Differential Effects of SGLT-2 Inhibitors on Liver Function and Noctur

Introduction

Sodium-glucose cotransporter-2 inhibitors (SGLT-2is) were initially introduced as treatments for type 2 diabetes but have since shown utility in various areas, such as preventing and improving renal and heart failure.1–4 Consequently, the use of SGLT-2is has expanded across multiple fields. Recently, studies have highlighted their efficacy in addressing fatty liver disease.5–7 Approximately 65% of patients with type 2 diabetes are estimated to have metabolic dysfunction-associated steatotic liver disease (MASLD).8,9 Moreover, the prevalence of metabolic dysfunction-associated steatohepatitis (MASH) in these patients can be as high as 66.44%,8 indicating that a considerable portion of MASLD in these patients corresponds to MASH. If left untreated, MASH can lead to progressive fibrosis, increasing the risk of severe liver failure and cardiovascular events.10–12 Therefore, improving liver function is as critical a therapeutic goal as glycemic control in patients with diabetes.

However, SGLT-2is are associated with several side effects, and frequent urination is one of the most common, affecting approximately 5% of patients who are prescribed these drugs.13

In particular, nocturia in older adults can lead to sleep disturbances because of nighttime awakenings, reduced quality of life,14,15 and even increased risks of depression and dementia,16 as well as falls and fractures.17,18 Therefore, addressing this issue is a critical challenge.

In this study, we investigated whether the fatty liver improvement effects of SGLT-2i differ by individual drugs or represent a class effect (a common effect across all drugs in this group), using three different SGLT-2is for evaluation. In addition, we focused on the fact that tofogliflozin has the shortest half-life in the blood and the shortest duration of action among SGLT-2is,19 and investigated whether its effect on nocturia differs from that of other SGLT-2is.

Materials and Methods

Study Design and Participants

The participants in this study were adults aged 30 years or older who were diagnosed with type 2 diabetes and fatty liver and were classified as having MASLD. Type 2 diabetes is diagnosed when at least two of the following criteria are fulfilled: a fasting plasma glucose level of 126 mg/dL (7.0 mmol/L) or higher, a 2-hour plasma glucose level of 200 mg/dL (11.1 mmol/L) or higher during a 75-g oral glucose tolerance test, a random plasma glucose of 200 mg/dL (11.1 mmol/L) or higher, and a hemoglobin A1c level of 6.5% (48 mmol/mol) or higher.20 Alternatively, patients who have already been diagnosed with type 2 diabetes based on the above tests. In addition, lipid droplets in more than 5% of hepatocytes, using ultrasonography, are defined as steatosis, referred to as fatty liver.21 Currently, hepatic steatosis involving more than 5% of the liver parenchyma can be diagnosed using B-mode ultrasonography.22,23

SGLT-2is were initiated or added to the treatment regimen for patients with HbA1c levels ≥7.5% (58 mmol/mol). MASLD was diagnosed according to the presence of fatty liver and at least one of the following five criteria:24 1. a BMI of ≥23.0 kg/m² or a waist circumference ≥94 cm for men and ≥80 cm for women; 2. prediabetes or diabetes treated with anti-diabetic medication; 3. blood pressure ≥130/85 mmHg or treatment with antihypertensive medication; 4. triglyceride (TG) levels ≥150 mg/dL or treatment with lipid-lowering medication; and 5. high-density lipoprotein cholesterol (HDL-C) levels ≤40 mg/dL for men or ≤50 mg/dL for women, or treatment for low HDL-C levels.

The exclusion criteria included patients already receiving SGLT-2is, those using insulin, and individuals with severe liver cirrhosis, severe renal dysfunction, pregnancy, or severe mental illness. In addition, patients with conditions known to cause nocturia, such as prostatic disorders or uterine prolapse, were excluded from the study.

Randomization

Eligible participants were randomized using a permuted block method stratified by age (≥65 years or <65 years), sex (male or female), HbA1c levels (≥9.0% or <9.0% [≥75 mmol/mol or <75 mmol/mol]), body weight (BMI ≥25.0 or <25.0 kg/m²), and duration of diabetes (≥10 years or <10 years). The participants were assigned to one of the following three groups in a 1:1:1 ratio: tofogliflozin group (Tofo group), empagliflozin group (Empa group), or dapagliflozin group (Dapa group). This trial is a prospective, randomized, open-label, blinded endpoint (PROBE) study conducted at a single center. The evaluation of liver fibrosis markers and nocturia frequency was independently performed by physicians and research staff from the adjudication committee, both of whom were blinded to treatment allocation.

