachi, M. Combination Therapy with Empagliflozin and Insulin Results in Successful Glycemic Control: A Case Report of Uncontrolled Diabetes Caused by Autoimmune Pancreatitis and Subsequent Steroid Treatment. Case Rep. Endocrinol. 2019, 2019, 9415347. [Google Scholar] [CrossRef]
  44. Al-Goblan, A.S.; Al-Alfi, M.A.; Khan, M.Z. Mechanism linking diabetes mellitus and obesity. Diabetes Metab. Syndr. Obes. 2014, 7, 587–591. [Google Scholar] [CrossRef] [PubMed]
  45. Rosenstock, J.; Jelaska, A.; Frappin, G.; Salsali, A.; Kim, G.; Woerle, H.J.; Broedl, U.C.; Investigators, E.-R.M.T. Improved glucose control with weight loss, lower insulin doses, and no increased hypoglycemia with empagliflozin added to titrated multiple daily injections of insulin in obese inadequately controlled type 2 diabetes. Diabetes Care 2014, 37, 1815–1823. [Google Scholar] [CrossRef] [PubMed]
  46. Bolinder, J.; Ljunggren, O.; Kullberg, J.; Johansson, L.; Wilding, J.; Langkilde, A.M.; Sugg, J.; Parikh, S. Effects of dapagliflozin on body weight, total fat mass, and regional adipose tissue distribution in patients with type 2 diabetes mellitus with inadequate glycemic control on metformin. J. Clin. Endocrinol. Metab. 2012, 97, 1020–1031. [Google Scholar] [CrossRef] [PubMed]
  47. Zhao, D.; Liu, H.; Dong, P. Empagliflozin reduces blood pressure and uric acid in patients with type 2 diabetes mellitus: A systematic review and meta-analysis. J. Hum. Hypertens. 2019, 33, 327–339. [Google Scholar] [CrossRef] [PubMed]
  48. Boorsma, E.M.; Beusekamp, J.C.; Ter Maaten, J.M.; Figarska, S.M.; Danser, A.H.J.; van Veldhuisen, D.J.; van der Meer, P.; Heerspink, H.J.L.; Damman, K.; Voors, A.A. Effects of empagliflozin on renal sodium and glucose handling in patients with acute heart failure. Eur. J. Heart Fail. 2021, 23, 68–78. [Google Scholar] [CrossRef] [PubMed]
  49. Schork, A.; Saynisch, J.; Vosseler, A.; Jaghutriz, B.A.; Heyne, N.; Peter, A.; Haring, H.U.; Stefan, N.; Fritsche, A.; Artunc, F. Effect of SGLT2 inhibitors on body composition, fluid status and renin-angiotensin-aldosterone system in type 2 diabetes: A prospective study using bioimpedance spectroscopy. Cardiovasc. Diabetol. 2019, 18, 46. [Google Scholar] [CrossRef] [PubMed]
  50. Ferdinand, K.C.; Izzo, J.L.; Lee, J.; Meng, L.; George, J.; Salsali, A.; Seman, L. Antihyperglycemic and Blood Pressure Effects of Empagliflozin in Black Patients With Type 2 Diabetes Mellitus and Hypertension. Circulation 2019, 139, 2098–2109. [Google Scholar] [CrossRef]
  51. Palmiero, G.; Cesaro, A.; Vetrano, E.; Pafundi, P.C.; Galiero, R.; Caturano, A.; Moscarella, E.; Gragnano, F.; Salvatore, T.; Rinaldi, L.; et al. Impact of SGLT2 Inhibitors on Heart Failure: From Pathophysiology to Clinical Effects. Int. J. Mol. Sci. 2021, 22, 5863. [Google Scholar] [CrossRef]
  52. Salvatore, T.; Caturano, A.; Galiero, R.; Di Martino, A.; Albanese, G.; Vetrano, E.; Sardu, C.; Marfella, R.; Rinaldi, L.; Sasso, F.C. Cardiovascular Benefits from Gliflozins: Effects on Endothelial Function. Biomedicines 2021, 9, 1356. [Google Scholar] [CrossRef]
  53. Perrone-Filardi, P.; Avogaro, A.; Bonora, E.; Colivicchi, F.; Fioretto, P.; Maggioni, A.P.; Sesti, G.; Ferrannini, E. Mechanisms linking empagliflozin to cardiovascular and renal protection. Int. J. Cardiol. 2017, 241, 450–456. [Google Scholar] [CrossRef]
