Next Article in Journal
The Changing Complementary Role of Multimodality Imaging in Clinical Cardiology
Previous Article in Journal
Evaluation of a Post-Operative Therapy Protocol after Epithelium-Off Corneal Cross-Linking in Patients Affected by Keratoconus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Choice of Glucose-Lowering Drugs as Initial Monotherapy for Type 2 Diabetes Patients with Contraindications or Intolerance to Metformin: A Systematic Review and Meta-Analysis

1
Center for Health Policy and Management Studies, School of Government, Nanjing University, Nanjing 210023, China
2
College of Politics and Public Administration, Qingdao University, Qingdao 266071, China
3
Department of Global Health Management and Policy, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
4
Centre for Health Management and Policy Research, School of Public Health, Cheeloo College of Medicine (NHC Key Laboratory of Health Economics and Policy Research), Shandong University, Jinan 250012, China
5
Center for Health Policy Studies, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
*
Authors to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(23), 7094; https://doi.org/10.3390/jcm11237094
Submission received: 31 October 2022 / Revised: 24 November 2022 / Accepted: 27 November 2022 / Published: 30 November 2022
(This article belongs to the Section Endocrinology & Metabolism)

Abstract

:
Background: There are multiple glucose-lowering drugs available as alternative initial monotherapy for type 2 diabetes patients with contraindications or intolerance to metformin. However, little comparative and systematic data are available for them as initial monotherapy. This study estimated and compared the treatment effects of glucose-lowering drugs as initial monotherapy for type 2 diabetes. Methods: PubMed, Web of Science, Embase, CNKI, Chongqing VIP, and WanFang Data from 1 January 1990 until 31 December 2020 were searched for randomized controlled trials which compared a glucose-lowering drug with placebo/lifestyle-intervention for type 2 diabetes. Drug classes included metformin, sulfonylureas (SUs), thiazolidinediones (TZDs), glinides (NIDEs), α-glucosidase inhibitors (AGIs), dipeptidyl peptidase-4 inhibitors (DPP-4is), sodium-glucose cotransporter-2 inhibitors (SGLT2is), insulins (INSs), and glucagon-like peptide-1 receptor agonists (GLP-1RAs). Results: A total of 185 trials were included, identifying 38,376 patients from 56 countries across six continents. When choosing an initial drug monotherapy alternative to metformin, SUs were most efficacious in reducing HbA1c (−1.39%; 95% CI −1.63, −1.16) and FPG (−2.70 mmol/L; 95% CI −3.18, −2.23), but increased hypoglycemia risks (5.44; 95% CI 2.11, 14.02). GLP-1RAs were most efficacious in reducing BMI (−1.05 kg/m2; 95% CI −1.81, −0.29) and TC (−0.42 mmol/L; 95% CI −0.61, −0.22). TZDs were most efficacious in increasing HDL-C (0.12 mmol/L; 95% CI 0.07, 0.17). SGLT2is were most efficacious in lowering SBP (−4.18 mmHg; 95% CI −4.84, −3.53). While AGIs conferred higher risk of AE-induced discontinuations (2.57; 95% CI 1.64, 4.03). Overall, only GLP-1RAs showed an integrated beneficial effect on all outcomes. Our results also confirmed the intraclass differences in treatment effects across drugs. Most trials were short-term, and no significant differences in mortality, total vascular events, myocardial infarction, heart failure, stroke, or diabetic nephropathy were observed across drug classes. Conclusions: Our results suggest a potential treatment hierarchy for decision-makers, with GLP-1RAs being the preferred alternative therapy to metformin regarding their favorable efficacy and safety profiles.

1. Introduction

Diabetes is among the top ten causes of death, imposing serious threats to global health and economy [1]. Globally, one in eleven adults were living with diabetes (463 million), 4.2 million deaths were due to diabetes, and 10% of the health expenditure (USD 760 billion) was spent on diabetes in 2019 [1]. Type 2 diabetes accounts for around 90% of all diabetes, which is characterized by chronic hyperglycemia [2]. Hyperglycemia, if left poorly managed, can lead to severe diabetes-related vascular events and even death. Conversely, if appropriately managed, the events can be prevented or delayed [1,3].
Clinical guidelines recommend metformin as the preferred initial monotherapy added to lifestyle interventions (LIs) for type 2 diabetes patients with proven effectiveness and safety [3,4]. However, in patients with contraindications or intolerance to metformin, initial monotherapy is recommended to consider a glucose-lowering drug from another class based on patient factors and drug-specific effects [3,4]. There are eight alternative drug classes available worldwide, including sulfonylureas (SUs), thiazolidinediones (TZDs), glinides (NIDEs), α-glucosidase inhibitors (AGIs), dipeptidyl peptidase-4 inhibitors (DPP-4is), sodium-glucose cotransporter-2 inhibitors (SGLT2is), insulins (INSs), and glucagon-like peptide-1 receptor agonists (GLP-1RAs), which have increased the complexity of clinical drug use. A national survey in China reported 35.8% of type 2 diabetes patients used monotherapy; among which, insulin secretagogues (SUs or NIDEs) were most commonly used (45.6%), followed by metformin (30%) and AGIs (18%) [5]. However, 32% of patients changed their therapies within a year, attributable to insufficient efficacy and/or intolerant adverse events (AEs) [5].
Regarding the heavy disease burden and frequent therapy alteration, it is of value to choose a rational drug as alternative initial monotherapy when metformin is contraindicated or intolerant. Evidence shows that focusing solely on glucose control has a limited effect on reducing the risks of diabetes-related vascular events and death, while a comprehensive control of cardiovascular risk factors (e.g., hypertension, dyslipidemia) can bring greater benefits, particularly among patients with special characteristics and needs [3,6]. In addition, the diabetes associations recommend a patient-centered approach to guide the choice of drugs, considering efficacy, weight impact, hypoglycemia risk, effect on cardiovascular and renal comorbidities, and patient preferences [4,7]. It is essential to fully understand the advantages and disadvantages of each drug before making a choice. However, there is little systematic and comparative data available for all the alternative drugs as initial monotherapy of type 2 diabetes [4]. Therefore, this study systematically estimated and compared the treatment effects of all available glucose-lowering drugs when used as initial monotherapy for type 2 diabetes, so as to enable decision-makers to make informed choice when choosing an alternative drug as initial monotherapy for patients with contraindications or intolerance to metformin.

2. Methods

This study was carried out in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) statement [8] and was registered at PROSPERO (CRD42020170769). We estimated the drug-specific effects of each drug after removing the effects of placebo (PBO) and LI and compared the drug classes against each other.

2.1. Eligibility Criteria

Studies were eligible if (1) participants were type 2 diabetes patients ≥18 years; (2) intervention was a glucose-lowering drug monotherapy; (3) comparator was PBO or LI such as diet and/or exercise; (4) only LI was allowed as background therapy; (5) study duration was ≥12 weeks; (6) outcomes were hemoglobin Alc (HbA1c), fasting plasma glucose (FPG), body mass index (BMI), total cholesterol (TC), high-density lipoprotein-cholesterol (HDL-C), systolic blood pressure (SBP), hypoglycemia, mortality, vascular outcomes, or discontinuation; (7) study design was randomized controlled trial (RCT); and (8) study was published in English or Chinese. Detailed eligibility criteria are shown in Supplementary Table S1.
27 glucose-lowering drugs from nine drug classes were targeted:
BiguanideMetformin
SUsGlyburide, glimepiride, gliclazide, glipizide, and gliquidone
TZDsRosiglitazone and pioglitazone
NIDEsRepaglinide, nateglinide, and mitiglinide
AGIsAcarbose, voglibose, and miglitol
DPP-4isSitagliptin, saxagliptin, vildagliptin, linagliptin, and alogliptin
SGLT2isDapagliflozin, empagliflozin, and canagliflozin
INSsInsulin and insulin analogs
GLP-1RAsExenatide, liraglutide, lixisenatide, and beinaglutide

2.2. Information Sources and Searches

We systematically searched PubMed, Web of Science, Embase, China National Knowledge Infrastructure (CNKI), Chongqing VIP, and WanFang Data for RCTs published between 1 January 1990 and 31 December 2020, which compared a glucose-lowering drug against PBO (or LI) with or without a background of LI for type 2 diabetes. Search terms used were type 2 diabetes, 27 glucose-lowering drugs, nine drug classes, placebo, diet, exercise, and lifestyle. Detailed search strategies are shown in Supplementary Table S2. The reference lists of relevant reviews and retrieved studies were manually screened to supplement for database searching.

2.3. Study Selection

After removing the duplicates, two researchers (S.G. and X.H.) screened the title and abstract of each retrieved record independently, and then examined potentially eligible records by reading full texts. The results were cross-checked by two other researchers (X.S. and Y.G.). Any potential disagreements were resolved through consensus by the research team.

2.4. Data Extraction and Quality Assessment

Two researchers (S.G. and X.H.) independently extracted data using a standardized form and assessed the risk of bias of each study by using the Cochrane Collaboration’s risk of bias assessment tool [9]. The extracted data included study characteristics, patient characteristics, intervention, comparator, outcomes, and other relevant data. The publication bias across studies was estimated using Egger’s test or Harbord’s modified test. The results were cross-checked by two other researchers (X.S. and Y.G.). Any potential discrepancies were resolved by consensus.

2.5. Data Synthesis and Analysis

Firstly, we conducted a series of meta-analyses to estimate the treatment effects of different drugs on each outcome, after removing the effects of PBO/LI. Then, we conducted the adjusted indirect treatment comparisons on the basis of the Bucher method to compare the treatment effects of the drugs with each other by using PBO/LI as common comparator [10,11]. For continuous outcomes, we calculated the weighted mean difference (WMD) with 95% confidence intervals (CIs). For dichotomous outcomes, we calculated the relative risk (RR) with 95% CIs. We used a random-effects model conservatively by assuming a substantial variability in effect size across studies and drugs [12,13]. We used the I2 statistic to evaluate heterogeneity, which was considered important when it was above 50% [14]. The subgroup analyses were performed based on individual drugs within the same class. The sensitivity analyses were performed using fixed-effects meta-analyses. All analyses were performed in Stata/SE 15.1.

3. Results

The initial search identified 30,124 records, of which 420 records were assessed in full texts. Finally, 185 trials were included [15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199], which identified 38,376 patients randomly assigned to 23 drugs from eight classes or PBO/LI (Figure 1). The sample size of the trials varied from 16 to 888. The study duration ranged from 12 weeks to 108 weeks. Metformin was studied in 40 trials, with pioglitazone (23 trials), acarbose (22 trials), and rosiglitazone (21 trials) being the three next most studied drugs. No eligible trial was detected for gliquidone, mitiglinide, beinaglutide, or insulin. The participants were from 56 countries across six continents. Mean age varied from 30.8 years to 74.5 years, with a diabetes duration of 0.23 year to 9.9 years. The characteristics with the risk of bias of the trials are presented in Supplementary Table S3. The publication bias is shown in Supplementary Figure S1.

3.1. Intermediate Outcomes

3.1.1. Hemoglobin Alc

A total of 167 trials (34,133 patients) provided data on HbA1c. All drugs significantly reduced HbA1c after removing the PBO/LI effects. The largest PBO/LI-subtracted reductions were evident with SUs (1.39%; 95% CI 1.16, 1.63), followed by GLP-1RAs (0.99%; 95% CI 0.78, 1.20); while NIDEs (0.44%; 95% CI 0.20, 0.69) resulted in the smallest reductions. There were great differences in HbA1c reductions detected within GLP-1RAs, ranging from 0.60% (95% CI 0.37, 0.83) with lixisenatide to 1.17% (95% CI 0.87, 1.47) with liraglutide; and within AGIs, ranging from 0.20% (95% CI 0.07, 0.33) with voglibose to 0.74% (95% CI 0.52, 0.96) with acarbose (Table 1; Supplementary Figures S2–S9).
Across the drug classes, SUs produced significantly greater HbA1c reductions than all other drug classes, ranging from 0.40% (95% CI 0.09, 0.72) vs. GLP-1RAs to 0.95% (95% CI 0.61, 1.29) vs. NIDEs. In addition, GLP-1RAs, metformin, and TZDs were superior to DPP-4is, AGIs, and NIDEs. SGLT2is worked better than DPP-4is and NIDEs. The greatest contrast among all comparisons was detected between SUs and NIDEs (Table 2).

3.1.2. Fasting Plasma Glucose

A total of 163 trials (33,963 patients) provided data on FPG. All drug classes significantly reduced FPG after removing the PBO/LI effects. The greatest PBO/LI-subtracted reductions were seen with SUs (2.70 mmol/L; 95% CI 2.23, 3.18), ranging from 2.22 mmol/L (95% CI 0.97, 3.47) with gliclazide to 3.02 mmol/L (95% CI 2.20, 3.85) with glipizide, followed by TZDs (1.91 mmol/L; 95% CI 1.60, 2.23), with reductions of 1.73 mmol/L (95% CI 1.14, 2.32) for rosiglitazone vs. 2.01 mmol/L (95% CI 1.67, 2.34) for pioglitazone. As a class, NIDEs conferred the smallest reductions (0.75 mmol/L; 95% CI 0.45, 1.04), which was inferior to AGIs (1.19 mmol/L; 95% CI 0.64, 1.73); but large differences within AGIs were noted, with a 1.17 mmol/L (95% CI 0.50, 1.83) reduction with acarbose vs. no significant reductions with other AGIs (Table 1; Supplementary Figures S10–S17).
Across the drug classes, SUs showed significantly greater FPG reductions than all other drug classes, ranging from 0.79 mmol/L (95% CI 0.22, 1.36) vs. TZDs to 1.95 mmol/L (95% CI 1.39, 2.51) vs. NIDEs. In addition, TZDs, metformin, GLP-1RAs, and SGLT2is performed better than DPP-4is and NIDEs. TZDs were superior to AGIs (Table 2).