Study Treatment

In the Tofo group, the participants received tofogliflozin at a dose of 20 mg. In the Empa group, the participants received empagliflozin at a dose of 10 mg. In the Dapa group, the participants received dapagliflozin at a dose of 5 mg. All these SGLT-2is were administered orally after breakfast.

During the study period, discontinuation of these SGLT-2is or the addition of other medications was not permitted unless adverse events occurred. The participants were followed up at outpatient visits initially after 1 month and subsequently every 2 months for a total of 6 months, resulting in a 7-month observation period. At each visit, measurements of height, weight, BMI, waist circumference, blood pressure, blood tests, body composition, and ultrasound elastography were conducted.

Outcomes

The primary endpoints were changes in the frequency of nocturia and liver function markers, namely aspartate aminotransferase (AST), alanine aminotransferase (ALT), gamma-glutamyl transpeptidase (γ-GPT), fibrosis-4 (FIB-4) index, and mac-2 binding protein glycan isomer (M2BPGi). In addition, liver fibrosis was evaluated using ultrasound elastography (Aplio i700, Canon Medical Systems, Tokyo). Shear wave speed measurements were interpreted as follows: values between 1.00 and 1.66 m/sec were classified as fibrosis grade (F) 0–1 (normal to mild fibrosis), with higher values indicating progression of fibrosis, ie, increased F score.25–27

Nocturia was defined as waking to urinate at least once during the night,28 with “nighttime” defined as the time between 11:00 pm and 5:00 am in this study.

Secondary endpoints included changes in HbA1c levels, fasting blood glucose, body weight, waist circumference, the lipid profile, and blood pressure. This study was conducted with the approval of the Institutional Review Board (IRB) of Shinkomonji Hospital. Written informed consent was obtained from all of the participants before enrollment. In addition, we confirmed that all research was performed following relevant guidelines/regulations.

This study has been performed in compliance with the Declaration of Helsinki. The trial protocol was registered with the UMIN Clinical Trial Registry (https://www.umin.ac.jp/ctr/) under the number UMIN000054278, with an initial registration date of 28/04/2024. The trial protocol is available in the Supplementary Material accompanying this manuscript.

Statistical Analyses

The sample size was calculated on the basis of the assumption that nighttime was defined as from 11:00 pm to 5:00 am, with the frequency of nighttime urination (nocturia) estimated to be 0.8 times in the Tofo group and 2.1 times in the Empa and Dapa groups. Nocturia frequency was calculated using a method based on the results of a pilot study (data not shown). Assuming a standard deviation of 2, an α error of 0.05, a statistical power of 0.8, and a 10% dropout rate over the 7-month follow-up, a 1:1:1 allocation required 43 participants in each group, resulting in a total sample size of 129 participants. Regarding the baseline characteristics, continuous variables are expressed as the mean ± standard deviation for descriptive analysis, and categorical variables are expressed as frequencies (%). To examine whether there were any variations in the samples between the groups at baseline, a test of homogeneity of variance (F-test) was conducted. Comparisons of continuous variables between the three groups at baseline and at each time point (1, 3, 5, and 7 months) were performed using an analysis of variance (ANOVA) with Dunnett’s test, and the Tofo group was set as the control. Temporal changes in continuous variables within each group were assessed using an ANOVA with Dunnett’s test, setting the baseline values as the control. In all cases, comparisons of categorical variables were performed using an m×n chi-square test. In this study, a p-value <0.05 was not considered significant. Instead, p-values adjusted using the Bonferroni correction (ie, 0.05 divided by the total number of pairwise comparisons) were considered significant under the concept of multiple comparisons. Statistical analyses were performed using SPSS ver. 17.0 (SPSS Inc., Chicago, IL, USA) and R software (version 4.1.1).

Results

Baseline Characteristics

The participants were recruited between April and the end of September 2024. A total of 135 participants were randomly assigned to the three groups (Figure 1). The participants had a mean age of 61 years and 43% were female. The mean BMI was 24.5 kg/m², the mean HbA1c level was 8.7%, the mean FIB-4 index was 1.84, the mean shear wave speed was 1.51 m/sec, and the mean number of nocturia episodes was 0.6 times. No significant differences in these variables were observed between the groups (Table 1).