  54. Pabel, S.; Wagner, S.; Bollenberg, H.; Bengel, P.; Kovacs, A.; Schach, C.; Tirilomis, P.; Mustroph, J.; Renner, A.; Gummert, J.; et al. Empagliflozin directly improves diastolic function in human heart failure. Eur. J. Heart Fail. 2018, 20, 1690–1700. [Google Scholar] [CrossRef]
  55. Cherney, D.; Lund, S.S.; Perkins, B.A.; Groop, P.H.; Cooper, M.E.; Kaspers, S.; Pfarr, E.; Woerle, H.J.; von Eynatten, M. The effect of sodium glucose cotransporter 2 inhibition with empagliflozin on microalbuminuria and macroalbuminuria in patients with type 2 diabetes. Diabetologia 2016, 59, 1860–1870. [Google Scholar] [CrossRef] [PubMed]
  56. Perry, R.J.; Samuel, V.T.; Petersen, K.F.; Shulman, G.I. The role of hepatic lipids in hepatic insulin resistance and type 2 diabetes. Nature 2014, 510, 84–91. [Google Scholar] [CrossRef] [PubMed]
  57. Kern, M.; Kloting, N.; Mark, M.; Mayoux, E.; Klein, T.; Bluher, M. The SGLT2 inhibitor empagliflozin improves insulin sensitivity in db/db mice both as monotherapy and in combination with linagliptin. Metabolism 2016, 65, 114–123. [Google Scholar] [CrossRef]
  58. Hattori, S. Empagliflozin decreases remnant-like particle cholesterol in type 2 diabetes patients with insulin resistance. J Diabetes Investig. 2018, 9, 870–874. [Google Scholar] [CrossRef]
  59. Jojima, T.; Sakurai, S.; Wakamatsu, S.; Iijima, T.; Saito, M.; Tomaru, T.; Kogai, T.; Usui, I.; Aso, Y. Empagliflozin increases plasma levels of campesterol, a marker of cholesterol absorption, in patients with type 2 diabetes: Association with a slight increase in high-density lipoprotein cholesterol. Int. J. Cardiol. 2021, 331, 243–248. [Google Scholar] [CrossRef]
Figure 1. Change from baseline in HbA1c%, FPG mmol/L, and body weight kg at week 12 (t-test).
Figure 1. Change from baseline in HbA1c%, FPG mmol/L, and body weight kg at week 12 (t-test).
Jcm 11 04769 g001
Figure 2. Change from baseline in SBP mmHg, DBP mmHg, and pulse rate bpm at week 12 (t-test).
Figure 2. Change from baseline in SBP mmHg, DBP mmHg, and pulse rate bpm at week 12 (t-test).
Jcm 11 04769 g002
Figure 3. Change from baseline in UACR mg/g and eGFR mL/min/1.73 m2 at week 12 (t-test).
Figure 3. Change from baseline in UACR mg/g and eGFR mL/min/1.73 m2 at week 12 (t-test).
Jcm 11 04769 g003
Figure 4. Change from baseline in AST U/l, ALT U/L, GGT U/L, LDL mmol/L, HDL-c mmol/L, and TG mmol/L at week 12 (t-test).
Figure 4. Change from baseline in AST U/l, ALT U/L, GGT U/L, LDL mmol/L, HDL-c mmol/L, and TG mmol/L at week 12 (t-test).
Jcm 11 04769 g004
Table 1. Patient demographics.
Table 1. Patient demographics.
Patient Demographics
Sample volume, n256
Age (years), mean (SD)58.9 (10.75)
Sex Males, n113
Females, n143
Male age, mean (SD)58.2 (11.88)
Female age, mean (SD)59.4 (9.76)
Duration since Diagnosis of T2DM, (years)
Mean (SD)16.7 (8.47)
<1, n (%)0
1 to 522 (9%)
> 5234 (91%)
DM Treatment before Empagliflozin
Insulin + OHA (metformin)156 (64%)
OHA (metformin)93 (36%)
SD: Standard deviation, T2DM: Type 2 diabetes mellitus, DM: Diabetes mellitus, OHA: Orally administered antihyperglycemic.