3.1.3. Body Mass Index

A total of 67 trials (4990 patients) provided data on BMI. After removing the PBO/LI effects, metformin and GLP-1RAs significantly reduced BMI by 1.28 kg/m2 (95% CI 0.31, 2.26) and 1.05 kg/m2 (95% CI 0.29, 1.81), while SUs and TZDs significantly increased BMI by 1.22 kg/m2 (95% CI 0.13, 2.31) and 0.63 kg/m2 (95% CI 0.26, 0.99), respectively. Across individual drugs, exenatide twice-daily produced a larger BMI loss (1.65 kg/m2; 95% CI 1.04, 2.26), while glimepiride yielded a larger BMI increase (1.79 kg/m2; 95% CI 0.46, 3.12). Smaller BMI increases were observed with rosiglitazone and alogliptin, in the order of 0.91 kg/m2 (95% CI 0.48, 1.35) and 0.81 kg/m2 (95% CI 0.27, 1.35). The other drugs had a neutral effect on BMI (Table 1; Supplementary Figures S18–S25).
Across the drug classes, metformin, GLP-1RAs, and AGIs produced significantly greater BMI reductions than DPP-4is, TZDs, and SUs, whilst SGLT2is were superior to SUs. The largest difference was seen between metformin and SUs (−2.50 kg/m2; 95% CI −3.96, −1.04), with the smallest difference seen between AGIs and DPP-4is (−0.96 kg/m2; 95% CI −1.87, −0.05) (Table 2).

3.1.4. Total Cholesterol

A total of 71 trials (12,014 patients) provided data on TC. Only GLP-1RAs, metformin, and AGIs reported significant TC reductions after removing the PBO/LI effects. The largest reductions were evident with GLP-1RAs (0.42 mmol/L; 95% CI 0.22, 0.61), ranging from 0.28 mmol/L (95% CI 0.06, 0.50) with exenatide twice-daily to 0.56 mmol/L (95% CI 0.34, 0.78) with liraglutide. AGIs conferred the smallest reductions (0.29 mmol/L; 95% CI 0.00, 0.58), although there were large differences within AGIs, with a 0.34 mmol/L (95% CI 0.03, 0.65) reduction with acarbose vs. no significant reduction with voglibose. Conversely, SGLT2is reported significant TC increases (0.22 mmol/L; 95% CI 0.13, 0.31), by 0.20 mmol/L (95% CI 0.06, 0.34) with empagliflozin and 0.30 mmol/L (95% CI 0.19, 0.40) with canagliflozin. Within DPP-4is, although alogliptin yielded TC loss (0.19 mmol/L; 95% CI 0.07, 0.31), sitagliptin produced TC increase (0.06 mmol/L; 95% CI 0.01, 0.11) (Table 1; Supplementary Figures S26–S33).
Across the drug classes, GLP-1RAs and metformin showed significantly greater TC reductions than DPP-4is, TZDs, and SGLT2is, ranging from 0.26 mmol/L (95% CI 0.08, 0.44) with metformin vs. DPP-4is to 0.64 mmol/L (95% CI 0.43, 0.86) with GLP-1RAs vs. SGLT2is. In addition, AGIs and DPP-4is were superior to SGLT2is (Table 2).

3.1.5. High Density Lipoprotein-Cholesterol

A total of 72 trials (12,578 patients) provided data on HDL-C. After removing the PBO/LI effects, the greatest HDL-C increases were seen with TZDs (0.12 mmol/L; 95% CI 0.07, 0.17); although certain intraclass differences were noted, with a 0.06 mmol/L (95% CI 0.03, 0.08) increase with rosiglitazone vs. 0.18 mmol/L (95% CI 0.09, 0.26) increase with pioglitazone. The smallest increase was evident with metformin (0.05 mmol/L; 95% CI 0.02, 0.09). With the exception of dapagliflozin, SGLT2is also increased HDL-C (0.09 mmol/L; 95% CI 0.07, 0.11), in the order of 0.08 mmol/L (95% CI 0.07, 0.10) with empagliflozin and 0.10 mmol/L (95% CI 0.07, 0.12) with canagliflozin. Although no significant HDL-C effect was seen with exenatide twice-daily, liraglutide showed a similar HDL-C increase to TZDs (Table 1; Supplementary Figures S34–S41).
Across the drug classes, TZDs produced significantly greater HDL-C increases than metformin, DPP-4is, and GLP-1RAs by 0.07~0.09 mmol/L. In addition, SGLT2is worked better than DPP-4is and GLP-1RAs (Table 2).

3.1.6. Systolic Blood Pressure

A total of 35 trials (9553 patients) provided data on SBP. Only SGLT2is and GLP-1RAs showed significant SBP reductions after removing the PBO/LI effects. SGLT2is produced relatively larger reductions (4.18 mmHg; 95% CI 3.53, 4.84), ranging from 3.29 mmHg (95% CI 2.25, 4.34) with empagliflozin to 5.36 mmHg (95% CI 4.14, 6.59) with canagliflozin. GLP-1RAs yielded smaller reductions (2.98 mmHg; 95% CI 1.30, 4.67), with reductions of 2.74 mmHg (95% CI 0.25, 5.24) with liraglutide vs. 3.19 mmHg (95% CI 0.90, 5.47) with exenatide twice-daily (Table 1; Supplementary Figures S42–S49).
Across the drug classes, SGLT2is conferred significantly greater SBP reductions than metformin, DPP-4is, and TZDs, ranging from 2.68 mmHg (95% CI 0.30, 5.06) vs. metformin to 4.96 mmHg (95% CI 1.74, 8.18) vs. TZDs. In addition, GLP-1RAs were superior to DPP-4is and TZDs (Table 2).

3.2. Hypoglycemia

A total of 103 trials (28,678 patients) provided data on hypoglycemia. The relative risk increases vs. PBO/LI were highest with SUs (5.44; 95% CI 2.11, 14.02), although only glipizide (7.11; 95% CI 2.18, 23.24) reported a significantly increased hypoglycemia risk. In addition, alogliptin (2.97; 95% CI 1.00, 8.77) significantly increased hypoglycemia risk, while empagliflozin (0.31; 95% CI 0.14, 0.71) reduced hypoglycemia risk compared with PBO/LI. The risks for other drugs were generally low (Table 1; Supplementary Figures S50–S57).
Across the drug classes, SUs showed significantly higher hypoglycemia risks than all other drug classes except NIDEs, by a RR ranging from 3.45 (95% CI 1.09, 10.91) vs. GLP-1RAs to 11.08 (95% CI 3.33, 36.87) vs. TZDs. In addition, metformin had higher risks than DPP-4is and TZDs. GLP-1RAs had higher risks than TZDs (Table 2).

3.3. Mortality and Vascular Outcomes

A total of 116 trials (24,976 patients) provided data on mortality, while most of them (101/116) reported that no death occurred during the trial. For the fatal events, most were considered by the investigator as not related to the study drug. A total of 32 trials (11,397 patients) provided data on vascular outcomes, 11 of which reported that no vascular events occurred. The main events reported were myocardial infarction (17 trials), heart failure (10 trials), stroke (9 trials), and diabetic nephropathy (9 trials). Compared with PBO/LI, all drugs had a neutral effect on mortality, total vascular events, myocardial infarction, heart failure, stroke, and diabetic nephropathy (Table 1; Supplementary Figures S58–S95).
Across the drug classes, there was no significant difference observed regarding mortality, total vascular events, myocardial infarction, heart failure, stroke, or diabetic nephropathy (Table 2; Supplementary Table S4).

3.4. Discontinuation

A total of 152 trials (34,569 patients) provided data on AE-induced discontinuations, 77 of which reported that no AE-induced discontinuation occurred during the trial. Overall, compared with PBO/LI, AGIs (2.57; 95% CI 1.64, 4.03) yielded significantly higher risk of AE-induced discontinuations, although large differences existed within AGIs, with 5.37 (95% CI 2.11, 13.69) times higher risk with miglitol vs. no risk increase with voglibose (0.92; 95% CI 0.19, 4.46). Conversely, linagliptin (0.55; 95% CI 0.30, 0.99) and empagliflozin (0.56; 95% CI 0.36, 0.87) significantly reduced the risk (Table 1; Supplementary Figures S96–S103).
Across the drug classes, AGIs conferred significantly higher risk of AE-induced discontinuations than TZDs (2.05; 95% CI 1.09, 3.85), metformin (2.49; 95% CI 1.43, 4.35), DPP-4is (2.80; 95% CI 1.70, 4.61), or SGLT2is (2.89; 95% CI 1.65, 5.07). No significant difference was seen across other comparisons (Table 2).

3.5. Sensitivity Analysis

The sensitivity analyses results did not significantly change the overall results (Supplementary Figures S104–S110).

4. Discussion

Evidence-based healthcare decision-making needs comparisons of all relevant competing therapies, on a full range of intermediate outcomes (e.g., blood glucose, blood lipids, and blood pressure), important AEs (e.g., hypoglycemia), and long-term outcomes (e.g., vascular events and mortality). This study identified important interclass and intraclass differences in these outcomes across all available glucose-lowering drugs when used as initial monotherapy for type 2 diabetes, to help decision makers rationally choose an alternative initial drug when metformin is contraindicated or intolerant.
The core of managing type 2 diabetes is to control blood glucose [3,4]. Intensive glucose control plays an important role in reducing the risks of diabetes-related vascular events and death [200]. This study showed that SUs generated the greatest HbA1c reductions, followed by GLP-1RAs, metformin, TZDs, SGLT2is, DPP-4is, AGIs, and NIDEs; and the HbA1c reductions observed were around the recommendation of clinical guidelines [3]. This is partly consistent with the results of Sherifali’s (2010) study, which found SUs to have greater HbA1c effect than metformin, TZDs, DPP-4is, AGIs, and NIDEs by synthesizing 61 English-language studies that compared an oral drug with placebo in patients with or without a background of other drugs [201]. Regarding FPG effects, SUs were also shown to have the largest FPG reductions, followed by TZDs, metformin, GLP-1RAs, SGLT2is, AGIs, DPP-4is, and NIDEs. In addition, we found the HbA1c reduction with liraglutide (1.17%) was more similar to SUs (1.39%) than other GLP-1RAs, such as lixisenatide (0.60%). We also found large differences in FPG reductions within GLP-1RAs, with 1.97 mmol/L for liraglutide vs. 0.97 mmol/L for lixisenatide. These findings confirmed important intraclass differences in glucose effect across drugs. Thus, optimal therapeutic decisions should also consider the comparative effects across individual drugs, especially when there is a need to choose from drugs belonging to the same classes.
Overweight or obesity is a common comorbidity of type 2 diabetes (85%) [202]. Weight gain may increase medical costs, while weight loss can improve cardiovascular risk factors, and reduce mortality and costs in patients with both type 2 diabetes and overweight/obesity [203,204,205,206]. This study reported that metformin and GLP-1RAs reduced BMI; while NIDEs, AGIs, DPP-4is, and SGLT2is maintained BMI; and SUs and TZDs increased BMI. The results were roughly consistent with that of Maruthur’s (2016) study, which compared the weight effect across monotherapies and metformin-based combinations of six drug classes from 85 English-language studies, and found weight was maintained or reduced with metformin, GLP-1RAs, DPP-4is, and SGLT2is, but increased with SUs and TZDs [207].
Dyslipidemia and/or hypertension, as independent risk factors for cardiovascular diseases, are also common comorbidities of type 2 diabetes (72%) [3,6]. Patients with type 2 diabetes, dyslipidemia, and hypertension are six times more likely to have cardiovascular diseases compared to those with diabetes alone [6]. Their integrated controls play important roles in reducing vascular events and costs, especially for those with long diabetes duration, old age, a history of cardiovascular diseases, or multiple risk factors [3]. This study showed that GLP-1RAs, AGIs, and metformin reduced TC, but SGLT2is increased TC; GLP-1RAs were superior to other drug classes in reducing TC. We also noted that within DPP-4is, alogliptin and sitagliptin showed opposite TC effects, confirming important intraclass differences across drugs. Regarding HDL-C effects, only TZDs, SGLT2is, and metformin increased HDL-C, and TZDs were the most efficacious drugs. This result is partly consistent with that of Bolen’s (2007) study, which found TZDs to have beneficial effects on HDL-C compared with other drugs by synthesizing 53 English-language studies on comparing monotherapies or combination therapies, although it did not include DPP-4is, SGLT2is, or GLP-1RAs [208].
Blood pressure lowering is related to improved mortality and other clinical outcomes for type 2 diabetes patients, and even a small reduction of 2.4 mmHg in SBP has an effect in reducing cardiovascular events [209,210]. As this study showed that SGLT2is and GLP-1RAs conferred SBP reductions by 2.74~5.36 mmHg, they may have beneficial effects on cardiovascular events. Our findings roughly correspond to that of Tsapas’s (2021) study, which reported that treatment with SGLT2is or GLP-1RAs with or without a metformin-based background therapy were more efficacious than other drug classes in reducing SBP by estimating 204 English-language studies [211].
A patient-centered approach recommends minimizing AEs, vascular events, and mortality beyond optimizing efficacy. Hypoglycemia as a common AE in treating type 2 diabetes is associated with multiple complications and high economic burden, and fear of it may reduce treatment adherence and prevent optimal glucose control [212,213]. This study showed that SUs had higher hypoglycemia risks than other drug classes, which is consistent with the previous studies [207,208]. In addition, large intraclass differences were noted regarding hypoglycemia risk. For example, within DPP-4is, alogliptin was reported to increase the risk but linagliptin reduced the risk. Overall, amongst all monotherapies, only GLP-1RAs produced a comprehensive beneficial effect on HbA1c, FPG, BMI, TC, and SBP, while not increasing hypoglycemia risk. As GLP-1RAs did not have a significant beneficial effect on HDL-C, while TZDs conferred the best effect on HDL-C, GLP-1RAs and TZDs may be the best mixed therapy to control HbA1c, FPG and lipid profile all together for patients with contraindications or intolerance to metformin. In terms of long-term outcomes, no significant difference was observed regarding total vascular events, myocardial infarction, heart failure, stroke, diabetic nephropathy, or mortality across the drug classes. This may be because when patients start the initial drug monotherapy they are most likely in the early stages of diabetes and may not develop serious vascular events in the short term. However, most of the included studies were short-term trials, lasting less than one year, thus with limited ability to observe long-term vascular outcomes and deaths.
Only a few systematic reviews were detected to have compared several drug classes, however, they were not focused on monotherapy, not covered all available drug classes, and only included English-language studies [201,207,208,211]. Thus, our study has noteworthy strengths in that it is the first attempt to comprehensively and simultaneously estimate and compare the treatment effects of all available glucose-lowering drugs when used as initial monotherapy for type 2 diabetes across multiple clinically meaningful intermediate and long-term outcomes, by merging data from English- and Chinese-language studies. The inclusion of 62 Chinese-language studies allowed the inclusion of populations and drugs that had not been studied previously in other settings. Our observations expanded the knowledge by supplementing with evidence from China. Our results may enable decision makers to better understand the similarities and differences across drugs, as to make comparisons between drugs at a glance when choosing an alternative monotherapy to metformin for type 2 diabetes patients.
Certain limitations should be acknowledged. First, from the methodological point of view, a meta-analysis has some weakness, including high risk of bias within studies, potential publication bias, and clinical heterogeneity. Second, most of the included trials were short-term trials, lasting less than one year, with limited ability to observe long-term clinical outcomes, such as vascular events and mortality. Ideally, as therapeutic decision-making should depend on long-term effectiveness, the generalizability of our results may be limited due to paucity of long-term evidence. Third, we only searched PubMed, Web of Science, Embase, CNKI, Chongqing VIP, and WanFang Data for eligible RCTs, thus RCTs indexed only in other databases (e.g., Scopus) may not be included in our review, which may induce potential bias.

5. Conclusions

Rational choice of drugs should be individualized, based on unique patient characteristics and benefit-risk profile of each drug. When choosing an alternative drug as initial monotherapy for type 2 diabetes patients with contraindications or intolerance to metformin, our results suggest a potential treatment hierarchy, with GLP-1RAs being preferred in terms of their favorable efficacy and safety profiles. In addition, SUs are most suggested for patients whose HbA1c or FPG is far away from the target, but least suggested for those wishing to minimize hypoglycemia. GLP-1RAs are most suggested for patients for whom BMI management or TC management is an emphasis. TZDs are most suggested for patients for whom HDL-C control is a priority. SGLT2is are most suggested for patients having a need to reduce SBP. While AGIs are least suggested for patients wishing to avoid AE-induced discontinuations. Our observations also corroborated the intraclass differences in treatment effects among drugs. Future studies should pay more attention to long-term studies to obtain more precise data on vascular outcomes and mortality. Moreover, this study also provided comparative and systematic clinical data for international researchers to conduct cost-effectiveness modelling studies, which will add long-term costs and effectiveness evidence for the rational choice of initial monotherapies alternative to metformin and improving the diabetes management as well as healthcare resource allocation.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm11237094/s1.

Author Contributions

S.G. contributed to the study design, data search and collection, statistical analysis, data interpretation, and writing and review of the manuscript. H.D. contributed to the study design, data interpretation, and review of the manuscript. X.H., X.S. and Y.G. contributed to the data collection, statistical analysis, and data interpretation. L.S. and X.Z. contributed to review of the manuscript. M.H. contributed to the data collection and data interpretation. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by National Natural Science Foundation of China (72104102), Jiangsu Planning Office of Philosophy and Social Science (20JD001), and the Fundamental Research Funds for the Central Universities, NJU (011714370119).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The datasets used and/or analyzed during the current study are available from the corresponding authors on reasonable request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. International Diabetes Federation. IDF DIABETES ATLAS Ninth Edition 2019. Available online: https://diabetesatlas.org/upload/resources/material/20200302_133351_IDFATLAS9e-final-web.pdf (accessed on 3 May 2021).
  2. International Diabetes Federation. About Diabetes: Types of Diabetes. Available online: https://www.idf.org/aboutdiabetes/what-is-diabetes/types-of-diabetes.html (accessed on 1 March 2018).
  3. Jia, W.; Weng, J.; Zhu, D.; Ji, L.; Lu, J.; Zhou, Z.; Zou, D.; Guo, L.; Ji, Q.; Chen, L.; et al. Standards of medical care for type 2 diabetes in China 2019. Diabetes/Metab. Res. Rev. 2019, 35, e3158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. American Diabetes Association. Pharmacologic Approaches to Glycemic Treatment: Standards of Medical Care in Diabetes—2021. Diabetes Care 2021, 44 (Suppl. S1), S111–S124. [Google Scholar] [CrossRef] [PubMed]
  5. Ji, L.; Lu, J.; Weng, J.; Jia, W.; Tian, H.; Zhu, D.; Xing, X.; Guo, L. China type 2 diabetes treatment status survey of treatment pattern of oral drugs users. J. Diabetes 2015, 7, 166–173. [Google Scholar] [CrossRef] [PubMed]
  6. Ji, L.; Hu, D.; Pan, C.; Weng, J.; Huo, Y.; Ma, C.; Mu, Y.; Hao, C.; Ji, Q.; Ran, X.; et al. Primacy of the 3B Approach to Control Risk Factors for Cardiovascular Disease in Type 2 Diabetes Patients. Am. J. Med. 2013, 126, 925.e11–925.e22. [Google Scholar] [CrossRef]
  7. Davies, M.J.; D’Alessio, D.A.; Fradkin, J.; Kernan, W.N.; Mathieu, C.; Mingrone, G.; Rossing, P.; Tsapas, A.; Wexler, D.J.; Buse, J.B. Management of Hyperglycemia in Type 2 Diabetes, 2018. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care. 2018, 41, 2669–2701. [Google Scholar] [CrossRef] [Green Version]
  8. Liberati, A.; Altman, D.G.; Tetzlaff, J.; Mulrow, C.; Gotzsche, P.C.; Ioannidis, J.P.A.; Clarke, M.; Devereaux, P.J.; Kleijnen, J.; Moher, D. The PRISMA Statement for Reporting Systematic Reviews and Meta-Analyses of Studies That Evaluate Healthcare Interventions: Explanation and Elaboration. BMJ 2009, 339, b2700. [Google Scholar] [CrossRef] [Green Version]
  9. Higgins, J.P.T.; Green, S. Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [Updated March 2011]. Available online: http://handbook-5-1.cochrane.org/ (accessed on 1 June 2017).
  10. Bucher, H.C.; Guyatt, G.H.; Griffith, L.E.; Walter, S.D. The results of direct and indirect treatment comparisons in meta-analysis of randomized controlled trials. J. Clin. Epidemiol. 1997, 50, 683–691. [Google Scholar] [CrossRef]
  11. Song, F.; Altman, D.G.; Glenny, A.-M.; Deeks, J. Validity of indirect comparison for estimating efficacy of competing interventions: Empirical evidence from published meta-analyses. BMJ 2003, 326, 472. [Google Scholar] [CrossRef] [Green Version]
  12. Eng, C.; Kramer, C.K.; Zinman, B.; Retnakaran, R. Glucagon-like peptide-1 receptor agonist and basal insulin combination treatment for the management of type 2 diabetes: A systematic review and meta-analysis. Lancet 2014, 384, 2228–2234. [Google Scholar] [CrossRef]
  13. Musso, G.; Gambino, R.; Cassader, M.; Paschetta, E. Efficacy and safety of dual SGLT 1/2 inhibitor sotagliflozin in type 1 diabetes: Meta-analysis of randomised controlled trials. BMJ 2019, 365, l1328. [Google Scholar] [CrossRef]
  14. Higgins, J.P.T.; Thompson, S.G.; Deeks, J.J.; Altman, D.G. Measuring inconsistency in meta-analyses. BMJ 2003, 327, 557–560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Ji, L.; Han, P.; Wang, X.; Liu, J.; Zheng, S.; Jou, Y.; O’Neill, E.A.; Golm, G.T.; Engel, S.S.; Kaufman, K.D.; et al. Randomized clinical trial of the safety and efficacy of sitagliptin and metformin co-administered to Chinese patients with type 2 diabetes mellitus. J. Diabetes Investig. 2016, 7, 727–736. [Google Scholar] [CrossRef] [PubMed]
  16. Hassan, M.; Abd-Allah, G.M. Effects of metformin plus gliclazide versus metformin plus glimepiride on cardiovascular risk factors in patients with type 2 diabetes mellitus. Pak. J. Pharm. Sci. 2015, 28, 1723–1730. [Google Scholar] [PubMed]
  17. Pratley, R.E.; Fleck, P.; Wilson, C. Efficacy and safety of initial combination therapy with alogliptin plus metformin versus either as monotherapy in drug-naïve patients with type 2 diabetes: A randomized, double-blind, 6-month study. Diabetes Obes. Metab. 2014, 16, 613–621. [Google Scholar] [CrossRef]
  18. Ferrannini, E.; Seman, L.; Seewaldt-Becker, E.; Hantel, S.; Pinnetti, S.; Woerle, H.J. A Phase IIb, randomized, placebo-controlled study of the SGLT2 inhibitor empagliflozin in patients with type 2 diabetes. Diabetes Obes. Metab. 2013, 15, 721–728. [Google Scholar] [CrossRef]
  19. Haak, T.; Meinicke, T.; Von Eynatten, M.; Woerle, H.-J.; Jones, R.; Weber, S. Initial combination of linagliptin and metformin improves glycaemic control in type 2 diabetes: A randomized, double-blind, placebo-controlled study. Diabetes Obes. Metab. 2012, 14, 565–574. [Google Scholar] [CrossRef]
  20. Chakraborty, A.; Chowdhury, S.; Bhattacharyya, M. Effect of metformin on oxidative stress, nitrosative stress and inflammatory biomarkers in type 2 diabetes patients. Diabetes Res. Clin. Pract. 2011, 93, 56–62. [Google Scholar] [CrossRef]
  21. List, J.F.; Woo, V.; Morales, E.; Tang, W.; Fiedorek, F.T. Sodium-Glucose Cotransport Inhibition with Dapagliflozin in Type 2 Diabetes. Diabetes Care 2009, 32, 650–657. [Google Scholar] [CrossRef] [Green Version]
  22. Goldstein, B.J.; Feinglos, M.N.; Lunceford, J.K.; Johnson, J.; Williams-Herman, D.E. Effect of Initial Combination Therapy with Sitagliptin, a Dipeptidyl Peptidase-4 Inhibitor, and Metformin on Glycemic Control in Patients with Type 2 Diabetes. Diabetes Care 2007, 30, 1979–1987. [Google Scholar] [CrossRef] [Green Version]
  23. Wang, L.; Ning, J. Effects of metformin on C-reactive protein and complement factor C3 in patients of obese type 2 diabetes mellitus. Shanxi Med. J. 2006, 35, 888–889. [Google Scholar]
  24. Fujioka, K.; Brazg, R.L.; Raz, I.; Bruce, S.; Joyal, S.; Swanink, R.; Pans, M. Efficacy, dose-response relationship and safety of once-daily extended-release metformin (GlucophageR XR) in type 2 diabetic patients with inadequate glycaemic control despite prior treatment with diet and exercise: Results from two double-blind, placebo-controlled studies. Diabetes Obes. Metab. 2005, 7, 28–39. [Google Scholar] [CrossRef] [PubMed]
  25. Karlsson, H.K.; Hallsten, K.; Bjornholm, M.; Tsuchida, H.; Chibalin, A.V.; Virtanen, K.A.; Heinonen, O.J.; Lonnqvist, F.; Nuutila, P.; Zierath, J.R. Effects of metformin and rosiglitazone treatment on insulin signaling and glucose uptake in patients with newly diagnosed type 2 diabetes: A randomized controlled study. Diabetes 2005, 54, 1459–1467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Viljanen, A.P.M.; Virtanen, K.A.; Järvisalo, M.J.; Hällsten, K.; Parkkola, R.; Rönnemaa, T.; Lönnqvist, F.; Iozzo, P.; Ferrannini, E.; Nuutila, P. Rosiglitazone Treatment Increases Subcutaneous Adipose Tissue Glucose Uptake in Parallel with Perfusion in Patients with Type 2 Diabetes: A Double-Blind, Randomized Study with Metformin. J. Clin. Endocrinol. Metab. 2005, 90, 6523–6528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Horton, E.S.; Foley, J.E.; Shen, S.G.; Baron, M.A. Efficacy and tolerability of initial combination therapy with nateglinide and metformin in treatment-naïve patients with type 2 diabetes. Curr. Med. Res. Opin. 2004, 20, 883–889. [Google Scholar] [CrossRef]
  28. Manzella, D.; Grella, R.; Esposito, K.; Giugliano, D.; Barbagallo, M.; Paolisso, G. Blood pressure and cardiac autonomic nervous system in obese type 2 diabetic patients: Effect of metformin administration. Am. J. Hypertens. 2004, 17, 223–227. [Google Scholar] [CrossRef]
  29. Del Prato, S.; Erkelens, D.W.; Leutenegger, M. Six-month efficacy of benfluorex vs. placebo or metformin in diet-failed type 2 diabetic patients. Acta Diabetol. 2003, 40, 20–27. [Google Scholar] [CrossRef]
  30. Chiasson, J.-L.; Naditch, L.; for the Miglitol Canadian University Investigator Group. The Synergistic Effect of Miglitol Plus Metformin Combination Therapy in the Treatment of Type 2 Diabetes. Diabetes Care 2001, 24, 989–994. [Google Scholar] [CrossRef] [Green Version]
  31. Mather, K.J.; Verma, S.; Anderson, T.J. Improved endothelial function with metformin in type 2 diabetes mellitus. J. Am. Coll. Cardiol. 2001, 37, 1344–1350. [Google Scholar] [CrossRef] [Green Version]
  32. Wu, G.T.; Han, Y.Q.; Wang, J.Y.; Li, W.J.; Gu, Y.; Shi, Y. Effects of Glucophage on insulin resistance in type 2 diabetes mellitus patients. J. Tongji Univ. (Med. Sci.) 2001, 22, 31–32+35. [Google Scholar]
  33. Lee, A.; Morley, J.E. Metformin Decreases Food Consumption and Induces Weight Loss in Subjects with Obesity with Type II Non-Insulin-Dependent Diabetes. Obes. Res. 1998, 6, 47–53. [Google Scholar] [CrossRef]
  34. Hoffmann, J.; Spengler, M. Efficacy of 24-Week Monotherapy with Acarbose, Metformin, or Placebo in Dietary-Treated NIDDM Patients: The Essen-II Study. Am. J. Med. 1997, 103, 483–490. [Google Scholar] [CrossRef] [PubMed]
  35. Wang, D.L.; Xian, B. Effect of pioglitazone and metformin on patients with incipient type 2 diabetes and nonalcoholic fatty liver disease. Chin. Baby 2020, 4, 122. [Google Scholar]
  36. Yang, Y. Application study of metformin combined with individualized diet and exercise therapy in elderly patients with type 2 diabetes. Diet Health 2019, 6, 45–46. [Google Scholar]
  37. Han, S.Q.; Liu, R.; Liu, J.X.; Geng, Y.Q. Effects of metformin on hepatic inflammation, liver fibrosis and insulin resistance in patients with type 2 diabetes mellitus complicated with nonalcoholic fatty liver disease. Hebei Med. J. 2018, 40, 1769–1773. [Google Scholar]
  38. Zhou, R.F.; Zhong, S.; Guo, Y.; Sun, Y.J.; Shao, Z.X. Impact of exenatide combined with metformin on islet function in new-onset type 2 diabetes mellitus. Clin. Focus 2017, 32, 965–968. [Google Scholar]
  39. Bai, N. The efficacy and side effects of metformin in the treatment of type 2 diabetes mellitus complicated with nonalcoholic fatty liver disease. China Health Stand. Manag. 2016, 7, 78–80. [Google Scholar]
  40. Geng, N.; Qi, H.; Liu, J.Q. Efficacy of rosuvastatin in male patients with newly diagnosed type 2 diabetes and hyperlipidemia and its effect on serum vaspin level. Hebei Med. J. 2016, 38, 2775–2777, 2781. [Google Scholar]
  41. Guo, D.L. Analysis of the efficacy of metformin in the treatment of type 2 diabetes. Diabetes New World 2014, 11, 1–2. [Google Scholar]
  42. Guo, W.; Gao, M.S.; Ye, Z.H.; Li, Y.; Tu, J.J.; Lei, W.M. Effects of metformin on the serum nesfatin-1 and liver steatosis in type 2 diabetic patients with nonalcoholic fatty liver disease. Chin. J. Difficult Complicat. Cases 2014, 13, 374–377. [Google Scholar]
  43. Li, L.; Zhu, K.S.; Qu, J.C.; Xia, A.X.; Zhang, W. Clinical effect of liraglutide and metformin hydrochioride on overweight diabetic patients with poor glycemic control. Clin. Med. China 2014, 30, 67–69. [Google Scholar]
  44. Esteghamati, A.; Eskandari, D.; Mirmiranpour, H.; Noshad, S.; Mousavizadeh, M.; Hedayati, M.; Nakhjavani, M. Effects of metformin on markers of oxidative stress and antioxidant reserve in patients with newly diagnosed type 2 diabetes: A randomized clinical trial. Clin. Nutr. 2013, 32, 179–185. [Google Scholar] [CrossRef] [PubMed]
  45. Liu, Y.T.; Hu, B.; Jian, L.; Li, J.W.; Huang, L. Influence of metformin on the level of serum vaspin in T2DM patients. Chin. J. Diabetes 2013, 21, 705–708. [Google Scholar]
  46. Xi, Y. Efficacy of liraglutide in patients with new-onset type 2 diabetes and obesity and its impact on patients’ micro-inflammation. Chin. Gen. Pract. 2013, 16, 3339–3340, 3345. [Google Scholar]
  47. Yang, W.C.; Dang, Y.; Qiao, L.; Wang, H.; Li, C.Q.; Dang, X.Y. Comparison of efficacy of exercise therapy and metformin in patients with newly diagnosed type 2 diabetes. J. Henan Norm. Univ. (Nat. Sci. Ed.) 2013, 41, 122–126. [Google Scholar]
  48. Qu, J.C.; Zhu, K.S.; Wang, T.; Li, L.; Zhao, L. Effect of rosiglitazone and metformin on lipid metabolism in patients with type 2 diabetes. China Med. 2011, 6, 286–287. [Google Scholar]
  49. Nar, A.; Gedik, O. The effect of metformin on leptin in obese patients with type 2 diabetes mellitus and nonalcoholic fatty liver disease. Geol. Rundsch. 2009, 46, 113–118. [Google Scholar] [CrossRef] [PubMed]
  50. Chen, Y. Study on the Plasma Visfatin Level in Newly Diagnosed Obese Type 2 Diabetes Patients and the Influence of Pioglitazone on It; Guangdong Medical University: Zhanjiang, China, 2008. [Google Scholar]
  51. Cao, Y. Clinical Study on Multiple Factor Interventions in Improving the Insulin Resistance in Type 2 Diabetes; Shandong University of Traditional Chinese Medicine: Jinan, China, 2007. [Google Scholar]
  52. Deng, H.O.; Lin, K.; Li, D.F.; Li, Y.L. Effect of metformin on serum testosterone in male patients with type 2 diabetes. Guangdong Med. J. 2007, 28, 601–602. [Google Scholar]
  53. Li, H.Z.; Zhang, C.; Wang, M.; Liu, X.H. Effect of metformin and rosiglitazone maleate on lipid metabolism in patients with type 2 diabetes. J. Clin. Res. 2007, 24, 319–321. [Google Scholar]
  54. Mei, Q. Therapeutic effect of metformin on type 2 diabetes mellitus complicated with nonalcoholic fatty liver disease. Pract. Pharm. Clin. Remedies 2006, 9, 346–347. [Google Scholar]
  55. Hanefeld, M.; Haffner, S.; Menschikowski, M.; Koehler, C.; Temelkova-Kurktschiev, T.; Wildbrett, J.; Fischer, S. Different effects of acarbose and glibenclamide on proinsulin and insulin profiles in people with Type 2 diabetes. Diabetes Res. Clin. Pract. 2002, 55, 221–227. [Google Scholar] [CrossRef]
  56. Birkeland, K.I.; Furuseth, K.; Melander, A.; Mowinckel, P.; Vaaler, S. Long-Term Randomized Placebo-Controlled Double-Blind Therapeutic Comparison of Glipizide and Glyburide: Glycemic control and insulin secretion during 15 months. Diabetes Care 1994, 17, 45–49. [Google Scholar] [CrossRef] [PubMed]
  57. Takami, K.; Takeda, N.; Nakashima, K.; Takami, R.; Hayashi, M.; Ozeki, S.; Yamada, A.; Kokubo, Y.; Sato, M.; Kawachi, S.-I.; et al. Effects of Dietary Treatment Alone or Diet with Voglibose or Glyburide on Abdominal Adipose Tissue and Metabolic Abnormalities in Patients with Newly Diagnosed Type 2 Diabetes. Diabetes Care 2002, 25, 658–662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Kaku, K.; Araki, T.; Yoshinaka, R. Randomized, Double-Blind, Dose-Ranging Study of TAK-875, a Novel GPR40 Agonist, in Japanese Patients with Inadequately Controlled Type 2 Diabetes. Diabetes Care 2013, 36, 245–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Bautista, J.L.; Bugos, C.; Dirnberger, G.; Atherton, T. Efficacy and safety profile of glimepiride in Mexican American Patients with type 2 diabetes mellitus: A randomized, placebo-controlled study. Clin. Ther. 2003, 25, 194–209. [Google Scholar] [CrossRef] [PubMed]
  60. Yu, J.X.; Wu, Q.; Zhou, L.L. Efficacy of simvastatin in treatment of patients with type 2 diabetes and dyslipidemia and its effect on serum vaspin level. Chin. J. Gerontol. 2014, 34, 6588–6589. [Google Scholar]
  61. Wei, J.C.; Ruan, D.J.; Meng, X.Z.; Qu, S.H.; Zheng, W.W. Effect of glipizide on blood glucose and blood lipids in patients with type 2 diabetes. Prog. Mod. Biomed. 2010, 10, 3067–3069. [Google Scholar]
  62. Scott, R.; Wu, M.; Sanchez, M.; Stein, P. Efficacy and tolerability of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy over 12 weeks in patients with type 2 diabetes. Int. J. Clin. Pract. 2007, 61, 171–180. [Google Scholar] [CrossRef]
  63. Simonson, D.C.; Kourides, I.A.; Feinglos, M.; Shamoon, H.; Fischette, C.T.; Aronoff, S.; Blonde, L.; Cefalu, W.; Clement, S.; Greenberg, C.; et al. Efficacy, safety, and dose-response characteristics of glipizide gastrointestinal therapeutic system on glycemic control and insulin secretion in NIDDM-Results of two multicenter, randomized, placebo-controlled clinical trials. Diabetes Care 1997, 20, 597–606. [Google Scholar] [CrossRef]
  64. Gul, O.O.; Tuncel, E.; Yilmaz, Y.; Ulukaya, E.; Gul, C.B.; Kiyici, S.; Oral, A.Y.; Guclu, M.; Ersoy, C.; Imamoglu, S. Comparative effects of pioglitazone and rosiglitazone on plasma levels of soluble receptor for advanced glycation end products in type 2 diabetes mellitus patients. Metabolism 2010, 59, 64–69. [Google Scholar] [CrossRef]
  65. Rahman, S.; Ismail, A.A.S.; Ismail, S.B.; Naing, N.N.; Rahman, A.R.A. Effect of rosiglitazone and ramipril on macrovasculopathy in patients with type 2 diabetes: Needs longer treatment and/or higher doses? Clin. Pharmacol. 2010, 2, 83–87. [Google Scholar] [CrossRef]
  66. Oz, O.; Tuncel, E.; Eryilmaz, S.; Fazlioglu, M.; Gul, C.B.; Ersoy, C.; Ocak, N.; Dirican, M.; Cangur, S.; Baran, I.; et al. Arterial elasticity and plasma levels of adiponectin and leptin in type 2 diabetic patients treated with thiazolidinediones. Endocrine 2008, 33, 101–105. [Google Scholar] [CrossRef]
  67. Albertini, J.-P.; McMorn, S.O.; Chen, H.; Mather, R.A.; Valensi, P. Effect of rosiglitazone on factors related to endothelial dysfunction in patients with type 2 diabetes mellitus. Atherosclerosis 2007, 195, e159–e166. [Google Scholar] [CrossRef]
  68. Gastaldelli, A.; Miyazaki, Y.; Pettiti, M.; Santini, E.; Ciociaro, D.; DeFronzo, R.A.; Ferrannini, E. The Effect of Rosiglitazone on the Liver: Decreased Gluconeogenesis in Patients with Type 2 Diabetes. J. Clin. Endocrinol. Metab. 2006, 91, 806–812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Chen, H.Y.; Zhang, Y.; Wu, Y. Effects of rosiglitazone on C-reactive protein in patients with type 2 diabetes. J. Jilin Univ. (Med. Ed.) 2004, 30, 465–467. [Google Scholar]
  70. Juhl, C.B.; Hollingdal, M.; Pørksen, N.; Prange, A.; Lönnqvist, F.; Schmitz, O. Influence of Rosiglitazone Treatment on β-Cell Function in Type 2 Diabetes: Evidence of an Increased Ability of Glucose to Entrain High-Frequency Insulin Pulsatility. J. Clin. Endocrinol. Metab. 2003, 88, 3794–3800. [Google Scholar] [CrossRef] [Green Version]
  71. Carey, D.G.; Cowin, G.J.; Galloway, G.J.; Jones, N.P.; Richards, J.C.; Biswas, N.; Doddrell, D.M. Effect of rosiglitazone on insulin sensitivity and body composition in type 2 diabetic patients [corrected]. Obes. Res. 2002, 10, 1008–1015. [Google Scholar] [CrossRef] [PubMed]
  72. Haffner, S.M.; Greenberg, A.S.; Weston, W.M.; Chen, H.; Williams, K.; Freed, M.I. Effect of rosiglitazone treatment on nontraditional markers of cardiovascular disease in patients with type 2 diabetes mellitus. Circulation 2002, 106, 679–684. [Google Scholar] [CrossRef] [Green Version]