Table 1 Baseline Characteristics

Figure 1 Screening, randomization, and follow-up. A total of 135 participants were randomly assigned to three groups of 45, each receiving a prescribed drug: the Tofo group received 20 mg of tofogliflozin, the Dapa group received 5–10 mg of dapagliflozin, and the Empa group received 10–25 mg of empagliflozin. All participants were followed for seven months. During the study, there were dropouts: 4 in the Tofo group, 5 in the Dapa group, and 4 in the Empa group. However, all participants were included in the analysis according to the intention-to-treat and per-protocol set criteria.

Primary Outcome of MASLD

All groups showed significant reductions in AST, ALT, and γ-GTP levels and the FIB-4 index compared with baseline (all p-values <0.05). In addition, a gradual reduction in shear wave speed-based fibrosis grade was observed compared with baseline across all groups (all p-values <0.05). However, no significant differences in these variables were found between the three groups. Furthermore, no significant changes in M2BPGi levels were observed in any group compared with baseline (Figure 2).

Figure 2 Changes in liver function and nocturia frequency with SGLT-2i treatment. Blue square, tofogliflozin group; purple diamond, dapagliflozin group; sky-blue circle, empagliflozin group. T-bars showed the standard deviations. * To assess time effects using analysis of variance followed by Dunnett’s test, baseline measurements (0 months) were compared with those at 1, 3, 5, and 7 months, in each of the three groups. † Comparisons between the Tofo group and the Dapa and Empa groups were conducted at each time point (0, 1, 3, 5, 7 months). The Bonferroni correction defined statistical significance as p<0.05/([4×3] + [2×5]) = 0.00227. Therefore, * and † denote p <0.00227.

Abbreviations: AST, aspartate aminotransferase; ALT, alanine aminotransferase; γ-GPT, gamma-glutamyl transpeptidase; FIB-4, fibrosis-4; M2BPGi, mac-2 binding protein glycan isomer.

Primary Outcome of the Frequency of Nocturia

In the Empa and Dapa groups, the frequency of nocturia significantly increased to 1.7 and 1.9 times, respectively, 1 month after starting medication compared with baseline. In contrast, the Tofo group showed an increase in the frequency of nocturia by only 0.8 times, which was not statistically significant. Consequently, the Tofo group showed a significantly lower frequency of nocturia than the other two groups (p<0.001). This difference persisted until 3 months after the initiation of treatment; however, the frequencies were 1.3, 1.2, and 0.7 times, respectively, with a p-value of 0.010, which did not meet the significance threshold after Bonferroni correction (p < 0.00227). In addition, the difference diminished after the fifth month. Over time, the frequency of nocturia decreased in all three groups, with no significant changes from baseline or differences between the groups (Figure 2).

Secondary Outcomes

All three groups showed a significant reduction in weight and waist circumference compared with baseline, with no significant differences between the groups. No significant changes in systolic or diastolic blood pressure were observed in any group compared with baseline. Fasting plasma glucose and HbA1c levels were significantly decreased in all three groups compared with baseline. While the Dapa and Empa groups showed a slight trend of a greater reduction in these variables than the Tofo group, no significant differences were observed between the groups. A significant reduction in TG levels was observed in all three groups, with no significant intergroup differences. In females, no significant changes in HDL-C levels were observed in any group. In contrast, in males, all three groups showed a significant increase in HDL-C levels at 1 month compared with baseline, with no differences between the groups (Figure 3). Low-density lipoprotein-cholesterol levels remained unchanged in all groups compared with baseline, with no difference between the sexes (data not shown).

Figure 3 Changes in body weight, blood pressure, blood glucose, and lipid levels with SGLT-2i treatment. Blue square, tofogliflozin group; purple diamond, dapagliflozin group; sky-blue circle, empagliflozin group. T-bars showed standard deviations. *To assess time effects using analysis of variance followed by Dunnett’s test, baseline measurements (0 month) were compared with those at 1, 3, 5, and 7 months in each of the three groups. Comparisons between the Tofo group and the Dapa and Empa groups were conducted at each time point (0, 1, 3, 5, 7 months). The Bonferroni correction defined statistical significance as p<0.05/([4×3] + [2×5]) = 0.00227. Therefore, *denotes p <0.00227.

Abbreviations: BMI, body mass index; BP, blood pressure; HbA1c, glycated hemoglobin; FPG, fasting blood glucose; TG, triglycerides; HDL-C, high-density lipoprotein-cholesterol; LDL, low-density lipoprotein-cholesterol.