Table 2. Efficacy and Safety Parameters at baseline and week 12.
Table 2. Efficacy and Safety Parameters at baseline and week 12.
Baseline GroupEmpagliflozin GroupChange Amount
Mean (SD)Mean (SD)
Efficacy:
Haemoglobin A1c HbA1c, (%)9.77 ± 1.768.85 ± 4.83−0.92
Participants treated with Insulin + OHA, HbA1c, (%)9.94 ± 1.848.66 ± 1.47−1.27
Participants treated with9.46 ± 1.568.31 ± 1.26−1.15
OHA, HbA1c, (%)
Fasting Plasma Glucose FPG, mmol/L11.27 ± 4.798.95 ± 3.37−2.32
Body weight, kg89.99 ± 18.09 88.03 ± 18.47−1.96
Safety:
Systolic Blood Pressure SBP, mmHg142.6 ± 19.45138.8 ± 20.23−3.8
Diastolic Blood Pressure DBP, mmHg79.6 ± 20.3279.4 ± 21.14−0.2
Pulse Rate, (bpm)85.98 ± 11.3384.80 ± 13.52−1.18
Kidney Function Status
Urine Albumin/Creatinine Ratio UACR (mg/g)20.39 ± 43.7217.12 ± 40.05−3.27
eGFR, mL/min/1.73 m251.12 ± 120.4572.51 ± 22.8021.39
Liver Function Status
Aspartate aminotransferase AST, U/L22.92 ± 8.1021.65 ± 6.38−1.26
Alanine aminotransferase ALT, U/L25.96 ± 8.0923.91 ± 11.71−0.70
A gamma-glutamyl transferase (GGT), U/L30.29 ± 25.1527.12 ± 18.32−4.31
LDL- cholesterol, mmol/L2.543 ± 0.932.544 ± 1.500.0009
HDL- cholesterol, mmol/L1.69 ± 3.59 1.98 ± 0.22 0.29
Triglycerides TG, mmol/L2.11 ± 5.901.66 ± 0.20−0.45
HbA1c: haemoglobin A1c (glycosylated haemoglobin), FPG: fasting plasma glucose, SD: standard deviation, OHA: Orally administered antihyperglycemic, eGFR: Estimated glomerular filtration rate, LDL: Low-density lipoprotein, HDL: High-density lipoprotein, AST: Aspartate aminotransferase, ALT: Alanine transaminase and TG: Triglycerides. A comparison between Empagliflozin-treated group at week 12 with the same group at bassline. Data are n (%) or mean (SD).
Table 3. Change in Efficacy and Safety Parameters at week 12.
Table 3. Change in Efficacy and Safety Parameters at week 12.
Baseline Start Treatment EmpagliflozinChange from Baseline at Week 12Rate of Change
Efficacy variables
Mean change in HbA1c −0.93 (−0.32, −1.54) −0.106
from baseline, % (95% CI)
Mean ± SD−0.93 ± 4.93
<7.0%5 (0.02)21 (0.08)0.76
≥7% to <8%4 (0.016)76 (0.30)0.95
≥8% to <9%80 (0.31)82 (0.32)0.02
≥9%167 (0.65)77 (0.30)−0.54
Decrease in HbA1c (%) in participants:
≥ 0.5%, n, (%)
181(0.71)
Treated with insulin + OHA, HbA1c, (%)−1.28(−1.03, −1.53) −0.147
Treated with OHA, HbA1c (%)−1.11(−0.799, −1.42) −0.139
Decrease in insulin units in participants treated with insulin + OHA 9.94 ± 1.848.67 ± 1.47−1.27
Mean change in FPG −2.28 (−2.81, −1.75) −0.25
from baseline (95% CI)
Mean ± SD−2.27 ± 4.22
<7.8, n, (%)63 (0.25)115 (0.45)0.83
7.8 to <11.080 (0.32)86 (0.34)0.08
≥11113 (0.44)55 (0.21)−0.51
Mean change in body weight−0.874 (−4.36, −6.10) −0.02
from baseline (95% CI)
Mean ± SD−1.96 ± 11.98
Safety variables
Mean change in SBP−3.85 (−6.81, −0.88) −0.03