  73. Lebovitz, H.E.; Dole, J.F.; Patwardhan, R.; Rappaport, E.B.; Freed, M.I. Rosiglitazone monotherapy is effective in patients with type 2 diabetes. J. Clin. Endocrinol. Metab. 2001, 86, 280–288. [Google Scholar] [CrossRef]
  74. Miyazaki, Y.; Glass, L.; Triplitt, C.; Matsuda, M.; Cusi, K.; Mahankali, A.; Mahankali, S.; Mandarino, L.J.; DeFronzo, R.A. Effect of rosiglitazone on glucose and non-esterified fatty acid metabolism in Type II diabetic patients. Diabetologia 2001, 44, 2210–2219. [Google Scholar] [CrossRef] [Green Version]
  75. Patel, J.; Anderson, R.J.; Rappaport, E.B. Rosiglitazone monotherapy improves glycaemic control in patients with type 2 diabetes: A twelve-week, randomized, placebo-controlled study. Diabetes Obes. Metab. 1999, 1, 165–172. [Google Scholar] [CrossRef]
  76. Huang, D.; Wei, X.; Li, Z.Y.; Qu, W.J.; Wang, C.P.; Chen, Y.M.; Chen, D.Z. Effect of rosiglitazone on serum SPARC in patients with newly diagnosed type 2 diabetes mellitus. Mod. Med. Health 2016, 32, 1284–1286, 1289. [Google Scholar]
  77. Berberoglu, Z.; Yazici, A.C.; Demirag, N.G. ORIGINAL ARTICLE: Effects of rosiglitazone on bone mineral density and remodelling parameters in Postmenopausal diabetic women: A 2-year follow-up study. Clin. Endocrinol. 2010, 73, 305–312. [Google Scholar] [CrossRef] [PubMed]
  78. Lu, X.H.; Feng, Y.X.; Wu, X.M.; Bian, Q.X.; Zhang, J.H. Effect of aerobic exercise on blood glucose and blood lipids in obese patients with type 2 diabetes. China Foreign Med. Treat. 2009, 28, 53–54. [Google Scholar]
  79. Zhu, K.S.; Wang, P. Effect of rosiglitazone on type 2 diabetes with postprandial hypoglycemia as the first symptom. J. Pract. Diabetol. 2009, 5, 46–47. [Google Scholar]
  80. Berberoglu, Z.; Gursoy, A.; Bayraktar, N.; Yazici, A.C.; Tutuncu, N.B.; Demirag, N.G. Rosiglitazone Decreases Serum Bone-Specific Alkaline Phosphatase Activity in Postmenopausal Diabetic Women. J. Clin. Endocrinol. Metab. 2007, 92, 3523–3530. [Google Scholar] [CrossRef] [PubMed]
  81. Wang, J. Clinical comparison of pioglitazone hydrochloride in the treatment of patients with incipient type 2 diabetes. China Health Vis. 2020, 1, 100–101. [Google Scholar]
  82. Bajpeyi, S.; Pasarica, M.; Conley, K.E.; Newcomer, B.R.; Jubrias, S.A.; Gamboa, C.; Murray, K.; Sereda, O.; Sparks, L.M.; Smith, S.R. Pioglitazone-induced improvements in insulin sensitivity occur without concomitant changes in muscle mitochondrial function. Metabolism 2017, 69, 24–32. [Google Scholar] [CrossRef] [PubMed]
  83. Tang, X.Q. Observation on the effect of pioglitazone hydrochloride monotherapy in the treatment of elderly patients with incipient type 2 diabetes. Chin. J. Clin. Ration. Drug Use 2017, 10, 71–72. [Google Scholar]
  84. Sykes, A.P.; Kemp, G.L.; Dobbins, R.; O’Connor-Semmes, R.; Almond, S.R.; Wilkison, W.O.; Walker, S.; Kler, L. Randomized efficacy and safety trial of once-daily remogliflozin etabonate for the treatment of type 2 diabetes. Diabetes Obes. Metab. 2015, 17, 98–101. [Google Scholar] [CrossRef]
  85. Sykes, A.P.; O’Connor-Semmes, R.; Dobbins, R.; Dorey, D.J.; Lorimer, J.D.; Walker, S.; Wilkison, W.O.; Kler, L. Randomized trial showing efficacy and safety of twice-daily remogliflozin etabonate for the treatment of type 2 diabetes. Diabetes Obes. Metab. 2015, 17, 94–97. [Google Scholar] [CrossRef]
  86. Chou, H.S.; Truitt, K.E.; Moberly, J.B.; Merante, D.; Choi, Y.; Mun, Y.; Pfützner, A. A 26-week, placebo- and pioglitazone-controlled monotherapy study of rivoglitazone in subjects with type 2 diabetes mellitus. Diabetes Obes. Metab. 2012, 14, 1000–1009. [Google Scholar] [CrossRef] [PubMed]
  87. Kong, A.P.S.; Yamasaki, A.; Ozaki, R.; Saito, H.; Asami, T.; Ohwada, S.; Ko, G.T.C.; Wong, C.K.; Leung, G.T.C.; Lee, K.F.; et al. A randomized-controlled trial to investigate the effects of rivoglitazone, a novel PPAR gamma agonist on glucose-lipid control in type 2 diabetes. Diabetes Obes. Metab. 2011, 13, 806–813. [Google Scholar] [CrossRef] [PubMed]
  88. Wang, B. Clinical observation of pioglitazone hydrochloride on 136 patients with incipient type 2 diabetes. China Medical Herald. 2009, 6, 58–59. [Google Scholar]
  89. Gastaldelli, A.; Casolaro, A.; Pettiti, M.; Nannipieri, M.; Ciociaro, D.; Frascerra, S.; Buzzigoli, E.; Baldi, S.; Mari, A.; Ferrannini, E. Effect of Pioglitazone on the Metabolic and Hormonal Response to a Mixed Meal in Type II Diabetes. Clin. Pharmacol. Ther. 2007, 81, 205–212. [Google Scholar] [CrossRef] [PubMed]
  90. Goldstein, B.J.; Rosenstock, J.; Anzalone, D.; Tou, C.; Ohman, K.P. Effect of tesaglitazar, a dual PPAR alpha/gamma agonist, on glucose and lipid abnormalities in patients with type 2 diabetes: A 12-week dose-ranging trial. Curr. Med. Res. Opin. 2006, 22, 2575–2590. [Google Scholar] [CrossRef] [PubMed]
  91. Hao, Y.R.; Han, Q.W.; Yuan, W.T.; He, X.J. Effects of pioglitazone on elderly patients with type 2 diabetes mellitus. Med. J. Wuhan Univ. 2006, 27, 104–107. [Google Scholar]
  92. Khan, M.; Murray, F.T.; Karunaratne, M.; Perez, A. Pioglitazone and reductions in post-challenge glucose levels in patients with type 2 diabetes. Diabetes Obes. Metab. 2006, 8, 31–38. [Google Scholar] [CrossRef]
  93. Pang, W.Y.; Liu, Y.; Zhao, C.Y. Pioglitazone monotherapy for elderly patients with new-onset type 2 diabetes mellitus. Cent. Plains Med. J. 2006, 33, 6–7. [Google Scholar]
  94. Wallace, T.M.; Levy, J.C.; Matthews, D.R. An increase in insulin sensitivity and basal beta-cell fuction in diabetic subjects treated with pioglitazone in a placebo-controlled randomized study. Diabet. Med. 2004, 21, 568–576. [Google Scholar] [CrossRef]
  95. Liu, C.M. Observation of clinical effects of pioglitazone hydrochlodde in treatment of type 2 diabetic patients. J. Med. Forum. 2003, 24, 5–6. [Google Scholar]
  96. Miyazaki, Y.; Matsuda, M.; DeFronzo, R.A. Dose-Response Effect of Pioglitazone on Insulin Sensitivity and Insulin Secretion in Type 2 Diabetes. Diabetes Care 2002, 25, 517–523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Scherbaum, W.A.; Göke, B.; and the German Pioglitazone Study Group. Metabolic Efficacy and Safety of Once-Daily Pioglitazone Monotherapy in Patients with Type 2 Diabetes: A Double-Blind, Placebo-Controlled Study. Horm. Metab. Res. 2002, 34, 589–595. [Google Scholar] [CrossRef] [PubMed]
  98. Aronoff, S.; Rosenblatt, S.; Braithwaite, S.; Egan, J.W.; Mathisen, A.L.; Schneider, R.L. Pioglitazone hydrochloride monotherapy improves glycemic control in the treatment of patients with type 2 diabetes: A 6-month randomized placebo-controlled dose-response study. The Pioglitazone 001 Study Group. Diabetes Care 2000, 23, 1605–1611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Mirmiranpour, H.; Mousavizadeh, K.; Noshad, S.; Ghavami, M.; Ebadi, M.; Ghasemiesfe, M.; Nakhjavani, M.; Esteghamati, A. Comparative effects of pioglitazone and metformin on oxidative stress markers in newly diagnosed type 2 diabetes patients: A randomized clinical trial. J. Diabetes Its Complicat. 2013, 27, 501–507. [Google Scholar] [CrossRef]
  100. Bech, P.; Moses, R.; Gomis, R. The effect of prandial glucose regulation with repaglinide on treatment satisfaction, wellbeing and health status in patients with pharmacotherapy-naïve type 2 diabetes: A placebo-controlled, multicentre study. Qual. Life Res. 2003, 12, 413–425. [Google Scholar] [CrossRef]
  101. Xu, L. Efficacy of repaglinide on 31 patients with type 2 diabetes. J. Aerosp. Med. 2011, 22, 458–459. [Google Scholar]
  102. Zhong, X.Y.; Lv, L.L.; Chen, Z.H.; Xu, Z.H. The effect of repaglinide on 31 patients with type 2 diabetes. Guide China Med. 2010, 8, 200–201. [Google Scholar]
  103. Guo, X.D. Efficacy of repaglinide on 31 patients with type 2 diabetes. Guide China Med. 2009, 7, 83–84. [Google Scholar]
  104. Zhao, W.; Zhuang, L.F.; Peng, L.Y.; Luo, J.L. Observation on the efficacy of repaglinide in treatment of type 2 diabetes. World Health Dig. 2008, 5, 71–73. [Google Scholar]
  105. González-Clemente, J.-M.; on behalf of the Spanish Nateglinide Study Group. Improvement of glycaemic control by nateglinide decreases systolic blood pressure in drug-naive patients with type 2 diabetes. Eur. J. Clin. Investig. 2008, 38, 174–179. [Google Scholar] [CrossRef]
  106. Schwarz, S.L.; Gerich, J.E.; Marcellari, A.; Jean-louis, L.; Purkayastha, D.; Baron, M.A. Nateglinide, alone or in combination with metformin, is effective and well tolerated in treatment-naïve elderly patients with type 2 diabetes. Diabetes Obes. Metab. 2008, 10, 652–660. [Google Scholar] [CrossRef] [PubMed]
  107. Mari, A.; Gastaldelli, A.; Foley, J.E.; Pratley, R.E.; Ferrannini, E. β-Cell function in mild type 2 diabetic patients: Effects of 6-month glucose lowering with nateglinide. Diabetes Care 2005, 28, 1132–1138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Kato, T.; Inoue, T.; Node, K. Postprandial endothelial dysfunction in subjects with new-onset type 2 diabetes: An acarbose and nateglinide comparative study. Cardiovasc. Diabetol. 2010, 9, 12. [Google Scholar] [CrossRef] [Green Version]
  109. Kirkman, M.S.; Shankar, R.R.; Shankar, S.; Shen, C.; Brizendine, E.; Baron, A.; McGill, J. Treating postprandial hyperglycemia does not appear to delay progression of early type 2 diabetes: The Early Diabetes Intervention Program. Diabetes Care 2006, 29, 2095–2101. [Google Scholar] [CrossRef] [PubMed]
  110. Josse, R.G.; Chiasson, J.-L.; Ryan, E.A.; Lau, D.C.W.; Ross, S.A.; Yale, J.-F.; Leiter, L.A.; Maheux, P.; Tessier, D.; Wolever, T.M.S.; et al. Acarbose in the treatment of elderly patients with type 2 diabetes. Diabetes Res. Clin. Pract. 2003, 59, 37–42. [Google Scholar] [CrossRef]
  111. Wu, G.T.; Han, Y.Q.; Yu, Y.C.; Teng, X.H.; Zhang, J.C.; Qu, F.J. Influence of acarbose on insulin resistant of patients with type 2 diabetes mellitus. Chin. J. New Drugs Clin. Remedies 2003, 22, 535–538. [Google Scholar]
  112. Meneilly, G.S.; Ryan, E.A.; Radziuk, J.; Lau, D.C.; Yale, J.F.; Morais, J.; Chiasson, J.L.; Rabasa-Lhoret, R.; Maheux, P.; Tessier, D.; et al. Effect of acarbose on insulin sensitivity in elderly patients with diabetes. Diabetes Care 2000, 23, 1162–1167. [Google Scholar] [CrossRef] [Green Version]
  113. Scott, R.; Lintott, C.J.; Zimmet, P.; Campbell, L.; Bowen, K.; Welborn, T. Will acarbose improve the metabolic abnormalities of insulin-resistant type 2 diabetes mellitus? Diabetes Res. Clin. Pract. 1999, 43, 179–185. [Google Scholar] [CrossRef]
  114. Chan, J.C.; Chan, K.-W.A.; Ho, L.L.; MC Fuh, M.; Horn, L.C.; Sheaves, R.; Panelo, A.A.; Kim, D.-K.; Embong, M. For the Asian Acarbose Study Group An Asian Multicenter Clinical Trial to Assess the Efficacy and Tolerability of Acarbose Compared with Placebo in Type 2 Diabetic Patients Previously Treated with Diet. Diabetes Care 1998, 21, 1058–1061. [Google Scholar] [CrossRef]
  115. Fischer, S.; Hanefeld, M.; Spengler, M.; Boehme, K.; Temelkova-Kurktschiev, T. European study on dose-response relationship of acarbose as a first-line drug in non-insulin-dependent diabetes mellitus: Efficacy and safety of low and high doses. Geol. Rundsch. 1998, 35, 34–40. [Google Scholar] [CrossRef]
  116. Braun, D.; Schönherr, U.; Mitzkat, H.J. Efficacy of acarbose monotherapy in patients with type 2 diabetes: A double-blind study conducted in general practice. Endocrinol. Metab. Suppl. 1996, 3, 275–280. [Google Scholar]
  117. Hou, W.K.; Zhang, B.Z.; Xu, J.; Jiang, B.; Shi, Z.C. A double-blind controlled study of glucobay in the treatment of NIDDM. Chin. J. Diabetes 1996, 4, 56–59. [Google Scholar]
  118. Zheng, G.F.; Wang, J.P.; Zhang, H.; Hu, Z.X.; Liu, J.; Xiao, J.Z.; Chen, S.M.; Cao, H.B.; Li, G.W.; Hu, Y.H.; et al. The clinical investigation of the effect of acarbose on non-insulin-dependent diabetics. Chin. J. Endocrinol. Metab. 1995, 11, 163–164, 192. [Google Scholar]
  119. Hotta, N.; Kakuta, H.; Sano, T.; Matsumae, H.; Yamada, H.; Kitazawa, S.; Sakamoto, N. Long-term Effect of Acarbose on Glycaemic Control in Non-insulin-dependent Diabetes Mellitus: A Placebo-controlled Double-blind Study. Diabet. Med. 1993, 10, 134–138. [Google Scholar] [CrossRef]
  120. Santeusanio, F.; Ventura, M.M.; Contadini, S.; Compagnucci, P.; Moriconi, V.; Zaccarini, P.; Marra, G.; Amigoni, S.; Bianchi, W.; Brunetti, P. Efficacy and safety of two different dosages of acarbose in non-insulin dependent diabetic patients treated by diet alone. Diabetes Nutr. Metab.-Clin. Exp. 1993, 6, 147–154. [Google Scholar]
  121. Hanefeld, M.; Fischer, S.; Schulze, J.; Spengler, M.; Wargenau, M.; Schollberg, K.; Fücker, K. Therapeutic Potentials of Acarbose as First-Line Drug in NIDDM Insufficiently Treated with Diet Alone. Diabetes Care 1991, 14, 732–737. [Google Scholar] [CrossRef]
  122. Chen, D.L.; Han, C.K.; Zhang, H.J. Evaluation of clinical effect of acarbose in the treatment of type 2 diabetes complicated with nonalcoholic fatty liver disease. Chin. Foreign Med. Res. 2017, 15, 123–124. [Google Scholar]
  123. Yang, L.H. Clinical efficacy of acarbose in the treatment of patients with type 2 diabetes combined with nonalcoholic fatty liver disease. Guide China Med. 2015, 13, 17–18. [Google Scholar]
  124. Wu, H.X.; Li, J.X. Efficacy of acarbose on type 2 diabetes complicated with nonalcoholic fatty liver disease. J. Pract. Diabetol. 2014, 10, 32–33. [Google Scholar]
  125. Yang, Y.; Xing, L.F.; Wang, A.M. Clinical study for the influence of acarbose on TNF-α and IL-6 on type 2 diabetes mellitus. Chin. J. Pract. Med. 2014, 41, 8–10. [Google Scholar]
  126. Hao, H.R.; Hu, W.; Yu, W.N. Platelet CD62P expression in early type 2 diabetes and the effect of acarbose. Shandong Med. J. 2012, 52, 50–52. [Google Scholar]
  127. Ye, X.H.; Yan, X.D.; Liang, S.; Lao, D.H. Efficacy of domestic acarbose on new-onset type 2 diabetes. Guangxi Med. J. 2009, 31, 963–964. [Google Scholar]
  128. Seino, Y.; Fujita, T.; Hiroi, S.; Hirayama, M.; Kaku, K. Efficacy and safety of alogliptin in Japanese patients with type 2 diabetes mellitus: A randomized, double-blind, dose-ranging comparison with placebo, followed by a long-term extension study. Curr. Med. Res. Opin. 2011, 27, 1781–1792. [Google Scholar] [CrossRef] [PubMed]
  129. Guo, H.G.; Ling, H.W. Clinical effect of voglibose in the treatment of type 2 diabetes mellitus. Acta Acad. Med. Xuzhou 2001, 21, 311–313. [Google Scholar]
  130. Drent, M.L.; Tollefsen, A.T.M.; Van Heusden, F.H.J.A.; Hoenderdos, E.B.M.; Jonker, J.J.C.; Van Der Veen, E.A. Dose-dependent efficacy of miglitol, an alpha-glucosidase inhibitor, in type 2 diabetic patients on diet alone: Results of a 24-week double-blind placebo-controlled study. Diabetes Nutr. Metab.-Clin. Exp. 2002, 15, 152–159. [Google Scholar]
  131. Gantz, I.; Okamoto, T.; Ito, Y.; Okuyama, K.; O’Neill, E.A.; Kaufman, K.D.; Engel, S.S.; Lai, E.; and the Omarigliptin Study 020 Group. A randomized, placebo- and sitagliptin-controlled trial of the safety and efficacy of omarigliptin, a once-weekly dipeptidyl peptidase-4 inhibitor, in Japanese patients with type 2 diabetes. Diabetes Obes. Metab. 2017, 19, 1602–1609. [Google Scholar] [CrossRef] [Green Version]
  132. Tian, M.; Liang, Z.; Liu, R.; Li, K.; Tan, X.; Luo, Y.; Yang, M.; Gu, H.F.; Liu, H.; Li, L.; et al. Effects of sitagliptin on circulating zinc-α2-glycoprotein levels in newly diagnosed type 2 diabetes patients: A randomized trial. Eur. J. Endocrinol. 2016, 174, 147–155. [Google Scholar] [CrossRef]
  133. Roden, M.; on behalf of the EMPA-REG EXTEND™ MONO investigators; Merker, L.; Christiansen, A.V.; Roux, F.; Salsali, A.; Kim, G.; Stella, P.; Woerle, H.J.; Broedl, U.C. Safety, tolerability and effects on cardiometabolic risk factors of empagliflozin monotherapy in drug-naïve patients with type 2 diabetes: A double-blind extension of a Phase III randomized controlled trial. Cardiovasc. Diabetol. 2015, 14, 1–11. [Google Scholar] [CrossRef] [Green Version]
  134. Roden, M.; Weng, J.; Eilbracht, J.; Delafont, B.; Kim, G.; Woerle, H.J.; Broedl, U.C. Empagliflozin monotherapy with sitagliptin as an active comparator in patients with type 2 diabetes: A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Diabetes Endocrinol. 2013, 1, 208–219. [Google Scholar] [CrossRef]
  135. Sun, J.; Li, W. Effect of sitagliptin on insulin resistance in patients with type 2 diabetes. Chin. J. Gerontol. 2013, 33, 1886–1887. [Google Scholar]
  136. Barzilai, N.; Guo, H.; Mahoney, E.M.; Caporossi, S.; Golm, G.T.; Langdon, R.B.; Williams-Herman, D.; Kaufman, K.D.; Amatruda, J.M.; Goldstein, B.J.; et al. Efficacy and tolerability of sitagliptin monotherapy in elderly patients with type 2 diabetes: A randomized, double-blind, placebo-controlled trial. Curr. Med. Res. Opin. 2011, 27, 1049–1058. [Google Scholar] [CrossRef] [PubMed]
  137. Iwamoto, Y.; Taniguchi, T.; Nonaka, K.; Okamoto, T.; Okuyama, K.; Arjona, F.J.C.; Amatruda, J. Dose-ranging efficacy of sitagliptin, a dipeptidyl peptidase-4 inhibitor, in Japanese patients with type 2 diabetes mellitus. Endocr. J. 2010, 57, 383–394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Mohan, V.; Yang, W.; Son, H.-Y.; Xu, L.; Noble, L.; Langdon, R.B.; Amatruda, J.M.; Stein, P.P.; Kaufman, K.D. Efficacy and safety of sitagliptin in the treatment of patients with type 2 diabetes in China, India, and Korea. Diabetes Res. Clin. Pract. 2009, 83, 106–116. [Google Scholar] [CrossRef]
  139. Nonaka, K.; Kakikawa, T.; Sato, A.; Okuyama, K.; Fujimoto, G.; Kato, N.; Suzuki, H.; Hirayama, Y.; Ahmed, T.; Davies, M.J.; et al. Efficacy and safety of sitagliptin monotherapy in Japanese patients with type 2 diabetes. Diabetes Res. Clin. Pract. 2008, 79, 291–298. [Google Scholar] [CrossRef] [PubMed]
  140. Hanefeld, M.; Herman, G.A.; Wu, M.; Mickel, C.; Sanchez, M.; Stein, P.P. Once-daily sitagliptin, a dipeptidyl peptidase-4 inhibitor, for the treatment of patients with type 2 diabetes. Curr. Med. Res. Opin. 2007, 23, 1329–1339. [Google Scholar] [CrossRef] [PubMed]
  141. Aschner, P.; Kipnes, M.S.; Lunceford, J.K.; Sanchez, M.; Mickel, C.; Williams-Herman, D.E. Effect of the Dipeptidyl Peptidase-4 Inhibitor Sitagliptin as Monotherapy on Glycemic Control in Patients with Type 2 Diabetes. Diabetes Care 2006, 29, 2632–2637. [Google Scholar] [CrossRef] [Green Version]
  142. Raz, I.; Hanefeld, M.; Xu, L.; Caria, C.; Williams-Herman, D.; Khatami, H.; Sitagliptin Study 023 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy in patients with type 2 diabetes mellitus. Diabetologia 2006, 49, 2564–2571. [Google Scholar] [CrossRef] [Green Version]
  143. Deng, X.; Ma, R.; Zhu, H.; Zhu, J. A randomized-controlled study of sitagliptin for treating diabetes mellitus complicated by nonalcoholic fatty liver disease. Eur. J. Gastroenterol. Hepatol. 2017, 29, 297–301. [Google Scholar] [CrossRef]
  144. Zhang, J.B.; Lu, Y.Z.; Fan, M.S. Efficacy analysis of saxagliptin treatment of new-onset type 2 diabetes. Guide China Med. 2016, 14, 9+12. [Google Scholar]
  145. Xiu, S.L.; Wang, L. Efficacy and safety of saxagliptin in newly diagnosed type 2 diabetes mellitus patients. China Med. 2015, 10, 352–354. [Google Scholar]
  146. Han, B.Y.; Lu, L.M.; Zhang, L.P.; Fan, Z.Y. Effect of DPP-4 inhibitor saxagliptin on endothelium-dependent vasodilation in patients with type 2 diabetes. J. Shanxi Med. Univ. 2014, 45, 291–294. [Google Scholar]
  147. Prasanna Kumar, K.M.; Jain, S.M.; Tou, C.; Schutzer, K.-M. Saxagliptin as initial therapy in treatment-naive Indian adults with type 2 diabetes mellitus inadequately controlled with diet and exercise alone: A randomized, double-blind, placebo-controlled, phase IIIb clinical study. Int. J. Diabetes Dev. Ctries. 2014, 34, 201–209. [Google Scholar] [CrossRef]
  148. Wu, Y.F. Evaluation of the Efficacy of Saxagliptin Monotherapy in Incipient Type 2 Diabetes; Zhejiang University: Hangzhou, China, 2013. [Google Scholar]
  149. Frederich, R.; McNeill, R.; Berglind, N.; Fleming, D.; Chen, R. The efficacy and safety of the dipeptidyl peptidase-4 inhibitor saxagliptin in treatment-naïve patients with type 2 diabetes mellitus: A randomized controlled trial. Diabetol. Metab. Syndr. 2012, 4, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Pan, C.Y.; Tou, C.; Gause-Nilsson, I.; Yang, W.; Zhao, J. Efficacy and safety of saxagliptin in drug-naïve Asian patients with type 2 diabetes mellitus: A randomized controlled trial. Diabetes/Metab. Res. Rev. 2012, 28, 268–275. [Google Scholar] [CrossRef]
  151. Rosenstock, J.; Aguilar-Salinas, C.; Klein, E.; Nepal, S.; List, J.; Chen, R.; for the CV181-011 Study Investigators. Effect of saxagliptin monotherapy in treatment-naïve patients with type 2 diabetes. Curr. Med. Res. Opin. 2009, 25, 2401–2411. [Google Scholar] [CrossRef]
  152. Rosenstock, J.; Sankoh, S.; List, J.F. Glucose-lowering activity of the dipeptidyl peptidase-4 inhibitor saxagliptin in drug-naive patients with type 2 diabetes. Diabetes Obes. Metab. 2008, 10, 376–386. [Google Scholar] [CrossRef]
  153. Mari, A.; Scherbaum, W.A.; Nilsson, P.M.; Lalanne, G.; Schweizer, A.; Dunning, B.E.; Jauffret, S.; Foley, J.E. Characterization of the Influence of Vildagliptin on Model-Assessed β-Cell Function in Patients with Type 2 Diabetes and Mild Hyperglycemia. J. Clin. Endocrinol. Metab. 2008, 93, 103–109. [Google Scholar] [CrossRef] [Green Version]
  154. Scherbaum, W.A.; Schweizer, A.; Mari, A.; Nilsson, P.M.; Lalanne, G.; Jauffret, S.; Foley, J.E. Efficacy and tolerability of vildagliptin in drug-naïve patients with type 2 diabetes and mild hyperglycaemia. Diabetes Obes. Metab. 2008, 10, 675–682. [Google Scholar] [CrossRef]
  155. Scherbaum, W.A.; Schweizer, A.; Mari, A.; Nilsson, P.M.; Lalanne, G.; Wang, Y.; Dunning, B.E.; Foley, J.E. Evidence that vildagliptin attenuates deterioration of glycaemic control during 2-year treatment of patients with type 2 diabetes and mild hyperglycaemia. Diabetes Obes. Metab. 2008, 10, 1114–1124. [Google Scholar] [CrossRef]
  156. Dejager, S.; Razac, S.; Foley, J.E.; Schweizer, A. Vildagliptin in drug-naïve patients with type 2 diabetes: A 24-week, double-blind, randomized, placebo-controlled, multiple-dose study. Horm. Metab. Res. 2007, 39, 218–223. [Google Scholar] [CrossRef]
  157. Pi-Sunyer, F.X.; Schweizer, A.; Mills, D.; Dejager, S. Efficacy and tolerability of vildagliptin monotherapy in drug-naïve patients with type 2 diabetes. Diabetes Res. Clin. Pract. 2007, 76, 132–138. [Google Scholar] [CrossRef] [PubMed]
  158. Pratley, R.E.; Jauffret-Kamel, S.; Galbreath, E.; Holmes, D. Twelve-week Monotherapy with the DPP-4 Inhibitor Vildagliptin Improves Glycemic Control in Subjects with Type 2 Diabetes. Horm. Metab. Res. 2006, 38, 423–428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Ristić, S.; Byiers, S.; Foley, J.; Holmes, D. Improved glycaemic control with dipeptidyl peptidase-4 inhibition in patients with type 2 diabetes: Vildagliptin (LAF237) dose response. Diabetes Obes. Metab. 2005, 7, 692–698. [Google Scholar] [CrossRef] [PubMed]
  160. Chen, J.; Li, Y.C. Efficacy of vildagliptin on newly diagnosed type 2 diabetes complicated with nonalcoholic fatty liver disease. J. Clin. Intern. Med. 2016, 33, 709–710. [Google Scholar]
  161. Lin, N.; Liu, H.B. Effect of linagliptin on lipid metabolism and islet resistance in patients with type 2 diabetes. Clin. Res. 2017, 25, 77–78, 80. [Google Scholar]
  162. Chen, Y.; Ning, G.; Wang, C.; Gong, Y.; Patel, S.; Zhang, C.; Izumoto, T.; Woerle, H.; Wang, W. Efficacy and safety of linagliptin monotherapy in Asian patients with inadequately controlled type 2 diabetes mellitus: A multinational, 24-week, randomized, clinical trial. J. Diabetes Investig. 2015, 6, 692–698. [Google Scholar] [CrossRef]
  163. Wu, W.; Li, Y.; Chen, X.; Lin, D.; Xiang, S.; Shen, F.; Gu, X. Effect of Linagliptin on Glycemic Control in Chinese Patients with Newly-Diagnosed, Drug-Naïve Type 2 Diabetes Mellitus: A Randomized Controlled Trial. J. Pharmacol. Exp. Ther. 2015, 21, 2678–2684. [Google Scholar] [CrossRef] [Green Version]
  164. Wu, W.J.; Chen, X.; Shen, F.X.; Xiang, S.Y.; Lin, D.N.; Gu, X.M. Effects of linagliptin treatment on hyperglycemia and islet function in newly-diagnosed type 2 diabetic patients. Chin. J. New Drugs Clin. Remedies 2014, 33, 263–266. [Google Scholar]
  165. Barnett, A.H.; Patel, S.; Harper, R.; Toorawa, R.; Thiemann, S.; von Eynatten, M.; Woerle, H.-J. Linagliptin monotherapy in type 2 diabetes patients for whom metformin is inappropriate: An 18-week randomized, double-blind, placebo-controlled phase III trial with a 34-week active-controlled extension. Diabetes Obes. Metab. 2012, 14, 1145–1154. [Google Scholar] [CrossRef]
  166. Kawamori, R.; Inagaki, N.; Araki, E.; Watada, H.; Hayashi, N.; Horie, Y.; Sarashina, A.; Gong, Y.; Von Eynatten, M.; Woerle, H.J.; et al. Linagliptin monotherapy provides superior glycaemic control versus placebo or voglibose with comparable safety in Japanese patients with type 2 diabetes: A randomized, placebo and active comparator-controlled, double-blind study. Diabetes Obes. Metab. 2012, 14, 348–357. [Google Scholar] [CrossRef]
  167. Del Prato, S.; Barnett, A.H.; Huisman, H.; Neubacher, D.; Woerle, H.J.; Dugi, K.A. Effect of linagliptin monotherapy on glycaemic control and markers of β-cell function in patients with inadequately controlled type 2 diabetes: A randomized controlled trial. Diabetes Obes. Metab. 2011, 13, 258–267. [Google Scholar] [CrossRef] [PubMed]
  168. Pan, C.; Han, P.; Ji, Q.; Li, C.; Lu, J.; Yang, J.; Li, W.; Zeng, J.; Hsieh, A.-T.; Chan, J. Efficacy and safety of alogliptin in patients with type 2 diabetes mellitus: A multicentre randomized double-blind placebo-controlled Phase 3 study in mainland China, Taiwan, and Hong Kong. J. Diabetes 2017, 9, 386–395. [Google Scholar] [CrossRef] [PubMed]
  169. Inagaki, N.; Onouchi, H.; Maezawa, H.; Kuroda, S.; Kaku, K. Once-weekly trelagliptin versus daily alogliptin in Japanese patients with type 2 diabetes: A randomised, double-blind, phase 3, non-inferiority study. Lancet Diabetes Endocrinol. 2015, 3, 191–197. [Google Scholar] [CrossRef] [PubMed]
  170. DeFronzo, R.A.; Fleck, P.R.; Wilson, C.A.; Mekki, Q.; Group, A.S. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor alogliptin in patients with type 2 diabetes and inadequate glycemic control: A randomized, double-blind, placebo-controlled study. Diabetes Care 2008, 31, 2315–2317. [Google Scholar] [CrossRef] [PubMed]
  171. Kutoh, E.; Ukai, Y. Alogliptin as an initial therapy in patients with newly diagnosed, drug naïve type 2 diabetes: A randomized, control trial. Endocrine 2012, 41, 435–441. [Google Scholar] [CrossRef]
  172. Wang, W.Y. The clinical effect of dapagliflozin in the treatment of newly diagnosed type 2 diabetes. Electron. J. Clin. Med. Lit. 2020, 7, 159–160. [Google Scholar]
  173. Liao, X.; Wang, X.; Li, H.; Li, L.; Zhang, G.; Yang, M.; Yuan, L.; Liu, H.; Yang, G.; Gao, L. Sodium-Glucose Cotransporter 2 (SGLT2) Inhibitor Increases Circulating Zinc-A2-Glycoprotein Levels in Patients with Type 2 Diabetes. Sci. Rep. 2016, 6, 32887. [Google Scholar] [CrossRef] [Green Version]
  174. Ji, L.; Ma, J.; Li, H.; Mansfield, T.A.; T’Joen, C.L.; Iqbal, N.; Ptaszynska, A.; List, J.F. Dapagliflozin as Monotherapy in Drug-Naive Asian Patients with Type 2 Diabetes Mellitus: A Randomized, Blinded, Prospective Phase III Study. Clin. Ther. 2014, 36, 84–100.e9. [Google Scholar] [CrossRef] [Green Version]
  175. Kaku, K.; Kiyosue, A.; Inoue, S.; Ueda, N.; Tokudome, T.; Yang, J.; Langkilde, A.M. Efficacy and safety of dapagliflozin monotherapy in Japanese patients with type 2 diabetes inadequately controlled by diet and exercise. Diabetes Obes. Metab. 2014, 16, 1102–1110. [Google Scholar] [CrossRef]
  176. Liu, L.M.; Yang, Y.; Bao, M.J.; Li, P.Q.; Zhang, X.J.; Xian, Y.; Wu, J.C. Therapeutic effect of dapagliflozin on type 2 diabetes mellitus. J. Chin. Pract. Diagn. Ther. 2014, 28, 926–927, 929. [Google Scholar]
  177. Kaku, K.; Inoue, S.; Matsuoka, O.; Kiyosue, A.; Azuma, H.; Hayashi, N.; Tokudomé, T.; Langkilde, A.M.; Parikh, S. Efficacy and safety of dapagliflozin as a monotherapy for type 2 diabetes mellitus in Japanese patients with inadequate glycaemic control: A phase II multicentre, randomized, double-blind, placebo-controlled trial. Diabetes Obes. Metab. 2013, 15, 432–440. [Google Scholar] [CrossRef] [PubMed]
  178. Bailey, C.J.; Iqbal, N.; T’Joen, C.; List, J.F. Dapagliflozin monotherapy in drug-naïve patients with diabetes: A randomized-controlled trial of low-dose range. Diabetes Obes. Metab. 2012, 14, 951–959. [Google Scholar] [CrossRef] [PubMed]
  179. Ferrannini, E.; Ramos, S.J.; Salsali, A.; Tang, W.; List, J.F. Dapagliflozin monotherapy in type 2 diabetic patients with inadequate glycemic control by diet and exercise: A randomized, double-blind, placebo-controlled, phase 3 trial. Diabetes Care 2010, 33, 2217–2224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Kadowaki, T.; Haneda, M.; Inagaki, N.; Terauchi, Y.; Taniguchi, A.; Koiwai, K.; Rattunde, H.; Woerle, H.J.; Broedl, U.C. Empagliflozin Monotherapy in Japanese Patients with Type 2 Diabetes Mellitus: A Randomized, 12-Week, Double-Blind, Placebo-Controlled, Phase II Trial. Adv. Ther. 2014, 31, 621–638. [Google Scholar] [CrossRef] [PubMed]
  181. Inagaki, N.; Kondo, K.; Yoshinari, T.; Takahashi, N.; Susuta, Y.; Kuki, H. Efficacy and safety of canagliflozin monotherapy in Japanese patients with type 2 diabetes inadequately controlled with diet and exercise: A 24-week, randomized, double-blind, placebo-controlled, Phase III study. Expert Opin. Pharmacother. 2014, 15, 1501–1515. [Google Scholar] [CrossRef]
  182. Inagaki, N.; Kondo, K.; Yoshinari, T.; Maruyama, N.; Susuta, Y.; Kuki, H. Efficacy and safety of canagliflozin in Japanese patients with type 2 diabetes: A randomized, double-blind, placebo-controlled, 12-week study. Diabetes Obes. Metab. 2013, 15, 1136–1145. [Google Scholar] [CrossRef]
  183. Stenlöf, K.; Cefalu, W.T.; Kim, K.; Alba, M.; Usiskin, K.; Tong, C.; Canovatchel, W.; Meininger, G. Efficacy and safety of canagliflozin monotherapy in subjects with type 2 diabetes mellitus inadequately controlled with diet and exercise. Diabetes Obes. Metab. 2013, 15, 372–382. [Google Scholar] [CrossRef] [Green Version]
  184. Hong, L.H. Efficacy and Safety of Exenatide Treatment in Type 2 Diabetes Mellitus; Zhejiang University: Hangzhou, China, 2016. [Google Scholar]
  185. Gastaldelli, A.; Brodows, R.G.; D’Alessio, D. The effect of chronic twice daily exenatide treatment on β-cell function in new onset type 2 diabetes. Clin. Endocrinol. 2014, 80, 545–553. [Google Scholar] [CrossRef]
  186. Moretto, T.J.; Milton, D.R.; Ridge, T.D.; MacConell, L.A.; Okerson, T.; Wolka, A.M.; Brodows, R.G. Efficacy and tolerability of exenatide monotherapy over 24 weeks in antidiabetic drug-naive patients with type 2 diabetes: A randomized, double-blind, placebo-controlled, parallel-group study. Clin. Ther. 2008, 30, 1448–1460. [Google Scholar] [CrossRef]
  187. Zhu, H.Q. Effect of Exenatide on Vascular Endothelial Function and Cardiovascular Disease Risk Factors in Patients with Type 2 Diabetes; Guangxi Medical University: Nanning, China, 2017. [Google Scholar]
  188. Yamada, Y.; Katagiri, H.; Hamamoto, Y.; Deenadayalan, S.; Navarria, A.; Nishijima, K.; Seino, Y.; Fukushima, Y.; Hisatomi, A.; Ide, Y.; et al. Dose-response, efficacy, and safety of oral semaglutide monotherapy in Japanese patients with type 2 diabetes (PIONEER 9): A 52-week, phase 2/3a, randomised, controlled trial. Lancet Diabetes Endocrinol. 2020, 8, 377–391. [Google Scholar] [CrossRef]