Sub-Analysis

A sub-analysis was conducted by stratifying the participants according to diabetes duration, age, baseline HbA1c levels, and sex. Based on the baseline distributions, participants were categorized into tertiles: 33–59, 60–71, and 72–89 years for age, and 7.5–8.9%, 9.0–10.3%, and 10.4–12.4% for HbA1c levels.

This analysis revealed that older age and higher baseline HbA1c levels were associated with greater effectiveness of tofogliflozin in reducing the frequency of nocturia (Table 2). No significant differences in these variables according to sex were observed. In addition, the frequency of daytime (5:00 a.m. to 11:00 p.m.) urinations increased to 7.9 times in the Tofo group, 7.1 times in the Dapa group, and 6.9 times in the Empa group after one month, with a significant difference among the three groups (p = 0.0014). Subsequently, urinary frequency gradually declined, and no significant differences were observed among the groups after three months (Figure 4).

Table 2 Frequency of Nocturia

Figure 4 Frequency of daytime urination. The daytime urinary frequency increased after one month to an average of 7.9 times in the Tofo group, 6.9 times in the Dapa group, and 7.1 times in the Empa group, with a statistically significant difference among the three groups (p = 0.0014). Subsequently, urinary frequency gradually declined, and no significant differences were observed among the groups after three months. * To assess time effects using analysis of variance followed by Dunnett’s test, baseline measurements (0 month) were compared with those at 1, 3, 5, and 7 months in each of the three groups. † Comparisons between the Tofo group and the Dapa and Empa groups were conducted at each time point (0, 1, 3, 5, 7 months). The Bonferroni correction defined statistical significance as p <0.05/([4×3] + [2×5]) = 0.00227. Therefore, * and † denote p < 0.00227.

Adverse Events

Genital mycotic infections and urinary tract infections were observed in all three groups (Table 3). No significant differences in the incidence rates of these events were observed between the three groups.

Table 3 Frequency of Adverse Events

Discussion

In this study, tofogliflozin, dapagliflozin, and empagliflozin showed reduced and improved liver function and fibrosis markers associated with MASLD. However, no significant differences in these markers were detected between the three groups. Notably, tofogliflozin significantly reduced the frequency of nocturia 1 month after initiating treatment compared with the other two drugs. However, the difference disappeared after three months. There was no significant difference in weight loss, an improvement in blood glucose levels, or a change in lipid profile between the groups.

Effect of SGLT-2is on MASLD

Recently, there has been an increase in reports showing the efficacy of SGLT-2is in improving MASLD.5–7 However, whether this effect represents a class effect of SGLT-2is remains unclear. Our findings suggest that SGLT-2is have a class effect in improving fatty liver or, at least, that the three drugs used in this study have beneficial effects on MASLD.

A total of 68.8% of patients with type 2 diabetes have been reported to have coexisting MASLD,8 which is associated with an increased risk of liver fibrosis.29 However, randomized controlled trials on SGLT-2is have shown a reduction in hepatic fat content as measured by magnetic resonance imaging.30,31

SGLT-2is lower blood glucose levels by inhibiting SGLT-2, a protein predominantly expressed in the renal tubules, thereby reducing glucose reabsorption in the kidneys. This mechanism is associated with an improvement in insulin resistance, a reduction in free fatty acids, decreased hepatic fat deposition, and amelioration of intrahepatic inflammation and fibrosis.32 In our study as well, significant improvement in liver fibrosis was observed at five months after the initiation of treatment. Furthermore, SGLT-2is have been shown to enhance insulin sensitivity by increasing adiponectin levels33 and to promote glucagon-like peptide 1 secretion,34,35 both of which may contribute to the improvement of MASLD. Notably, SGLT-2 expression is not limited to the kidneys; it has also been detected in hepatocytes and bile duct epithelial cells.36 This finding suggests that SGLT-2is exert direct effects on the liver or affect hepatic function indirectly through bile acids and the gut microbiota, resulting in reduced hepatic fat deposition and inflammation. The use of SGLT-2is in the treatment of type 2 diabetes has shown potential therapeutic benefits in MASLD, possibly through mechanisms that may indirectly influence the progression of liver disease.37,38 In addition, as discussed later, although tofogliflozin has a shorter half-life compared to the other two agents, no significant differences in liver function improvement, including antifibrotic effects, were observed among the three groups. These findings suggest that, similar to its glucose-lowering effects, the improvement in liver function may be independent of the drug’s half-life duration.

Effect of SGLT-2is on Nocturia Frequency

Nocturia is defined as the need to urinate during the night, interrupting sleep at least once.28 Nocturia is associated with sleep disturbances,15 an increased risk of falls and fractures,17,18,39 and an elevated risk of depression and dementia.16 These effects can greatly reduce quality of life, especially in older adults,40–42 highlighting the importance of preventing and treating nocturia.

Frequent urination is a common side effect of SGLT-2is, leading to increased urination during the day and at night, which may contribute to nocturia. A randomized controlled trial reported that approximately 5% of individuals experience this side effect.13 However, other studies have shown that empagliflozin increases daily urine volume by 500 mL,43 and nearly all individuals taking SGLT-2is exhibit some increase in urinary frequency.44 The change in frequency of urination can vary according to the type of SGLT-2i used and the patients’ underlying conditions. However, SGLT-2is contribute to the development of frequent urination. This effect is particularly prominent in patients with poor glycemic control at the time of prescription, because osmotic diuresis and nephropathy are thought to further increase the frequency of urination.13 On the contrary, several studies have noted that while polyuria is a frequent early complaint, it often subsides within weeks of treatment initiation.1,45,46 This timeline coincides with the period in which blood glucose begins to normalize, suggesting a strong correlation between the resolution of polyuria and improved glycemic control. The EMPA-REG and CANVAS studies involving SGLT-2is demonstrated the greatest improvement in glycemic control at 12 to 13 weeks (approximately 3 months) following drug initiation,1,46 aligning with the 3-month time point observed in the present study. Frequent urination caused by SGLT-2is is largely due to osmotic diuresis driven by glucosuria. As blood glucose control improves, the filtered load of glucose decreases, leading to reduced glucosuria and, consequently, decreased urinary frequency.47

The blood half-life of tofogliflozin is 5–6 hours,19 which is shorter than that of empagliflozin (9.88–13.1 hours)48 and dapagliflozin (12.9 hours).49 Therefore, when tofogliflozin is taken in the morning, its effects are likely to diminish by nighttime. In this study, the frequency of nocturia in the Tofo group did not significantly increase (0.8 times), and this small increase was notably less than that in the other two groups (1.7 and 1.9 times, respectively).

This trend was particularly evident in patients with higher baseline HbA1c levels or older age. Therefore, when initiating SGLT-2i therapy, tofogliflozin may be a more appropriate option for patients with baseline HbA1c levels ≥ 9.0% or those aged ≥ 72 years, given its potential benefit in reducing nocturia frequency. Conversely, the nocturia-reducing effect of tofogliflozin may be limited in patients with baseline HbA1c levels ≤ 8.9% or age ≤ 59 years.

In addition, we did not find any significant differences in the daytime urination frequency or a reduction in HbA1c levels among the three groups.

Adverse Events of SGLT-2is

Regarding adverse events, urinary tract infections and genital infections were observed in all SGLT-2i groups. However, the incidence of these events is comparable to previously reported rates,13 with no significant differences between the groups.

Study Limitation

This study has several limitations, such as its single-center design, the fact that only Japanese participants were included, and the 7-month study duration. Liver function may continue to change beyond 7 months. While a significant difference in the frequency of nocturia was observed at 1 and 3 months, this difference diminished over time, suggesting that additional long-term studies may not be required.

Conclusion

All three SGLT-2is led to a reduction in MASLD-related parameters, but no significant differences in these parameters were observed between the groups. These findings suggest that the improvement in fatty liver associated with SGLT-2is may be a class effect. SGLT-2 is are now widely recognized not only for their glucose-lowering effects in patients with diabetes but also for their cardioprotective and renoprotective effects. The findings of this study may contribute to the growing body of evidence supporting the potential hepatoprotective effects of SGLT-2 is. Tofogliflozin significantly reduced the frequency of nocturia compared with the other two SGLT-2is. This finding suggests that the shorter half-life in the blood of tofogliflozin may be particularly beneficial for patients with baseline HbA1c levels ≥ 9.0% or age ≥ 72 years.

Data Sharing Statement

Some or all datasets generated during and/or analyzed during the current study are not publicly available but are available from the corresponding author on reasonable request.

Ethics Approval and Consent to Participate

This study was conducted with the approval of the Institutional Review Board (IRB) of Shinkomonji Hospital. Written informed consent was obtained from all of the participants before enrollment. In addition, we confirmed that all research was performed following relevant guidelines/regulations.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Funding

This study was supported by a grant from the Fukuoka Medical Association (T.K.). The sponsor did not contribute to the design, collection, management, analysis, interpretation of data, writing of the manuscript, or the decision to submit the manuscript for publication.

Disclosure

All the authors declare no competing interests in this work.

References

1. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373:2117–2128.

2. Mosenzon O, Wiviott SD, Cahn A, et al. Effects of dapagliflozin on development and progression of kidney disease in patients with type 2 diabetes: an analysis from the DECLARE-TIMI 58 randomised trial. Lancet Diabetes Endocrinol. 2019;7(8):606–617. doi:10.1016/S2213-8587(19)30180-9

3. Heerspink HJL, Stefánsson BV, Correa-Rotter R, et al. Dapagliflozin in patients with chronic kidney disease. N Engl J Med. 2020;383(15):1436–1446. doi:10.1056/NEJMoa2024816

4. The E-KCG. Empagliflozin in patients with chronic kidney disease. N Engl J Med. 2023;388:117–127.

5. Seko Y, Sumida Y, Sasaki K, et al. Effects of canagliflozin, an SGLT2 inhibitor, on hepatic function in Japanese patients with type 2 diabetes mellitus: pooled and subgroup analyses of clinical trials. J Gastroenterol. 2018;53(1):140–151. doi:10.1007/s00535-017-1364-8

6. Takahashi H, Kessoku T, Kawanaka M, et al. Ipragliflozin improves the hepatic outcomes of patients with diabetes with NAFLD. Hepatol Commun. 2022;6(1):120–132. doi:10.1002/hep4.1696

7. Mantovani A, Byrne CD, Targher G. Efficacy of peroxisome proliferator-activated receptor agonists, glucagon-like peptide-1 receptor agonists, or sodium-glucose cotransporter-2 inhibitors for treatment of non-alcoholic fatty liver disease: a systematic review. Lancet Gastroenterol Hepatol. 2022;7(4):367–378. doi:10.1016/S2468-1253(21)00261-2

8. Younossi ZM, Golabi P, Price JK, et al. The global epidemiology of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis among patients with type 2 diabetes. Clin Gastroenterol Hepatol. 2024;22(10):1999–2010e1998. doi:10.1016/j.cgh.2024.03.006

9. Fujii H, Suzuki Y, Sawada K, et al. Prevalence and associated metabolic factors of nonalcoholic fatty liver disease in the general population from 2014 to 2018 in Japan: a large-scale multicenter retrospective study. Hepatol Res. 2023;53(11):1059–1072. doi:10.1111/hepr.13947

10. Sanyal AJ, Van Natta ML, Clark J, et al. Prospective study of outcomes in adults with nonalcoholic fatty liver disease. N Engl J Med. 2021;385(17):1559–1569. doi:10.1056/NEJMoa2029349

11. Targher G, Corey KE, Byrne CD. NAFLD, and cardiovascular and cardiac diseases: factors influencing risk, prediction and treatment. Diabetes Metab. 2021;47(2):101215. doi:10.1016/j.diabet.2020.101215

12. Fujii H, Iwaki M, Hayashi H, et al. Clinical outcomes in biopsy-proven nonalcoholic fatty liver disease patients: a multicenter registry-based cohort study. Clin Gastroenterol Hepatol. 2023;21(2):370–379. doi:10.1016/j.cgh.2022.01.002

13. Krepostman N, Kramer H. Lower urinary tract symptoms should be queried when initiating sodium glucose co-transporter 2 inhibitors. Kidney. 2021;2(4):751–754. doi:10.34067/KID.0000472021

14. van Doorn B, Blanker MH, Kok ET, Westers P, Bosch JL. Prevalence, incidence, and resolution of nocturnal polyuria in a longitudinal community-based study in older men: the Krimpen study. Eur Urol. 2013;63(3):542–547. doi:10.1016/j.eururo.2012.10.004

15. Abdel Rahman TT, El Gaafary MM. Nocturia among elderly men living in a rural area in Egypt, and its impact on sleep quality and health-related quality of life. Geriatrics Gerontol Int. 2014;14(3):613–619. doi:10.1111/ggi.12145