from baseline, % (95% CI)
Mean ± SD−3.82 ± 23.4
Mean change in DBP−0.06 (−0.81, −0.88) −0.001
from baseline, % (95% CI)
Mean ± SD−0.05 ± 0.28
Mean change in pulse rate−1.18 (−0.79, −3.15) −0.01
from baseline (95% CI)
Mean ± SD−1.18 ± 15.46
Kidney Function Status
Mean change in Urine Albumin/ −0.067
Creatinine Ratio UACR (mg/g) −1.76 (−1.07, −34.25)
Mean ± SD−1.76 ± 103.5
<30, n, (%)126 (0.50)177(0.70)0.4
30 to <300 n, (%)51(0.21)67 (0.27)0.31
≥300, n, (%)75 (0.29)8 (0.03)−0.89
Mean change in body eGFR3.54 (2.78, 9.87) −0.418
from baseline, kg (95% CI)
Mean ± SD3.54 ± 45.98
≥90, n, (%)49 (0.19)55 (0.21)0.02
60 to < 90120 (0.47)116 (0.45)−0.018
30 to < 6087 (0.34)84 (0.33)−0.016
Liver Function Status
Mean change in AST −1.26 (−0.3011, −2.227) −0.058
from baseline (95% CI)
Mean ± SD−1.263 ± 7.72
Mean change in ALT−2.36 (−1.031, −3.69) −0.029
from baseline (95% CI)
Mean ± SD−2.36 ± 10.75
Mean change in GGT−4.31 (−2.33, −6.28) −0.159
from baseline (95% CI)
Mean ± SD−4.31 ± 15.66
Mean change in LDL0.005 (0.192, 0.18) 0.0004
from baseline (95% CI)
Mean ± SD0.005 ± 1.46
Mean change in HDL0.29 (0.74, 0.15) 0.171
from baseline, kg (95% CI)
Mean ± SD0.293 ± 3.61
Mean change in TG−0.43 (−0.31, −1.17) −0.271
from baseline (95% CI)
Mean ± SD−0.45 ± 5.98
A comparison between EMPA treated group at week 12 with the same group at baseline. Data are n (%) or mean (SD).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Althobaiti, F.M.; Alsanosi, S.M.; Falemban, A.H.; Alzahrani, A.R.; Fataha, S.A.; Salih, S.O.; Alrumaih, A.M.; Alotaibi, K.N.; Althobaiti, H.M.; Al-Ghamdi, S.S.; et al. Efficacy and Safety of Empagliflozin in Type 2 Diabetes Mellitus Saudi Patients as Add-On to Antidiabetic Therapy: A Prospective, Open-Label, Observational Study. J. Clin. Med. 2022, 11, 4769. https://doi.org/10.3390/jcm11164769

AMA Style

Althobaiti FM, Alsanosi SM, Falemban AH, Alzahrani AR, Fataha SA, Salih SO, Alrumaih AM, Alotaibi KN, Althobaiti HM, Al-Ghamdi SS, et al. Efficacy and Safety of Empagliflozin in Type 2 Diabetes Mellitus Saudi Patients as Add-On to Antidiabetic Therapy: A Prospective, Open-Label, Observational Study. Journal of Clinical Medicine. 2022; 11(16):4769. https://doi.org/10.3390/jcm11164769

Chicago/Turabian Style

Althobaiti, Fahad M., Safaa M. Alsanosi, Alaa H. Falemban, Abdullah R. Alzahrani, Salma A. Fataha, Sara O. Salih, Ali M. Alrumaih, Khalid N. Alotaibi, Hazim M. Althobaiti, Saeed S. Al-Ghamdi, and et al. 2022. "Efficacy and Safety of Empagliflozin in Type 2 Diabetes Mellitus Saudi Patients as Add-On to Antidiabetic Therapy: A Prospective, Open-Label, Observational Study" Journal of Clinical Medicine 11, no. 16: 4769. https://doi.org/10.3390/jcm11164769

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop