Systematic Reviews Open Access
Copyright ©The Author(s) 2019. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Diabetes. Feb 15, 2019; 10(2): 114-132
Published online Feb 15, 2019. doi: 10.4239/wjd.v10.i2.114
SGLT-2 inhibitors in non-alcoholic fatty liver disease patients with type 2 diabetes mellitus: A systematic review
Henith Raj, Harsh Durgia, Rajan Palui, Sadishkumar Kamalanathan, Jayaprakash Sahoo, Department of Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India
Sandhiya Selvarajan, Department of Clinical Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India
Sitanshu Sekhar Kar, Department of Preventive and Social Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India
ORCID number: Henith Raj (0000-0002-1499-4021); Harsh Durgia (0000-0002-8404-5729); Rajan Palui (0000-0002-2429-3595); Sadishkumar Kamalanathan (0000-0002-2371-0625); Sandhiya Selvarajan (0000-0002-7948-7821); Sitanshu Sekhar Kar (0000-0001-7122-523X); Jayaprakash Sahoo (0000-0002-8805-143X).
Author contributions: Raj H, Durgia H, and Palui R designed the work; Kamalanathan SK, Selvarajan S, Kar SS, and Sahoo JP interpreted the data; Raj H, Durgia H, and Palui R revised it critically for important intellectual content; Kamalanathan SK, Selvarajan S, Kar SS, and Sahoo JP drafted the work; all authors approved the final version of the manuscript.
Conflict-of-interest statement: All authors have no conflicts of interest to report.
PRISMA 2009 Checklist statement: The authors have read the PRISMA 2009 Checklist, and the manuscript was prepared and revised according to the PRISMA 2009 Checklist.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Corresponding author: Jayaprakash Sahoo, MD, DM, Associate Professor, Department of Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research, Room No. 5444, the 4th Floor, Superspeciality block, Puducherry 605006, India. jayaprakash.s@jipmer.edu.in
Telephone: +91-9629158368
Received: October 5, 2018
Peer-review started: October 6, 2018
First decision: November 15, 2018
Revised: December 14, 2018
Accepted: December 29, 2018
Article in press: December 30, 2018
Published online: February 15, 2019

Abstract
BACKGROUND

Non-alcoholic fatty liver disease (NAFLD) is a common comorbidity with type 2 diabetes. The existing therapeutic options for NAFLD are not adequate. Hypocaloric diet and exercise is the cornerstone of therapy in NAFLD. Pioglitazone is the only drug recommended in diabetes patients with biopsy proven non-alcoholic steatohepatitis. The frequent coexistence of NAFLD and type 2 diabetes with their combined adverse health consequences and inadequate therapeutic options makes it necessary to search for newer alternatives.

AIM

To assess the effect of sodium glucose cotransporter-2 (SGLT-2) inhibitors on liver enzymes in type 2 diabetes patients with NAFLD.

METHODS

We searched PubMed/MEDLINE, Cochrane library, Google scholar, and Clinicaltrials.gov for the relevant articles to be included in this systematic review. Human studies done in type 2 diabetes patients with NAFLD treated with SGLT-2 inhibitors for at least 12 wk were included. Data from eight studies (four randomised controlled trials and four observational studies) were extracted and a narrative synthesis was done. A total of 214 patients were treated with SGLT-2 inhibitors in these studies (94 in randomised controlled trials and 120 in observational studies).

RESULTS

The primary outcome measure was change in serum alanine aminotransferase level. Out of eight studies, seven studies showed a significant decrease in serum alanine aminotransferase level. Most of the studies revealed reduction in serum level of other liver enzymes like aspartate aminotransferase and gamma glutamyl transferase. Five studies that reported a change in hepatic fat exhibited a significant reduction in hepatic fat content in those treated with SGLT-2 inhibitors. Likewise, among the three studies that evaluated a change in indices of hepatic fibrosis, two studies revealed a significant improvement in liver fibrosis. Moreover, there was an improvement in obesity, insulin resistance, glycaemia, and lipid parameters in those subjects taking SGLT-2 inhibitors. The studies disclosed that about 17% (30/176) of the subjects taking SGLT-2 inhibitors developed adverse events and more than 40% (10/23) of them had genitourinary tract infections.

CONCLUSION

Based on low to moderate quality of evidence, SGLT-2 inhibitors improve the serum level of liver enzymes, decrease liver fat, and fibrosis with additional beneficial effects on various metabolic parameters in type 2 diabetes patients with NAFLD.

Key Words: Alanine aminotransferase, Hepatic fat, Hepatic fibrosis, Non-alcoholic fatty liver disease, Sodium-glucose cotransporter-2 inhibitor, Type 2 diabetes mellitus

Core tip: The frequent coexistence of non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes, their adverse health consequences, and lack of adequate therapeutic options makes it necessary to search for newer alternatives. Currently, pioglitazone and vitamin E are recommended in addition to lifestyle modifications for the management of NAFLD. Animal studies have shown that sodium glucose cotransporter-2 inhibitors might be beneficial in NAFLD present in diabetes patients. The current systematic review shows that sodium glucose cotransporter-2 inhibitors improve the serum level of liver enzymes, liver fat, and liver fibrosis with additional beneficial effects on various metabolic parameters in type 2 diabetes patients with NAFLD.



INTRODUCTION

Non-alcoholic fatty liver disease (NAFLD) is an emerging public health issue worldwide. The prevalence of NAFLD in type 2 diabetes mellitus patients is three times greater as compared to the general population. Its prevalence in diabetic subjects ranges from 69%-87% depending upon the imaging modality used[1]. The spectrum of NAFLD includes simple steatosis, steatohepatitis, and cirrhosis[2]. Besides NAFLD is a risk factor for extrahepatic complications like cardiovascular disease, chronic kidney disease, and type 2 diabetes. In addition, the prevalence of both microvascular and macrovascular complications is increased in patients with NAFLD and type 2 diabetes[3].

The existing therapeutic options for NAFLD are not adequate. Hypocaloric diet and exercise is the cornerstone of therapy in NAFLD. Pioglitazone and vitamin E are recommended only in biopsy-proven non-alcoholic steatohepatitis (NASH), but vitamin E is not recommended in diabetic patients due to inadequate evidence[4]. The frequent coexistence of NAFLD and type 2 diabetes with their combined adverse health consequences and inadequate therapeutic options makes it necessary to search for newer alternatives. Based on the information from animal studies, sodium glucose cotransporter-2 (SGLT-2) inhibitors appear promising in the management of NAFLD[5-7]. This systematic review is an effort to review the available literature on the effect of SGLT-2 inhibitors on NAFLD in type 2 diabetes patients.

MATERIALS AND METHODS
Protocol and registration

This systematic review was performed according to the predefined protocol registered in PROSPERO (Registration ID: CRD42018104572). The protocol can be accessed at the website address https://http://www.crd.york.ac.uk/prospero. We followed the Preferred Reporting Items for Systematic Reviews and Meta-Analysis 2009 guidelines for reporting this systematic review[8]. Ethics committee approval was not required for this systematic review because it was done using published data found in the public domain.

Eligibility criteria

All observational and randomised controlled trials (RCTs) done using SGLT-2 inhibitors among type 2 diabetes patients with NAFLD having both baseline and post-treatment serum alanine aminotransferase (ALT) level data with a minimum follow-up duration of 12 wk were included in this systematic review. The studies with concomitant pharmacological therapy like pioglitazone or α-tocopherol (vitamin E) for treating NAFLD were excluded to avoid the confounding effects of these drugs on liver function tests. Only those studies that were done in humans and published in English were considered for inclusion. We excluded abstract-only articles, case reports, conference presentations, editorials, reviews, expert opinions, and studies with five participants and less.