16. Bliwise DL, Wagg A, Sand PK. Nocturia: a highly prevalent disorder with multifaceted consequences. Urology. 2019;133S:3–13. doi:10.1016/j.urology.2019.07.005

17. Pesonen JS, Vernooij RWM, Cartwright R, et al. The impact of nocturia on falls and fractures: a systematic review and meta-analysis. J Urol. 2020;203(4):674–683. doi:10.1097/JU.0000000000000459

18. Committee for establishment of the clinical guidelines for nocturia of the neurogenic bladder, S. Clinical guidelines for nocturia. Int J Urol. 2010;17(5):397–409. doi:10.1111/j.1442-2042.2010.02527.x

19. Kasahara-Ito N, Fukase H, Ogama Y, et al. Pharmacokinetics and pharmacodynamics of tofogliflozin (a selective SGLT2 inhibitor) in healthy male subjects. Drug Res. 2017;67(06):349–357. doi:10.1055/s-0043-104779

20. American Diabetes A. 2. Classification and diagnosis of diabetes: standards of medical care in diabetes-2021. Diabetes Care. 2021;44:S15–S33.

21. Chalasani N, Younossi Z, Lavine JE, et al. The diagnosis and management of nonalcoholic fatty liver disease: practice guidance from the American association for the study of liver diseases. Hepatology. 2018;67(1):328–357. doi:10.1002/hep.29367

22. Karlas T, Petroff D, Sasso M, et al. Individual patient data meta-analysis of controlled attenuation parameter (CAP) technology for assessing steatosis. J Hepatol. 2017;66(5):1022–1030. doi:10.1016/j.jhep.2016.12.022

23. Fujiwara Y, Kuroda H, Abe T, et al. The B-mode image-guided ultrasound attenuation parameter accurately detects hepatic steatosis in chronic liver disease. Ultrasound Med Biol. 2018;44(11):2223–2232. doi:10.1016/j.ultrasmedbio.2018.06.017

24. Rinella ME, Lazarus JV, Ratziu V, et al. A multisociety Delphi consensus statement on new fatty liver disease nomenclature. Hepatology. 2023;78(6):1966–1986. doi:10.1097/HEP.0000000000000520

25. Trout AT, Xanthakos SA, Bennett PS, Dillman JR. Liver shear wave speed and other quantitative ultrasound measures of liver parenchyma: prospective evaluation in healthy children and adults. AJR Am J Roentgenol. 2020;214(3):557–565. doi:10.2214/AJR.19.21796

26. Dhyani M, Xiang F, Li Q, et al. Ultrasound shear wave elastography: variations of liver fibrosis assessment as a function of depth, force and distance from central axis of the transducer with a comparison of different systems. Ultrasound Med Biol. 2018;44(11):2209–2222. doi:10.1016/j.ultrasmedbio.2018.07.003

27. Sugimoto K, Moriyasu F, Dioguardi Burgio M, et al. US markers and necroinflammation, steatosis, and fibrosis in metabolic dysfunction-associated steatotic liver disease: the iLEAD study. Radiology. 2024;312(2):e233377. doi:10.1148/radiol.233377

28. Hashim H, Blanker MH, Drake MJ, et al. International Continence Society (ICS) report on the terminology for nocturia and nocturnal lower urinary tract function. Neurourol Urodyn. 2019;38(2):499–508. doi:10.1002/nau.23917

29. Lonardo A, Nascimbeni F, Mantovani A, Targher G. Hypertension, diabetes, atherosclerosis and NASH: cause or consequence? J Hepatol. 2018;68(2):335–352. doi:10.1016/j.jhep.2017.09.021

30. Kuchay MS, Krishan S, Mishra SK, 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(8):1801–1808. doi:10.2337/dc18-0165

31. Brunt EM, Kleiner DE, Wilson LA, et al. Nonalcoholic fatty liver disease (NAFLD) activity score and the histopathologic diagnosis in NAFLD: distinct clinicopathologic meanings. Hepatology. 2011;53(3):810–820. doi:10.1002/hep.24127

32. Kothari S, Dhami-Shah H, Shah SR. Antidiabetic drugs and statins in nonalcoholic fatty liver disease. J Clin Exp Hepatol. 2019;9(6):723–730. doi:10.1016/j.jceh.2019.06.003

33. Kahl S, Gancheva S, Straßburger K, et al. Empagliflozin effectively lowers liver fat content in well-controlled type 2 diabetes: a randomized, double-blind, phase 4, placebo-controlled trial. Diabetes Care. 2020;43(2):298–305. doi:10.2337/dc19-0641

34. Ito D, Shimizu S, Inoue K, et al. 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(10):1364–1372. doi:10.2337/dc17-0518

35. Kadowaki T, Inagaki N, Kondo K, et al. Efficacy and safety of canagliflozin as add-on therapy to teneligliptin in J apanese patients with type 2 diabetes mellitus: results of a 24-week, randomized, double-blind, placebo-controlled trial. Diabetes Obes Metab. 2017;19(6):874–882. doi:10.1111/dom.12898

36. Nakano D, Akiba J, Tsutsumi T, et al. Hepatic expression of sodium-glucose cotransporter 2 (SGLT2) in patients with chronic liver disease. Med Mol Morphol. 2022;55(4):304–315. doi:10.1007/s00795-022-00334-9

37. Tang H, Dai Q, Shi W, et al. SGLT2 inhibitors and risk of cancer in type 2 diabetes: a systematic review and meta-analysis of randomised controlled trials. Diabetologia. 2017;60(10):1862–1872. doi:10.1007/s00125-017-4370-8

38. Kaji K, Nishimura N, Seki K, et al. Sodium glucose cotransporter 2 inhibitor canagliflozin attenuates liver cancer cell growth and angiogenic activity by inhibiting glucose uptake. Int J Cancer. 2018;142(8):1712–1722. doi:10.1002/ijc.31193

39. Saran R, Shahinian V. CKD: a pandemic calling for concerted public health action. Adv Chronic Kidney Dis. 2010;17(3):213–214. doi:10.1053/j.ackd.2010.03.009

40. Andersson F, Anderson P, Holm-Larsen T, et al. Assessing the impact of nocturia on health-related quality-of-life and utility: results of an observational survey in adults. J Med Econ. 2016;19(12):1200–1206. doi:10.1080/13696998.2016.1211136

41. Miranda Ede P, Gomes CM, Torricelli FCM, et al. Nocturia is the lower urinary tract symptom with greatest impact on quality of life of men from a community setting. Int Neurourol J. 2014;18(2):86–90. doi:10.5213/inj.2014.18.2.86

42. Kupelian V, Wei JT, O’Leary MP, et al. Nocturia and quality of life: results from the Boston area community health survey. Eur Urol. 2012;61(1):78–84. doi:10.1016/j.eururo.2011.05.065

43. Mordi NA, Mordi IR, Singh JS, et al. Renal and cardiovascular effects of sglt2 inhibition in combination with loop diuretics in patients with type 2 diabetes and chronic heart failure: the RECEDE-CHF trial. Circulation. 2020;142(18):1713–1724. doi:10.1161/CIRCULATIONAHA.120.048739

44. Saijo Y, Okada H, Hata S, et al. Reasons for discontinuing treatment with sodium-glucose cotransporter 2 inhibitors in patients with diabetes in real-world settings: the KAMOGAWA-A study. J Clin Med. 2023;12(22):6993. doi:10.3390/jcm12226993

45. Vallon V:. State-of-the-art-review: mechanisms of action of SGLT2 inhibitors and clinical implications. Am J Hypertens. 2024;37(11):841–852. doi:10.1093/ajh/hpae092

46. Neal B, Perkovic V, Mahaffey KW, et al. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med. 2017;377(7):644–657. doi:10.1056/NEJMoa1611925

47. Heerspink HJ, Perkins BA, Fitchett DH, Husain M, Cherney DZ. Sodium glucose cotransporter 2 inhibitors in the treatment of diabetes mellitus: cardiovascular and kidney effects, potential mechanisms, and clinical applications. Circulation. 2016;134(10):752–772. doi:10.1161/CIRCULATIONAHA.116.021887

48. Scheen AJ. Pharmacokinetic and pharmacodynamic profile of empagliflozin, a sodium glucose co-transporter 2 inhibitor. Clin Pharmacokinet. 2014;53(3):213–225. doi:10.1007/s40262-013-0126-x

49. Kasichayanula S, Liu X, Lacreta F, Griffen SC, Boulton DW. Clinical pharmacokinetics and pharmacodynamics of dapagliflozin, a selective inhibitor of sodium-glucose co-transporter type 2. Clin Pharmacokinet. 2014;53(1):17–27. doi:10.1007/s40262-013-0104-3

Continue Reading