Primary and secondary outcomes

The primary outcome was the change in serum ALT levels in type 2 diabetes patients with NAFLD treated with SGLT-2 inhibitors. The secondary outcomes were change in serum aspartate aminotransferase (AST) and gamma-glutamyl transferase (GGT) levels, hepatic fat, hepatic fibrosis, metabolic profile, anthropometric parameters, and the adverse effects of SGLT-2 inhibitors.

Information sources

PubMed/MEDLINE, Cochrane library, Google scholar, and Clinicaltrials.gov were searched from their date of inception until 31st August, 2018.

Literature search and study selection

The search terms/MeSH terms used were “NAFLD”, “Nonalcoholic fatty liver disease”, “Non-alcoholic fatty liver disease”, “Non alcoholic fatty liver disease”, “NASH”, “Non-alcoholic steatohepatitis”, “Nonalcoholic steatohepatitis”, “Non alcoholic steatohepatitis”, “Fatty liver”, “Type 2 diabetes mellitus”, “Type 2 diabetes”, “Diabetes mellitus type 2”, “Diabetes type 2”, “SGLT-2 inhibitors”, “Sodium glucose cotransporter-2 inhibitors”, “SGLT-2”, “SGLT2”, “SGLT 2”, “Canagliflozin”, “Dapagliflozin”, “Empagliflozin”, “Ipragliflozin”, “Luseogliflozin”, “Tofogliflozin”, “Sotagliflozin”, “Remogliflozin”, “Ertugliflozin”, and “Sergliflozin”(Table 1). The references of the search articles were scrutinised for relevant articles.

Table 1 Literature search strategy.
S. NoSearch terms
1NAFLD
2Nonalcoholic fatty liver disease
3Non-alcoholic fatty liver disease
4Non alcoholic fatty liver disease
5NASH
6Non-alcoholic steatohepatitis
7Nonalcoholic steatohepatitis
8Non alcoholic steatohepatitis
9Fatty liver
101 OR 2 OR 3 OR 4 OR 5 OR 6 OR 7 OR 8 OR 9
11Type 2 diabetes mellitus
12Type 2 diabetes
13Diabetes mellitus type 2
14Diabetes type 2
1511 OR 12 OR 13 OR 14
16SGLT-2 inhibitors
17Sodium glucose cotransporter-2 inhibitors
18SGLT-2
19SGLT2
20SGLT 2
21Canagliflozin
22Dapagliflozin
23Empagliflozin
24Ipragliflozin
25Luseogliflozin
26Tofogliflozin
27Sotagliflozin
28Remogliflozin
29Ertugliflozin
30Sergliflozin
3116 OR 17 OR 18 OR 19 OR 20 OR 21 OR 22 OR 23 OR 24 OR 25 OR 26 OR 27 OR 28 OR 29 OR 30
3210 AND 15 AND 31
Data collection process

The titles and/or abstracts of studies were retrieved using the search strategy and those from additional sources were scrutinised independently by two review authors (HR and JPS) to identify studies that potentially met the inclusion criteria as outlined above. The full texts of these potentially eligible studies were retrieved and independently assessed for eligibility by three review team members (HD, SS, and RP). Any disagreements between the reviewers over the eligibility of particular studies were resolved through discussion with a fourth senior reviewer (SKK). A standardised, pre-formatted excel form was used to extract data from the included studies for the assessment of study quality.

Data items and synthesis of results

The extracted data included the author of the study with year, the study methodology, the recruitment and study completion rates, the types of population, the exposure/intervention (dose of SGLT-2 inhibitor, duration), the results (outcome measures like change in serum ALT, AST, GGT, hepatic fat, markers of liver fibrosis, fasting plasma glucose (FPG), glycosylated haemoglobin (HbA1c), lipid profile, homeostasis model assessment-estimated insulin resistance (HOMA-IR), body mass index (BMI), any adverse effects, information for the assessment of the risk of bias, and sources of funding/support.

The statistical review of the study was performed by a biomedical statistician (SSK). A narrative synthesis of the results of individual studies was done. The change in the difference in means and difference in proportions and the respective P values as mentioned in the original manuscripts were tabulated and explained in our study.

Risk of study bias

The risk of bias of the RCTs was done using Cochrane risk of bias tool[9]. The studies were graded as “good quality” or “fair quality” or “poor quality” according to the level of risk. Methodological Index for Non-Randomized Studies (MINORS) scale was used to assess the risk of bias of observational studies[10]. A study was considered to be an ideal study if the score was 16 for single arm and 24 for comparative studies.

RESULTS
Study selection

Our literature search from all the aforementioned databases yielded 73 articles (including references of the relevant articles). After eliminating duplicate articles, 55 articles were screened. Eight articles met all of the inclusion criteria (total 214 patients were on SGLT-2 inhibitors) (Figure 1).

Figure 1
Figure 1 Literature search and study selection.
Study characteristics

The summary of all studies included in this systematic review is given in Tables 2 and 3. Out of the eight studies, four are RCTs[11-14] and four are observational[15-18]. Five studies were conducted amongst the Japanese population. Ipragliflozin was used in three studies whereas canagliflozin and luseogliflozin were used in two studies each, but dapagliflozin and empagliflozin were used in one study each. All studies used one type of SGLT-2 inhibitor except the one authored by Seko et al[16], where both canagliflozin and ipragliflozin were used. The change in serum ALT was a secondary outcome while the effect of SGLT- 2 inhibitors on liver fat was the primary outcome in all RCTs.

Table 2 Randomised controlled trials.
S. NoRef.Inclusion criteriaAge (yr)Male genderIntervention armControl armFollow-up durationPrimary outcome
1Kuchay et al[11], 2018Age > 20 yr, hepatic steatosis (MRI-PDFF > 6%), HbA1c > 7.0% to < 10.0%Intervention arm: 50.7 (12.8)Intervention arm: 16 (64%)Standard treatment + Empagliflozin 10 mg daily (n = 25)Standard treatment (n = 25)20 wkChange in liver fat content by MRI-PDFF
Control arm: 49.1 (10.3)Control arm: 17 (68%)
2Ito et al[12], 2017Age 20-75 yr, HbA1c 7.0–11.0%, BMI < 45 kg/m2, On diet and exercise therapy alone or with oral hypoglycaemic agents other than SGLT-2 inhibitors and thiazolidinediones and/or insulin, NAFLD, findings suggesting hepatic steatosis and hepatic dysfunction on clinical laboratory tests or on imaging studies (e.g., computed tomography or ultrasound)Pioglitazone arm: 59.1 (9.8)Pioglitazone arm: 18 (53%)Ipragliflozin 50 mg daily (n = 32)Pioglitazone 15-30 mg daily (n = 34)24 wkChange in L/S attenuation ratio
Ipragliflozin arm: 57.3 (12.1)Ipragliflozin arm: 14 (44%)
3Shibuya et al[13], 2018Fatty liver diagnosed on the basis of computed tomography or abdominal sonography, HbA1c 6.0%–10.0%, age 20–70 yrLuseogliflozin arm: 51 (47-62)Luseogliflozin arm: 10 (62.5%)Luseogliflozin 2.5 mg daily (n = 16)Metformin 1.5 g daily (n = 16)24 wkChange in L/S attenuation ratio
Metformin arm: 60 (53-66)Metformin arm: 8 (50%)
4Eriksson et al[14], 2018Age 40–75 yr, treated with a stable dose of metformin or sulfonylurea alone or in combination for at least 3 mo, MRI-PDFF > 5.5%, BMI 25–40 kg/m2Dapagliflozin arm: 65 (6.5)Dapagliflozin arm: 16 (76.2%)Dapagliflozin 10 mg daily (n = 21) or Omega 3-carboxylic acid 4 g daily (n = 20) or Combination (n = 22)Placebo (n = 21)12 wkChange in liver fat content by MRI-PDFF
Omega 3-carboxylic acid arm: 66.2 (5.9)Omega 3-carboxylic acid arm: 11 (55%)
O + D arm: 65(5.4)O + D arm: 15 (68.2%)
Placebo arm: 65.6 (6.1)Placebo arm: 17 (81%)
Table 3 Observational studies.
S. NoRef.DesignInclusion criteriaAge (yr)Male genderSample sizeSGLT-2 inhibitorFollow-up duration
1Ohki et al[15], 2016Prospective studyType 2 diabetes with NAFLD treated with GLP-1 analogues or DPP-4 inhibitors and failed to normalise serum ALT levels54.2 (49.3-60.1)19 (79.2%)24Ipragliflozin 25-50 mg daily320 d (302-329)
2Seko et al[16], 2016Retrospective cohort studyType 2 diabetes with NAFLDSGLT-2 inhibitor arm: 60.3 (1.8)SGLT-2 inhibitor arm: 9 (37.5%)24 (SGLT-2 inhibitor); 21 (Sitagliptin )Canagliflozin 100 mg (n = 18) or Ipragliflozin 50 mg daily (n = 6)24 wk
Sitagliptin arm: 59.4 (3.7)Sitagliptin arm: 8 (38.1%)
3Gautam et al[17], 2018Prospective studyType 2 diabetes with NAFLD--32Canagliflozin 100 mg daily24 wk
4Sumida et al[18], 2018Prospective studyAge > 20 yr, HbA1c > 6.5% to < 8.5%, NAFLD55.4 (13.6)28 (70%)40Luseogliflozin 2.5 mg daily24 wk
Risk of bias within studies

The risk of bias of RCTs was assessed using the Cochrane risk of bias tool. Among the four RCTs, the studies done by Kuchay et al[11] and Eriksson et al[14] were of good quality however those done by Ito et al[12] and Shibuya et al[13] were of fair quality (Table 4). The risk of bias of observational studies was assessed using the MINORS scale. All the observational studies were of less than ideal quality (Table 5).

Table 4 Assessment of study quality of randomised controlled trials.
StudyCriteriaRisk of biasStudy quality
Kuchay et al[11]Random sequence generationLow riskGood quality
Allocation concealmentLow risk
Selective reportingLow risk
Other biasLow risk
Blinding of participants and personnelLow risk
Blinding of outcome assessmentLow risk
Incomplete outcome dataLow risk
Ito et al[12]Random sequence generationLow riskFair quality
Allocation concealmentUnclear risk
Selective reportingLow risk
Other biasLow risk
Blinding of participants and personnelLow risk
Blinding of outcome assessmentLow risk
Incomplete outcome dataLow risk
Shibuya et al[13]Random sequence generationUnclear riskFair quality
Allocation concealmentUnclear risk
Selective reportingLow risk
Other biasLow risk
Blinding of participants and personnelLow risk
Blinding of outcome assessmentLow risk
Incomplete outcome dataLow risk
Eriksson et al[14]Random sequence generationLow riskGood quality
Allocation concealmentLow risk
Selective reportingLow risk
Other biasLow risk
Blinding of participants and personnelLow risk
Blinding of outcome assessmentLow risk
Incomplete outcome dataLow risk
Table 5 Assessment of study quality of observational studies.
S. NoCriteriaOhki et al[15]Seko et al[16]Gautam et al[17]Sumida et al[18]
1A clearly stated aim2222
2Inclusion of consecutive patients0221
3Prospective collection of data2022
4Endpoints appropriate to the aim of the study2222
5Unbiased assessment of the study endpoint0000
6Follow-up period appropriate to the aim of the study2222
7Loss to follow up less than 5%2222
8Prospective calculation of the study size0000
9An adequate control groupNA0NANA
10Contemporary groupsNA2NANA
11Baseline equivalence of groupsNA2NANA
12Adequate statistical analysesNA2NANA
13Total score10/1616/2412/1611/16
Primary outcome

Change in serum ALT levels: In all of the studies, there was a decrease in serum ALT levels from the baseline in those treated with SGLT-2 inhibitors (Table 6) but in the study done by Shibuya et al[13] it did not reach statistical significance.

Table 6 Change in serum alanine aminotransferase levels in individual studies.
StudySerum ALT level (IU/L)
P valueP value between groups
GroupBaselineStudy completion
Kuchay et al[11]Empagliflozin64.3 (20.2)49.7 (25.8)0.0010.005
Control65.3 (40.3)61.6 (38.4)0.422
Ito et al[12]Ipragliflozin57.4 (27.3)38.2 (20.5)< 0.050.642
Pioglitazone53.1 (26.6)36.8 (15.1)< 0.05
Shibuya et al[13]Luseogliflozin49.5 (31.0, 70.0)31 (26.0, 55.0)0.0570.064
Metformin39 (23.0, 56.0)39 (27.0, 51.0)0.518
Eriksson et al[14]Placebo33.53 (12.4)-0.2 (8.8)1--
Omega-3 CA37.65 (14.7)+5.9 (16.5)1-Non-significant2
Dapagliflozin39.41 (14.7)-8.2 (8.2)1-< 0.052
O + D35.88 (17.1)+0.1 (12.9)1-Non-significant2
Ohki et al[15]Ipragliflozin62 (43.0-75.0)38.0 (31.0-65.0)0.01-
Seko et al[16]SGLT-2 inhibitor70.8 (8.1)48.8 (5.5)0.0020.039
Sitagliptin92.4 (11.2)71.1 (10.0)0.012
Gautam et al[17]Canagliflozin96 (18.7)60.0 (17.6)< 0.00001-
Sumida et al[18]Luseogliflozin54.7 (28.2)42.4 (26.5)< 0.001-

Kuchay et al[11] found a significant decrease in serum ALT levels in the empagliflozin arm compared to the control arm at the end of the study (difference between the two arms was -10.9 IU/L, P = 0.005). In the study done by Ito et al[12] ALT levels decreased equally in both the groups [Change from baseline in ipragliflozin group: -17.5 (4) and pioglitazone group: -20 (3.4), P = 0.642]. Similar results were found in the study by Shibuya et al[13] [ΔALT in luseogliflozin arm was 9 (-20, 1) and in metformin arm was 4.5 (-5, 9), P = 0.064]. Eriksson et al[14] found that the ALT reduction in the dapagliflozin arm was more compared to placebo [ΔALT in dapagliflozin arm was -8.24 (8.24) and in the placebo arm was -0.18 (8.82), P < 0.05]. Seko et al[16] demonstrated that the serum ALT levels in SGLT-2 inhibitor arm was lower compared to the sitagliptin arm at the end of the study [48.8 (5.5) vs 71.1 (10), P = 0.039]

Secondary outcomes

Change in serum AST levels: Seven of the included studies had data regarding change in serum AST levels (Table 7). The study done by Shibuya et al[13] did not have data on AST levels. All the studies showed a significant reduction in serum AST levels in those treated with SGLT-2 inhibitors. The decrease in AST with empagliflozin and ipragliflozin was similar compared to placebo and pioglitazone respectively whereas dapagliflozin was better than placebo.

Table 7 Change in serum aspartate aminotransferase levels in individual studies.
StudySerum AST levels (IU/L)
P valueP value between groups
GroupBaselineStudy completion
Kuchay et al[11]Empagliflozin44.6 (23.5)36.2 (9.0)0.040.212
Control45.3 (24.3)44.6 (23.8)0.931
Ito et al[12]Ipragliflozin39.7 (16.7)27.3 (8.9)< 0.050.802
Pioglitazone43.3 (20.5)32.4 (15.4)< 0.05
Eriksson et al[14]Placebo29.4 (13.2)-1.2 (7.2)1--
Omega-3 CA30.6 (10.2)+4.8 (9.0)1-Non-significant2
Dapagliflozin31.2 (11.4)-4.2 (5.4)1-< 0.052
O + D30 (10.2)+1.2 (5.4)1-Non-significant2
Ohki et al[15]Ipragliflozin37 (29.0-52.0)28 (23.0-31.0)0.03-
Seko et al[16]SGLT-2 inhibitor54.4 (5.6)38 (3.1)0.001-
Sitagliptin67 (7.7)52.5 (7.7)0.016
Gautam et al[17]Canagliflozin72 (16.7)53 (10.3)< 0.00001-
Sumida et al[18]Luseogliflozin40.7 (22.2)31.9 (18.2)< 0.001-

Change in serum GGT levels: Seven studies had data regarding GGT levels. Six studies reported a significant decrease in serum GGT levels in those treated with SGLT-2 inhibitors (Table 8). In the study done by Seko et al[16], there was an insignificant decrease in both the SGLT-2 inhibitor and DPP-4 inhibitor groups. The decrease in GGT with empagliflozin and ipragliflozin was similar compared to placebo and pioglitazone correspondingly while dapagliflozin was better than placebo.

Table 8 Change in serum gamma-glutamyl transferase levels in individual studies.
StudySerum GGT (IU/L )
P valueP value between groups
GroupBaselineStudy completion
Kuchay et al[11]Empagliflozin65.8 (36.1)50.9 (24.6)0.0020.057
Control63.9 (45.3)60.0 (39.0)0.421
Ito et al[12]Ipragliflozin62.8 (58.3)44.0 (38.3)< 0.050.642
Pioglitazone71.6 (54.1)48.8 (61.2)< 0.05
Eriksson et al[14]Placebo32.4 (17.4)+2.4 (9.6)1--
Omega-3 CA54.0 (57.6)+2.4 (12.0)1-Non-significant2
Dapagliflozin58.2 (43.2)-4.8 (13.8)1-< 0.052
O + D40.2 (14.4)-0.6 (13.8)1-Non-significant2
Ohki et al[15]Ipragliflozin75.0 (47.0-105.0)60.0 (40.0-101.0)0.03-
Seko et al[16]SGLT-2 inhibitor61.7 (9.1)58.7 (11.5)0.051-
Sitagliptin89.2 (11.8)82.4 (11.9)0.36
Gautam et al[17]Canagliflozin75.1 (31.8)69.2 (26.2)0.003-
Sumida et al[18]Luseogliflozin62.4 (77.1)48.2 (56.3)0.003-

Change in hepatic fat: Kuchay et al[11] and Eriksson et al[14] evaluated hepatic fat using magnetic resonance imaging- derived proton density fat fraction (Table 9). It was found that there was a significant reduction in hepatic fat in the empagliflozin arm compared to the control arm in the study done by Kuchay et al[11]. In the study done by Eriksson et al[14], dapagliflozin or omega-3 carboxylic acid when administered alone or in combination reduced hepatic fat fraction significantly. When compared with placebo, only the combination of both drugs reduced hepatic fat fraction significantly. Sumida et al[18] showed that luseogliflozin significantly reduced hepatic fat fraction using magnetic resonance imaging-hepatic fat fraction. Ito et al[12] and Shibuya et al[13] used liver/spleen attenuation ratio for measuring hepatic fat. They found that ipragliflozin was equivalent to pioglitazone in improving liver/spleen attenuation ratio while luseogliflozin was found to be superior to metformin in the same aspect.

Table 9 Change in hepatic fat in individual studies.
StudyParameterGroupBaselineStudy completionP valueP value between groups
Kuchay et al[11]MRI-PDFFEmpagliflozin16.2 (7)11.3 (5.3)< 0.0001< 0.0001
Control16.4 (7.3)15.5 (6.7)0.054
Ito et al[12]L/S ratioIpragliflozin0.8 (0.2)1.0 (0.2)< 0.050.90
Pioglitazone0.8 (0.3)1.0 (0.2)< 0.05
Shibuya et al[13]L/S ratioLuseogliflozin0.9 (0.6-1.0)1.0 (0.8-1.2)0.00080.00002
Metformin1.0 (0.8-1.1)0.9 (0.7-1.0)0.017
Eriksson et al[14]MRI-PDFFPlacebo15.1 (6.5)-0.6 (1.9)1--
Omega-3 CA22.2 (11.0)-3.2 (2.9)1-Non-significant2
Dapagliflozin17.3 (9.1)-2.2 (3.3)1-Non-significant2
O + D17.8 (9.2)-3.2 (3.5)1-< 0.052
Sumida et al[18]MRI-HFFLuseogliflozin21.5 (7.2)15.7 (6.8)< 0.001-
Effect on liver fibrosis indices

Ito et al[12] and Ohki et al[15] evaluated liver fibrosis using the FIB-4 index (Table 10). There was a significant decrease in the FIB-4 index in the ipragliflozin arms compared to baseline. Ipragliflozin was similar to pioglitazone in decreasing the FIB-4 index. Sumida et al[18] used both the FIB-4 index and NAFLD fibrosis score. There was no significant change in either indices.

Table 10 Assessment of liver fibrosis in individual studies.
StudyParameterGroupBaselineStudy completionP valueP value between groups
Ito et al[12]FIB-4 indexIpragliflozin1.44 (0.64)1.22 (0.55)< 0.050.596
Pioglitazone1.84 (1.13)1.71 (1.19)Non-significant
Ohki et al[15]FIB-4 indexIpragliflozin1.75 (0.82-1.93)1.39 (0.77-1.99)0.04-
Sumida et al[18]FIB-4 indexLuseogliflozin1.63 (1.19)1.52 (0.92)0.17-
NAFLD fibrosis scoreLuseogliflozin1.61 (0.71)1.62 (0.88)0.86-
Change in metabolic and anthropometric parameters

Seven studies reported changes in FPG and HbA1c (Tables 11 and 12). The majority of the studies showed a decrease in FPG and HbA1c.

Table 11 Change in fasting plasma glucose in individual studies.
StudyFasting plasma glucose (mg/dL)
P valueP value between groups
GroupBaselineStudy completion
Kuchay et al[11]Empagliflozin173.0 (44.0)124.0 (17.0)< 0.0010.85
Control176.0 (57.0)120.0 (19.0)< 0.0001
Ito et al[12]Ipragliflozin160.1 (38.7)136.5 (26.7)< 0.050.785
Pioglitazone169.4 (50.9)139.0 (26.6)< 0.05
Shibuya et al[13]Luseogliflozin127.0 (116.0, 136.0)125.0 (113.0, 138.0)0.870.583
Metformin147.0 (126.0, 161.0)134.0 (122.0, 145.0)0.32
Eriksson et al[14]Placebo169.2 (29.7)+6.7 (14.8)1--
Omega-3 CA162.4 (26.6)+3.8 (19.3)1-Non-significant2
Dapagliflozin161.8 (33.3)-17.6 (26.8)1-< 0.052
O + D168.8 (35.5)-16.4 (36.0)1-< 0.052
Ohki et al[15]Ipragliflozin162.0 (135.0-189.0)135.0 (120.0-166.0)0.3-
Seko et al[16]SGLT-2 inhibitor125.0 (6.0)116.6 (4.2)0.07Non-significant
Sitagliptin114.6 (7.0)134.0 (10.5)0.067
Sumida et al[18]Luseogliflozin142.0 (30.3)135.4 (25.6)0.04-
Table 12 Change in glycosylated haemoglobin in individual studies.
StudyGlycosylated haemoglobin (%)
P valueP value between groups
GroupBaselineStudy completion
Kuchay et al[11]Empagliflozin9.0 (1.0)7.2 (0.6)< 0.0010.88
Control9.1 (1.4)7.1 (0.9)< 0.0001
Ito et al[12]Ipragliflozin8.5 (1.5)7.6 (1.0)< 0.050.522
Pioglitazone8.3 (1.4)7.1 (0.9)< 0.05
Shibuya et al[13]Luseogliflozin7.8 (7.2, 7.9)6.5 (6.4, 7.0)0.0020.023
Metformin7.4 (6.9, 7.7)7.3 (6.7, 7.6)0.362
Eriksson et al[14]Placebo7.4 (0.8)-0.1 (0.4)1--
Omega-3 CA7.4 (0.7)+0.1 (0.4)1-Non-significant2
Dapagliflozin7.4 (0.6)-0.6 (0.7)1-< 0.052
O + D7.5 (0.8)-0.5 (0.5)1-Non-significant2
Ohki et al[15]Ipragliflozin8.4 (7.8-8.9)7.6 (6.9-8.2)< 0.01-
Seko et al[16]SGLT-2 inhibitor6.7 (0.1)6.5 (0.1)0.055Non-significant
Sitagliptin7.0 (0.3)6.9 (0.3)0.331
Sumida et al[18]Luseogliflozin7.3 (0.7)7.0 (0.7)0.002-

In the study done by Ito et al[12] there was no difference in the change in HOMA-IR in those treated with either ipragliflozin or pioglitazone (P = 0.401) (Table 13). There was a significant decrease in HOMA-IR in those treated with dapagliflozin compared to placebo in the study done by Eriksson et al[14]. Surprisingly there was an insignificant increase in HOMA-IR in those treated with either a SGLT-2 inhibitor or a gliptin in the study done by Seko et al[16].

Table 13 Change in homeostasis model assessment-estimated insulin resistance in individual studies.
StudyHOMA-IR
P valueP value between groups
GroupBaselineStudy completion
Ito et al[12]Ipragliflozin5.2 (2.5)4.8 (5.5)Non-significant0.401
Pioglitazone5.7 (3.4)4.5 (2.7)< 0.05
Eriksson et al[14]Placebo4.2 (2.4)-0.2 (1.4)1--
Omega 3-CA5.4 (2.9)+0.3 (2.4)1-Non-significant2
Dapagliflozin4.3 (1.9)-1.1 (1.4)1-< 0.052
O + D4.4 (1.7)-0.9 (1.6)1-< 0.052
Seko et al[16]SGLT-2 inhibitor4.5 (0.5)7.9 (2.3)0.955-
Sitagliptin4.4 (0.5)6.5 (0.8)0.163

Six studies included data on the changes in lipid profile (Tables 14, 15, and 16). There was a significant decrease in serum triglycerides in two studies (Kuchay et al[11] and Ito et al[12]). Three studies exhibited an increase in high-density lipoprotein cholesterol levels (Ito et al[12], Ohki et al[15], and Seko et al[16]). Most of the studies (Ito et al[12], Eriksson et al[14], Ohki et al[15], Seko et al[16], and Sumida et al[18]) showed no change in serum LDL levels.

Table 14 Change in serum triglycerides in individual studies.
StudySerum triglycerides (mg/dL)
P valueP value between groups
GroupBaselineStudy completion
Kuchay et al[11]Empagliflozin201.0 (124.0)155.0 (52.0)0.010.678
Control212.0 (115.0)175.0 (43.0)0.019
Ito et al[12]Ipragliflozin166.9 (76.4)143.4 (81.4)< 0.050.938
Pioglitazone188.4 (148.8)169.3 (131.3)Non-significant
Eriksson et al[14]Placebo169.2 (84.1)-11.5 (45.6)1--
Omega-3 CA186.9 (81.5)-15.9 (47.4)1-Non-significant2
Dapagliflozin178.0 (103.6)+14.2 (40.5)1-Non-significant2
O + D168.3 (72.6)-25.7 (57.1)1-Non-significant2
Ohki et al[15]Ipragliflozin148.0 (107.0, 222.)145.0 (114.0, 172.0)0.75-
Seko et al[16]SGLT-2 inhibitor153.8 (15.9)137.8 (10.5)0.236-
Sitagliptin193.4 (25.2)191.1 (23.8)0.986
Sumida et al[18]Luseogliflozin158.1 (110.5)129.4 (59.5)0.062-
Table 15 Change in serum low-density lipoprotein cholesterol in individual studies.
StudySerum low-density lipoprotein cholesterol (mg/dL)
P valueP value between groups
GroupBaselineStudy completion
Kuchay et al[11]Empagliflozin112.0 (35.0)95.0 (22.0)0.0180.512
Control114.0 (30.0)96.0 (17.0)0.001
Ito et al[12]Ipragliflozin108.3 (36.2)110.7 (40.1)Non-significant0.057
Pioglitazone104.0 (27.9)114.6 (29.5)< 0.05
Eriksson et al[14]Placebo98.2 (34.4)+1.6 (15.5)1--
Omega-3 CA111.8 (34.4)+2.3 (17.4)1-Non-significant2
Dapagliflozin109.4 (34.8)+7.7 (20.5)1-Non-significant2
O + D88.9 (23.2)+5.8 (21.7)1-Non-significant2
Ohki et al[15]Ipragliflozin113.0 (89.0-142.0)103.0 (92.0-122.0)0.08-
Seko et al[16]SGLT-2 inhibitor119.2 (5.8)119.8 (5.7)0.943-
Sitagliptin112.9 (4.9)127.1 (8.8)0.063
Sumida et al[18]Luseogliflozin101.0 (22.4)105.0 (24.4)0.11-
Table 16 Change in serum high-density lipoprotein cholesterol in individual studies.
StudySerum high-density lipoprotein cholesterol (mg/dL)
P valueP value between groups
GroupBaselineStudy completion
Kuchay et al[11]Empagliflozin42.0 (12.0)45.0 (12.0)0.0870.752
Control45.0 (15.0)47.0 (12.0)0.097
Ito et al[12]Ipragliflozin48.9 (9.3)54.7 (10.4)< 0.050.82
Pioglitazone47.4 (11.6)52.7 (13.5)< 0.05
Eriksson et al[14]Placebo51.4 (14.9)-0.4 (5.0)1--
Omega-3 CA49.9 (14.1)+0.4 (3.2)1-Non-significant2
Dapagliflozin49.9 (9.5)+0.4 (4.8)1-Non-significant2
O + D51.4 (10.2)+1.6 (5.0)1-Non-significant2
Ohki et al[15]Ipragliflozin42.0 (40.0-50.0)44.0 (42.0-59.0)0.01-
Seko et al[16]SGLT-2 inhibitor53.9 (2.5)55.4 (2.6)0.043-
Sitagliptin54.8 (3.3)55.6 (2.3)0.531
Sumida et al[18]Luseogliflozin55.6 (11.7)57.5 (13.4)0.062-

Five studies included BMI change (Table 17). There was a reduction in BMI in the SGLT-2 inhibitor arms in all the studies. Empagliflozin was similar to placebo in reducing BMI whereas luseogliflozin was superior to metformin in reducing BMI.

Table 17 Change in body mass index in individual studies.
StudyBody mass index (kg/m2)
P valueP value between groups
GroupBaselineStudy completion
Kuchay et al[11]Empagliflozin30.0 (3.8)28.7 (3.5)0.0010.124
Control29.4 (3.1)28.8 (2.8)0.019
Shibuya et al[13]Luseogliflozin27.9 (26.2, 28.7)27.0 (25.6, 28.3)0.0020.031
Metformin27.2 (24.8, 32.1)27.3 (24.3, 31.6)0.646
Ohki et al[15]Ipragliflozin30.1 (26.1-31.4)27.6 (25.3-30.2)< 0.01-
Seko et al[16]SGLT-2 inhibitor29.6 (0.7)28.3 (0.7)< 0.001-
Sitagliptin29.2 (1.5)28.9 (1.4)0.295
Sumida et al[18]Luseogliflozin27.8 (3.6)27.2 (1.0)< 0.001-
Adverse effects of SGLT-2 inhibitors

Out of the eight studies, six studies reported the adverse effects of SGLT-2 inhibitors. There were a total of 30 reported adverse events in 176 patients taking SGLT-2 inhibitors (Table 18). The most common adverse event was genitourinary tract infection (10 events).

Table 18 Adverse effects of sodium glucose cotransporter-2 inhibitors in individual studies.
StudyNo. of adverse eventsNo. of patientsTypes of adverse events
Kuchay et al[11]325Nonspecific fatigue: 1
Arthralgia: 1
Balanoposthitis: 1
Ito et al[12]932UTI: 3
Increased appetite: 2
Nausea: 1
Headache: 1
Diarrhoea: 1
Vaginal candidiasis: 1
Eriksson et al[14]721-
Seko et al[16]226UTI: 2
Gautam et al[17]132Recurrent UTI with genital candidiasis: 1
Sumida et al[18]840Low blood pressure: 3
Vaginal itching: 2
Constipation: 1
Vertigo: 1
Dehydration: 1
Total30176Most common adverse event: Genitourinary tract infections-10
DISCUSSION

Type 2 diabetes is commonly associated with NAFLD. Serum ALT levels are commonly above the upper limit of normal with AST levels lesser than ALT levels[19]. Animal studies have shown that SGLT-2 inhibitors decrease liver enzymes (ALT, AST), liver weight, and hepatic steatosis[20-23]. There are several mechanisms for improvement in serum liver enzymes in the patients taking SGLT-2 inhibitors. These drugs cause hyperglucagonemia by increasing glucagon secretion from the pancreatic α cells. Glucagon stimulates gluconeogenesis and β-oxidation of fatty acids in the liver via stimulation of peroxisome proliferator-activated receptor alpha and carnitine palmitoyl transferase-1[13]. Thus SGLT-2 inhibitors help to reduce hepatic fat. They reduce collagen deposition and inflammatory cytokine expression in liver[5,22]. They decrease liver enzymes by additionally improving glycaemic parameters and insulin resistance. Out of eight studies, seven showed a decrease in serum ALT and AST levels in our systematic review. Shibuya et al[13] observed a decrease in ALT that almost reached statistical significance, however data regarding AST was unavailable[13]. Out of seven studies, six illustrated a significant decrease in GGT levels while in the study by Seko et al[16] the change in serum GGT level almost reached statistical significance.

Liver enzymes are surrogate markers of liver histological response, but an improvement in liver histology is not always associated with a decrease in serum liver enzymes[11]. The five studies that evaluated changes in hepatic fat showed a decrease in hepatic fat. There was no correlation of a change in ALT with a change in hepatic fat in the study by Shibuya et al[13], however there was a correlation between these two parameters in the study by Sumida et al[18]. The decrease in hepatic fat in the SGLT-2 inhibitor arm was comparable to pioglitazone, which is an approved drug for treatment of NAFLD irrespective of the presence of diabetes. Eriksson et al[14] observed that although the hepatic fat content decreased in the dapagliflozin arm it did not reach statistical significance compared to placebo. The lesser duration of this study (12 wk) compared to other studies may have contributed to this difference.

The progression of NAFLD to cirrhosis is determined to a large extent by the liver histology. Studies with up to 20 years follow-up have shown that the risk of progression to cirrhosis for simple steatosis, NASH, and NASH with fibrosis are 0%-4%, 25%, and 38%, respectively[24]. The FIB-4 index is a non-invasive tool to assess liver fibrosis[25]. It is calculated from the patient’s age, platelet count, ALT levels, and AST levels. The FIB-4 index was decreased with SGLT-2 inhibitor therapy in two out of three studies. Sumida et al[18] used the NAFLD fibrosis score in addition to the FIB-4 index to assess liver fibrosis. The NAFLD fibrosis score is a composite score of six variables (age, BMI, hyperglycaemia, platelet count, albumin, and AST/ALT ratio)[26]. There was no significant change in either indices in this study.

It has been shown that NAFLD is more common in those with poor glycaemic control than those with good glycaemic control[27]. SGLT-2 inhibitors promote glycosuria by inhibiting SGLT-2 in the proximal convoluted tubule. Therefore their action is dependent on blood glucose levels but insulin independent[28]. They cause a significant reduction in FPG[29]. A meta-analysis of RCTs has concluded that the average HbA1c reduction at 52 wk of SGLT-2 inhibitor therapy to be 0.6%[30]. Another meta-analysis has shown that SGLT-2 inhibitor monotherapy is equivalent to metformin monotherapy in reducing HbA1c levels[31]. However, the decrease in HbA1c was more in the luseogliflozin arm compared to the metformin arm in the study by Shibuya et al[13]. Four out of seven studies and six out of seven studies showed a decrease in FPG and HbA1c, respectively, in the SGLT-2 inhibitor arm. Thus, the improved glycaemic status is one of the mechanisms by which SGLT-2 inhibitors ameliorate NAFLD.

SGLT-2 inhibitors ameliorate insulin resistance in numerous ways. SGLT-2 inhibitors improve obesity associated insulin resistance by regulating macrophage recruitment and altering the proportion of pro-inflammatory and anti-inflammatory macrophages. They enhance fat utilization by promoting β-oxidation of fatty acids and browning of white adipose tissue by inducing the expression of thermogenin leading to an improvement in the lipid profile. Similar to other antidiabetic drugs, SGLT-2 inhibitors reduce insulin resistance by decreasing glucotoxicity. Dapagliflozin has been shown to improve insulin sensitivity by increasing adiponectin and zinc-A2-glycoprotein levels[32]. Only dapagliflozin was shown to decrease insulin resistance in the study by Eriksson et al[14].

SGLT-2 inhibitors cause weight reduction. The major mechanism that causes weight reduction is the decrease in fat mass. The decrease in fat mass is due to the shift in substrate utilization to lipids instead of carbohydrates[33,34]. Ito et al[12] and Shibuya et al[13] demonstrated that SGLT-2 inhibitors caused a significant reduction in abdominal visceral and subcutaneous fat area as measured by computed tomography scan. Similarly, Eriksson et al[14] showed that dapagliflozin significantly reduced abdominal visceral and subcutaneous adipose tissue volume as assessed by magnetic resonance imaging. The other mechanisms of weight loss are the urinary glucose loss which amounts to approximately 200 Kcal/d and osmotic diuresis[33,35]. Unlike the other weight-reducing effects of SGLT-2 inhibitors, which are potentially beneficial, osmotic diuresis is clearly an adverse effect. Seko et al[16] showed that ipragliflozin and canagliflozin significantly reduced total body water in addition to body fat mass as measured by bioelectrical impedance analysis. Five studies showed a significant decrease in BMI in patients on SGLT-2 inhibitor therapy. Thus, the major beneficial effects of SGLT-2 inhibitors on NAFLD are exerted via reduction in hepatic fat and fibrosis, improved glycaemic control, decrease in insulin resistance, and weight loss.

The most common adverse effects of SGLT-2 inhibitors are genitourinary tract infections. In addition, they may cause diabetic ketoacidosis, dizziness, acute kidney injury, lower limb amputations, and bone fractures[36,37]. A meta-analysis concluded that there was no difference between placebo and SGLT-2 inhibitors for serious adverse events[38]. Among the 30 adverse events reported in all the studies, the most common was genitourinary tract infections (10 out of 23 characterised events).

The major strength of this systematic review was that the effect of five SGLT-2 inhibitors on NAFLD in patients with type 2 diabetes was evaluated in both RCTs and observational studies. Moreover, liver fat, liver fibrosis, metabolic, and anthropometric parameters in addition to liver enzymes were assessed as outcome variables following SGLT-2 inhibitor therapy. Yet this systematic review has a few limitations. First, most of the studies were done amongst the Japanese population. As a result, the study findings may not be applicable to patients from other ethnicities. Second, the sample size was considerably small and the duration of follow-up was of limited period in most of the studies. Third, the confounding effect of concomitant anti-diabetes drugs like metformin, DPP- 4 inhibitors, and glucagon like peptide-1 analogues on NAFLD cannot be ruled out, particularly in observational studies. Fourth, two studies (Eriksson et al[14] and Sumida et al[18]) were funded by pharmaceutical companies, which is a source of potential conflicts of interest.

Summary and conclusion

In conclusion based on the available evidence, SGLT-2 inhibitors were found to improve serum levels of liver enzymes, liver fibrosis indices, and liver fat without significant side effects in type 2 diabetes patients with NAFLD. They showed additional beneficial effects on obesity, glycaemic parameters, insulin resistance, and dyslipidaemia in these subjects. However, the quality of evidence was low to moderate. Prospective studies, preferably RCTs, comparing different SGLT-2 inhibitors with standard treatments of NAFLD in multi-ethnic populations with a longer follow-up period are needed in the future.

ARTICLE HIGHLIGHTS
Research background

Non-alcoholic fatty liver disease (NAFLD) is a common comorbidity with type 2 diabetes. The existing therapeutic options for NAFLD are not adequate. Hypocaloric diet and exercise is the cornerstone of therapy in NAFLD. Pioglitazone is the only drug recommended in diabetes patients with biopsy proven non-alcoholic steatohepatitis. The frequent coexistence of NAFLD and type 2 diabetes along with their combined adverse health consequences and inadequate therapeutic options makes it necessary to search for newer alternatives. This systematic review is an effort to review the available literature on the effect of sodium glucose cotransporter-2 (SGLT-2) inhibitors on NAFLD in type 2 diabetes patients.

Research motivation

Because the existing therapeutic options are not adequate for NAFLD patients, there is a need for finding newer alternatives. SGLT-2 inhibitors have shown promise in the management of NAFLD in animals. Hence, we reviewed the available literature on the effect of SGLT-2 inhibitors in NAFLD in type 2 diabetes patients. This will promote further high quality research on the effect of SGLT-2 inhibitors in NAFLD.

Research objectives

The primary outcome was the change in serum alanine aminotransferase levels in type 2 diabetes patients with NAFLD treated with SGLT-2 inhibitors. The secondary outcomes were change in serum aspartate aminotransferase and gamma-glutamyl transferase levels, hepatic fat, hepatic fibrosis, metabolic profile, anthropometric parameters, and the adverse effects of SGLT-2 inhibitors.

Research methods

This systematic review was registered in PROSPERO and performed according to Preferred Reporting Items for Systematic Reviews and Meta-Analysis guidelines. We searched PubMed/MEDLINE, Cochrane library, Google scholar, and Clinicaltrials.gov for the relevant articles to be included in this systematic review. A narrative synthesis of the results of individual studies was done. The change in the difference in means and difference in proportions and the respective P values as mentioned in the original manuscripts were tabulated and explained. The quality of the randomised controlled trials and observational studies was analysed using the Cochrane risk of bias tool and MINORS scale, respectively.

Research results

Eight articles (four randomised controlled trials and four observational studies) were included in this systematic review. A total of 214 patients were treated with SGLT-2 inhibitors. SGLT-2 inhibitors caused a significant improvement in liver enzymes, hepatic fat, hepatic fibrosis, glycaemia, insulin resistance, obesity, and lipid parameters with minimal adverse effects. However, the quality of evidence is low to moderate.

Research conclusions

We found that SGLT-2 inhibitors improved the serum levels of liver enzymes, liver fat, and liver fibrosis with additional beneficial effects on various metabolic and anthropometric parameters in type 2 diabetes patients with NAFLD. However, the number of patients treated with SGLT-2 inhibitors was small. The findings of this systematic review will have impact in choosing anti-diabetes medication like SGLT-2 inhibitors to treat NAFLD associated with type 2 diabetes.

Research perspectives

The studies included in this systematic review were heterogeneous with regard to study design and intervention drugs. Most of the studies were done amongst the Japanese population. Prospective studies, preferably randomised controlled trials, comparing different SGLT-2 inhibitors with standard treatments of NAFLD in multi-ethnic populations with a longer follow-up period are needed in the future.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Endocrinology and metabolism

Country of origin: India

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C, C, C

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Joseph PM, Serhiyenko VA, Tzamaloukas AHH S- Editor: Ma YJ L- Editor: Filipodia E- Editor: Song H

References
1.  Saponaro C, Gaggini M, Gastaldelli A. Nonalcoholic fatty liver disease and type 2 diabetes: common pathophysiologic mechanisms. Curr Diab Rep. 2015;15:607.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Burt AD, Lackner C, Tiniakos DG. Diagnosis and Assessment of NAFLD: Definitions and Histopathological Classification. Semin Liver Dis. 2015;35:207-220.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Williams KH, Shackel NA, Gorrell MD, McLennan SV, Twigg SM. Diabetes and nonalcoholic Fatty liver disease: a pathogenic duo. Endocr Rev. 2013;34:84-129.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella M, Harrison SA, Brunt EM, Sanyal AJ. The diagnosis and management of nonalcoholic fatty liver disease: Practice guidance from the American Association for the Study of Liver Diseases. Hepatology. 2018;67:328-357.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Qiang S, Nakatsu Y, Seno Y, Fujishiro M, Sakoda H, Kushiyama A, Mori K, Matsunaga Y, Yamamotoya T, Kamata H, Asano T. Treatment with the SGLT2 inhibitor luseogliflozin improves nonalcoholic steatohepatitis in a rodent model with diabetes mellitus. Diabetol Metab Syndr. 2015;7:104.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Tahara A, Kurosaki E, Yokono M, Yamajuku D, Kihara R, Hayashizaki Y, Takasu T, Imamura M, Li Q, Tomiyama H, Kobayashi Y, Noda A, Sasamata M, Shibasaki M. Effects of SGLT2 selective inhibitor ipragliflozin on hyperglycemia, hyperlipidemia, hepatic steatosis, oxidative stress, inflammation, and obesity in type 2 diabetic mice. Eur J Pharmacol. 2013;715:246-255.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Yokono M, Takasu T, Hayashizaki Y, Mitsuoka K, Kihara R, Muramatsu Y, Miyoshi S, Tahara A, Kurosaki E, Li Q, Tomiyama H, Sasamata M, Shibasaki M, Uchiyama Y. SGLT2 selective inhibitor ipragliflozin reduces body fat mass by increasing fatty acid oxidation in high-fat diet-induced obese rats. Eur J Pharmacol. 2014;727:66-74.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Moher D, Liberati A, Tetzlaff J, Altman DG; PRISMA Group. Preferred Reporting Items for Systematic Reviews and Meta-Analyses: The PRISMA Statement. PLoS Med. 2009;6:e1000097.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Higgins JP, Altman DG, Gøtzsche PC, Jüni P, Moher D, Oxman AD, Savovic J, Schulz KF, Weeks L, Sterne JA; Cochrane Bias Methods Group; Cochrane Statistical Methods Group. The Cochrane Collaboration's tool for assessing risk of bias in randomised trials. BMJ. 2011;343:d5928.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Slim K, Nini E, Forestier D, Kwiatkowski F, Panis Y, Chipponi J. Methodological index for non-randomized studies (minors): development and validation of a new instrument. ANZ J Surg. 2003;73:712-716.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Kuchay MS, Krishan S, Mishra SK, Farooqui KJ, Singh MK, Wasir JS, Bansal B, Kaur P, Jevalikar G, Gill HK, Choudhary NS, Mithal A. 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.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Ito D, Shimizu S, Inoue K, Saito D, Yanagisawa M, Inukai K, Akiyama Y, Morimoto Y, Noda M, Shimada A. Comparison of Ipragliflozin and Pioglitazone Effects on Nonalcoholic Fatty Liver Disease in Patients With Type 2 Diabetes: A Randomized, 24-Week, Open-Label, Active-Controlled Trial. Diabetes Care. 2017;40:1364-1372.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Shibuya T, Fushimi N, Kawai M, Yoshida Y, Hachiya H, Ito S, Kawai H, Ohashi N, Mori A. Luseogliflozin improves liver fat deposition compared to metformin in type 2 diabetes patients with non-alcoholic fatty liver disease: A prospective randomized controlled pilot study. Diabetes Obes Metab. 2018;20:438-442.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Eriksson JW, Lundkvist P, Jansson PA, Johansson L, Kvarnström M, Moris L, Miliotis T, Forsberg GB, Risérus U, Lind L, Oscarsson J. Effects of dapagliflozin and n-3 carboxylic acids on non-alcoholic fatty liver disease in people with type 2 diabetes: a double-blind randomised placebo-controlled study. Diabetologia. 2018;61:1923-1934.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Ohki T, Isogawa A, Toda N, Tagawa K. Effectiveness of Ipragliflozin, a Sodium-Glucose Co-transporter 2 Inhibitor, as a Second-line Treatment for Non-Alcoholic Fatty Liver Disease Patients with Type 2 Diabetes Mellitus Who Do Not Respond to Incretin-Based Therapies Including Glucagon-like Peptide-1 Analogs and Dipeptidyl Peptidase-4 Inhibitors. Clin Drug Investig. 2016;36:313-319.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Seko Y, Sumida Y, Tanaka S, Mori K, Taketani H, Ishiba H, Hara T, Okajima A, Umemura A, Nishikawa T, Yamaguchi K, Moriguchi M, Kanemasa K, Yasui K, Imai S, Shimada K, Itoh Y. Effect of sodium glucose cotransporter 2 inhibitor on liver function tests in Japanese patients with non-alcoholic fatty liver disease and type 2 diabetes mellitus. Hepatol Res. 2017;47:1072-1078.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Gautam A, Agrawal PK, Doneria J, Nigam A. Effects of Canagliflozin on Abnormal Liver Function Tests in Patients of Type 2 Diabetes with Non-Alcoholic Fatty Liver Disease. JAPI. 2018;66:62-66.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Sumida Y, Murotani K, Saito M, Tamasawa A, Osonoi Y, Yoneda M, Osonoi T. Effect of luseogliflozin on hepatic fat content in type 2 diabetes patients with non-alcoholic fatty liver disease: A prospective, single-arm trial (LEAD trial). Hepatol Res. 2019;49:64-71.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Sattar N, Fitchett D, Hantel S, George JT, 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® trial. Diabetologia. 2018;61:2155-2163.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Nakano S, Katsuno K, Isaji M, Nagasawa T, Buehrer B, Walker S, Wilkison WO, Cheatham B. Remogliflozin Etabonate Improves Fatty Liver Disease in Diet-Induced Obese Male Mice. J Clin Exp Hepatol. 2015;5:190-198.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Komiya C, Tsuchiya K, Shiba K, Miyachi Y, Furuke S, Shimazu N, Yamaguchi S, Kanno K, Ogawa Y. Ipragliflozin Improves Hepatic Steatosis in Obese Mice and Liver Dysfunction in Type 2 Diabetic Patients Irrespective of Body Weight Reduction. PLoS One. 2016;11:e0151511.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Jojima T, Tomotsune T, Iijima T, Akimoto K, Suzuki K, Aso Y. Empagliflozin (an SGLT2 inhibitor), alone or in combination with linagliptin (a DPP-4 inhibitor), prevents steatohepatitis in a novel mouse model of non-alcoholic steatohepatitis and diabetes. Diabetol Metab Syndr. 2016;8:45.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Wang D, Luo Y, Wang X, Orlicky DJ, Myakala K, Yang P, Levi M. The Sodium-Glucose Cotransporter 2 Inhibitor Dapagliflozin Prevents Renal and Liver Disease in Western Diet Induced Obesity Mice. Int J Mol Sci. 2018;19.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Calzadilla Bertot L, Adams LA. The Natural Course of Non-Alcoholic Fatty Liver Disease. Int J Mol Sci. 2016;17.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Shah AG, Lydecker A, Murray K, Tetri BN, Contos MJ, Sanyal AJ; Nash Clinical Research Network. Comparison of noninvasive markers of fibrosis in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol. 2009;7:1104-1112.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Angulo P, Hui JM, Marchesini G, Bugianesi E, George J, Farrell GC, Enders F, Saksena S, Burt AD, Bida JP, Lindor K, Sanderson SO, Lenzi M, Adams LA, Kench J, Therneau TM, Day CP. The NAFLD fibrosis score: a noninvasive system that identifies liver fibrosis in patients with NAFLD. Hepatology. 2007;45:846-854.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Afolabi BI, Ibitoye BO, Ikem RT, Omisore AD, Idowu BM, Soyoye DO. The Relationship Between Glycaemic Control and Non-Alcoholic Fatty Liver Disease in Nigerian Type 2 Diabetic Patients. J Natl Med Assoc. 2018;110:256-264.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Kalra S. Sodium Glucose Co-Transporter-2 (SGLT2) Inhibitors: A Review of Their Basic and Clinical Pharmacology. Diabetes Ther. 2014;5:355-366.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Abdul-Ghani MA, Norton L, Defronzo RA. Role of sodium-glucose cotransporter 2 (SGLT 2) inhibitors in the treatment of type 2 diabetes. Endocr Rev. 2011;32:515-531.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Monami M, Nardini C, Mannucci E. Efficacy and safety of sodium glucose co-transport-2 inhibitors in type 2 diabetes: a meta-analysis of randomized clinical trials. Diabetes Obes Metab. 2014;16:457-466.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Palmer SC, Mavridis D, Nicolucci A, Johnson DW, Tonelli M, Craig JC, Maggo J, Gray V, De Berardis G, Ruospo M, Natale P, Saglimbene V, Badve SV, Cho Y, Nadeau-Fredette AC, Burke M, Faruque L, Lloyd A, Ahmad N, Liu Y, Tiv S, Wiebe N, Strippoli GF. Comparison of Clinical Outcomes and Adverse Events Associated With Glucose-Lowering Drugs in Patients With Type 2 Diabetes: A Meta-analysis. JAMA. 2016;316:313-324.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Mohammad SH, Fadhil NN, Mahmood MD. Effects of metformin and dapagliflozin on glycemic indices and HOMA-IR in type 2 diabetes mellitus patients. Int J Pharm Biol Sci. 2018;8:66-73.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Trujillo JM, Nuffer WA. Impact of Sodium-Glucose Cotransporter 2 Inhibitors on Nonglycemic Outcomes in Patients with Type 2 Diabetes. Pharmacotherapy. 2017;37:481-491.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Ferrannini E, Baldi S, Frascerra S, Astiarraga B, Heise T, Bizzotto R, Mari A, Pieber TR, Muscelli E. Shift to Fatty Substrate Utilization in Response to Sodium-Glucose Cotransporter 2 Inhibition in Subjects Without Diabetes and Patients With Type 2 Diabetes. Diabetes. 2016;65:1190-1195.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Ferrannini G, Hach T, Crowe S, Sanghvi A, Hall KD, Ferrannini E. Energy Balance After Sodium-Glucose Cotransporter 2 Inhibition. Diabetes Care. 2015;38:1730-1735.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Esteban-Jiménez O, Navarro-Pemán C, Urieta-González L. Seguridad de los iSGLT-2. Revisión de las reacciones adversas notificadas a nivel nacional. Med Fam SEMERGEN. 2018;44:23–29.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Blau JE, Taylor SI. Adverse effects of SGLT2 inhibitors on bone health. Nat Rev Nephrol. 2018;14:473–474.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Storgaard H, Gluud LL, Bennett C, Grøndahl MF, Christensen MB, Knop FK, Vilsbøll T. Benefits and Harms of Sodium-Glucose Co-Transporter 2 Inhibitors in Patients with Type 2 Diabetes: A Systematic Review and Meta-Analysis. PLoS One. 2016;11:e0166125.  [PubMed]  [DOI]  [Cited in This Article: ]