  189. Nino, A.; Okuda, I.; Wilson, T.H.; Yue, L.; Nakajima, H.; Tsuboi, M.; Carr, M.C.; Seino, Y. Weekly glucagon-like peptide-1 receptor agonist albiglutide as monotherapy improves glycemic parameters in Japanese patients with type 2 diabetes mellitus: A randomized, double-blind, placebo-controlled study. J. Diabetes Investig. 2018, 9, 558–566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Tran, S.; Kramer, C.K.; Zinman, B.; Choi, H.; Retnakaran, R. Effect of chronic liraglutide therapy and its withdrawal on time to postchallenge peak glucose in type 2 diabetes. Am. J. Physiol. Metab. 2018, 314, E287–E295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Miyagawa, J.; Odawara, M.; Takamura, T.; Iwamoto, N.; Takita, Y.; Imaoka, T. Once-weekly glucagon-like peptide-1 receptor agonist dulaglutide is non-inferior to once-daily liraglutide and superior to placebo in Japanese patients with type 2 diabetes: A 26-week randomized phase III study. Diabetes Obes Metab. 2015, 17, 974–983. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Tan, X.R.; Liang, Z.R.; Li, S.B.; Tang, S.G.; Fang, C.; Tang, L.H.; Pi, Y.; Qin, F.; Wang, Y. Effects of GLP-1 receptor agonist on Zinc-α2-glycoprotein and its relationship with insulin resistance in patients with newly diagnosed type 2 diabetes. J. Chongqing Med. Univ. 2015, 40, 1395–1400. [Google Scholar]
  193. Retnakaran, R.; Kramer, C.K.; Choi, H.; Swaminathan, B.; Zinman, B. Liraglutide and the Preservation of Pancreatic Beta-Cell Function in Early Type 2 Diabetes: The LIBRA Trial. Can. J. Diabetes 2014, 38, S4–S5. [Google Scholar] [CrossRef]
  194. Seino, Y.; Rasmussen, M.; Zdravkovic, M.; Kaku, K. Dose-dependent improvement in glycemia with once-daily liraglutide without hypoglycemia or weight gain: A double-blind, randomized, controlled trial in Japanese patients with type 2 diabetes. Diabetes Res. Clin. Pract. 2008, 81, 161–168. [Google Scholar] [CrossRef]
  195. Vilsbøll, T.; Brock, B.; Perrild, H.; Levin, K.; Lervang, H.-H.; Kølendorf, K.; Krarup, T.; Schmitz, O.; Zdravkovic, M.; Le-Thi, T.; et al. Liraglutide, a once-daily human GLP-1 analogue, improves pancreatic B-cell function and arginine-stimulated insulin secretion during hyperglycaemia in patients with Type 2 diabetes mellitus. Diabet. Med. 2008, 25, 152–156. [Google Scholar] [CrossRef]
  196. Fan, P. Efficacy of long-acting GLP-1 analogue combined with low-carbohydrate diet in the treatment of obese type 2 diabetes patients. J. Pract. Diabetol. 2020, 16, 43–44. [Google Scholar]
  197. Li, Q.Y.; Liu, X.; Li, H.X.; Yang, C.W.; Zhang, W.H.; Wang, T.T.; Guo, Z.H.; Zhou, Y.R. Clinical effect of liraglutide on type 2 diabetes mellitus complicated with polycystic ovary syndrome. China Med. 2018, 13, 549–552. [Google Scholar]
  198. Liu, M.R. The Clinical Efficacy and Safety Observation on a Low Carbohydrate Diet Combined with Liraglutide in Obese Patients with Diabetes; University of South China: Hengyang, China, 2017. [Google Scholar]
  199. Fonseca, V.A.; Alvarado-Ruiz, R.; Raccah, D.; Boka, G.; Miossec, P.; Gerich, J.E. Efficacy and safety of the once-daily GLP-1 receptor agonist lixisenatide in monotherapy: A randomized, double-blind, placebo-controlled trial in patients with type 2 diabetes (GetGoal-Mono). Diabetes Care 2012, 35, 1225–1231. [Google Scholar] [CrossRef] [Green Version]
  200. Holman, R.R.; Paul, S.K.; Bethel, M.A.; Matthews, D.R.; Neil, H.A. 10-year follow-up of intensive glucose control in type 2 diabetes. N. Engl. J. Med. 2008, 359, 1577–1589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Sherifali, D.; Nerenberg, K.; Pullenayegum, E.; Cheng, J.E.; Gerstein, H.C. The effect of oral antidiabetic agents on A1C levels: A systematic review and meta-analysis. Diabetes Care 2010, 33, 1859–1864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Centers for Disease Control and Prevention (CDC). Prevalence of overweight and obesity among adults with diagnosed diabetes—United States, 1988–1994 and 1999–2002. MMWR Morb Mortal Wkly Rep. 2004, 53, 1066–1068. [Google Scholar]
  203. Nichols, G.A.; Bell, K.; Kimes, T.M.; O’Keeffe-Rosetti, M. Medical Care Costs Associated with Long-term Weight Maintenance Versus Weight Gain among Patients with Type 2 Diabetes. Diabetes Care 2016, 39, 1981–1986. [Google Scholar] [CrossRef] [Green Version]
  204. Wing, R.R.; Lang, W.; Wadden, T.A.; Safford, M.; Knowler, W.C.; Bertoni, A.G.; Hill, J.O.; Brancati, F.L.; Peters, A.; Wagenknecht, L.; et al. Benefits of Modest Weight Loss in Improving Cardiovascular Risk Factors in Overweight and Obese Individuals with Type 2 Diabetes. Diabetes Care 2011, 34, 1481–1486. [Google Scholar] [CrossRef] [Green Version]
  205. Williamson, D.F.; Thompson, T.J.; Thun, M.; Flanders, D.; Pamuk, E.; Byers, T. Intentional weight loss and mortality among overweight individuals with diabetes. Diabetes Care 2000, 23, 1499–1504. [Google Scholar] [CrossRef] [Green Version]
  206. Redmon, J.B.; Bertoni, A.G.; Connelly, S.; Feeney, P.A.; Glasser, S.P.; Glick, H.; Greenway, F.; Hesson, L.A.; Lawlor, M.S.; Montez, M.; et al. Effect of the Look AHEAD Study Intervention on Medication Use and Related Cost to Treat Cardiovascular Disease Risk Factors in Individuals with Type 2 Diabetes. Diabetes Care 2010, 33, 1153–1158. [Google Scholar] [CrossRef] [Green Version]
  207. Maruthur, N.M.; Tseng, E.; Hutfless, S.; Wilson, L.M.; Suarez-Cuervo, C.; Berger, Z.; Chu, Y.; Iyoha, E.; Segal, J.B.; Bolen, S. Diabetes Medications as Monotherapy or Metformin-Based Combination Therapy for Type 2 Diabetes: A Systematic Review and Meta-analysis. Ann. Intern. Med. 2016, 164, 740–751. [Google Scholar] [CrossRef]
  208. Bolen, S.; Feldman, L.; Vassy, J.; Wilson, L.; Yeh, H.-C.; Marinopoulos, S.; Wiley, C.; Selvin, E.; Wilson, R.; Bass, E.; et al. Systematic Review: Comparative Effectiveness and Safety of Oral Medications for Type 2 Diabetes Mellitus. Ann. Intern. Med. 2007, 147, 386–399. [Google Scholar] [CrossRef]
  209. Emdin, C.A.; Rahimi, K.; Neal, B.; Callender, T.; Perkovic, V.; Patel, A. Blood pressure lowering in type 2 diabetes: A systematic review and meta-analysis. JAMA 2015, 313, 603–615. [Google Scholar] [CrossRef]
  210. Heart Outcomes Prevention Evaluation Study Investigators. Effects of ramipril on cardiovascular and microvascular outcomes in people with diabetes mellitus: Results of the HOPE study and MICRO-HOPE substudy. Lancet 2000, 355, 253–259. [Google Scholar] [CrossRef]
  211. Tsapas, A.; Karagiannis, T.; Kakotrichi, P.; Avgerinos, I.; Mantsiou, C.; Tousinas, G.; Manolopoulos, A.; Liakos, A.; Malandris, K.; Matthews, D.R.; et al. Comparative efficacy of glucose-lowering medications on body weight and blood pressure in patients with type 2 diabetes: A systematic review and network meta-analysis. Diabetes Obes. Metab. 2021, 23, 2116–2124. [Google Scholar] [CrossRef] [PubMed]
  212. Frier, B.M. Hypoglycaemia in diabetes mellitus: Epidemiology and clinical implications. Nat. Rev. Endocrinol. 2014, 10, 711–722. [Google Scholar] [CrossRef] [PubMed]
  213. Shi, L.; Fonseca, V.; Childs, B. Economic burden of diabetes-related hypoglycemia on patients, payors, and employers. J. Diabetes Its Complicat. 2021, 35, 107916. [Google Scholar] [CrossRef] [PubMed]
Figure 1. PRISMA flow diagram of study search and selection.
Figure 1. PRISMA flow diagram of study search and selection.
Jcm 11 07094 g001
Table 1. Treatment effects of glucose-lowering drugs after removing the effect of placebo/lifestyle intervention.
Table 1. Treatment effects of glucose-lowering drugs after removing the effect of placebo/lifestyle intervention.
Glucose-Lowering DrugHbA1c, %FPG, mmol/LBMI, kg/m2TC, mmol/LHDL-C, mmol/LSBP, mmHgHypoglycemia
Metformin−0.96 (−1.16, −0.76) *−1.65 (−2.02, −1.27) *−1.28 (−2.26, −0.31) *−0.30 (−0.47, −0.14) *0.05 (0.02, 0.09) *−1.50 (−3.78, 0.79)1.53 (0.98, 2.40)
SUs−1.39 (−1.63, −1.16) *−2.70 (−3.18, −2.23) *1.22 (0.13, 2.31) *−0.40 (−1.20, 0.40)−0.00 (−0.18, 0.18)1.84 (−4.57, 8.25)5.44 (2.11, 14.02) *
Glyburide−1.50 (−2.69, −0.30) *−2.35 (−3.59, −1.12) *0.27 (−1.48, 2.03)−0.80 (−1.94, 0.34)−0.20 (−0.63, 0.23)
Glimepiride−1.36 (−1.57, −1.16) *−2.41 (−3.09, −1.73) *1.79 (0.46, 3.12) *−0.22 (−1.60, 1.17)0.12 (−0.04, 0.29)1.84 (−4.57, 8.25)2.88 (0.45, 18.58)
Gliclazide−1.40 (−2.70, −0.10) *−2.22 (−3.47, −0.97) *−0.75 (−1.84, 0.34)0.07 (−0.08, 0.22)5.00 (0.25, 99.95)
Glipizide−1.47 (−1.87, −1.06) *−3.02 (−3.85, −2.20) *0.20 (−3.19, 3.59)0.41 (−1.64, 2.47)7.11 (2.18, 23.24) *
TZDs−0.89 (−1.04, −0.73) *−1.91 (−2.23, −1.60) *0.63 (0.26, 0.99) *0.01 (−0.19, 0.22)0.12 (0.07, 0.17) *0.78 (−2.37, 3.93)0.49 (0.23, 1.03)
Rosiglitazone−0.68 (−0.98, −0.38) *−1.73 (−2.32, −1.14) *0.91 (0.48, 1.35) *0.25 (0.03, 0.46) *0.06 (0.03, 0.08) *2.43 (−1.55, 6.42)0.43 (0.15, 1.26)
Pioglitazone−1.00 (−1.17, −0.82) *−2.01 (−2.34, −1.67) *0.38 (−0.07, 0.82)−0.09 (−0.32, 0.14)0.18 (0.09, 0.26) *−1.79 (−6.33, 2.74)0.55 (0.20, 1.53)
NIDEs−0.44 (−0.69, −0.20) *−0.75 (−1.04, −0.45) *0.08 (−1.29, 1.44)0.21 (−0.39, 0.81)0.08 (−0.21, 0.37)−5.98 (−12.33, 0.37)1.37 (0.34, 5.59)
Repaglinide−0.45 (−0.81, −0.09) *−0.64 (−1.27, −0.01) *0.97 (0.14, 6.77)
Nateglinide−0.45 (−0.79, −0.10) *−0.70 (−1.04, −0.36) *0.08 (−1.29, 1.44)0.21 (−0.39, 0.81)0.08 (−0.21, 0.37)−5.98 (−12.33, 0.37)2.00 (0.26, 15.33)
AGIs−0.62 (−0.79, −0.45) *−1.19 (−1.73, −0.64) *−0.49 (−1.26, 0.28)−0.29 (−0.58, −0.00) *0.03 (−0.12, 0.17)−1.40 (−4.71, 1.90)0.86 (0.51, 1.45)
Acarbose−0.74 (−0.96, −0.52) *−1.17 (−1.83, −0.50) *−0.60 (−1.66, 0.46)−0.34 (−0.65, −0.03) *0.07 (−0.11, 0.25)−1.40 (−4.71, 1.90)1.19 (0.34, 4.23)
Voglibose−0.20 (−0.33, −0.07) *−1.78 (−3.58, 0.02)0.10 (−0.13, 0.33)−0.15 (−0.92, 0.62)−0.17 (−0.48, 0.14)0.93 (0.10, 8.79)
Miglitol−0.53 (−0.85, −0.21) *−0.01 (−0.88, 0.86)0.79 (0.44, 1.44)
DPP−4is−0.63 (−0.68, −0.58) *−0.94 (−1.03, −0.85) *0.47 (−0.01, 0.95)−0.04 (−0.11, 0.02)0.03 (−0.01, 0.06)0.02 (−1.10, 1.14)0.89 (0.67, 1.18)
Sitagliptin−0.73 (−0.82, −0.65) *−1.07 (−1.20, −0.95) *0.10 (−1.24, 1.44)0.06 (0.01, 0.11) *0.06 (−0.02, 0.14)0.18 (−1.27, 1.64)0.79 (0.52, 1.20)
Saxagliptin−0.52 (−0.61, −0.44) *−0.83 (−1.00, −0.67) *−0.46 (−2.04, 1.12)1.21 (0.52, 2.81)
Vildagliptin−0.48 (−0.57, −0.38) *−0.56 (−0.82, −0.30) *−0.58 (−2.09, 0.93)−0.12 (−0.83, 0.59)1.07 (0.54, 2.13)
Linagliptin−0.68 (−0.79, −0.58) *−0.94 (−1.16, −0.73) *−0.06 (−0.21, 0.09)0.03 (−0.06, 0.12)−1.74 (−4.75, 1.26)0.52 (0.26, 1.01)
Alogliptin−0.68 (−0.76, −0.61) *−1.07 (−1.27, −0.86) *0.81 (0.27, 1.35) *−0.19 (−0.31, −0.07) *−0.00 (−0.04, 0.03)0.59 (−1.59, 2.76)2.97 (1.00, 8.77) *
SGLT2is−0.80 (−0.87, −0.72) *−1.58 (−1.81, −1.36) *−0.60 (−1.89, 0.69)0.22 (0.13, 0.31) *0.09 (0.07, 0.11) *−4.18 (−4.84, −3.53) *0.86 (0.55, 1.37)
Dapagliflozin−0.68 (−0.77, −0.59) *−1.27 (−1.49, −1.06) *−0.60 (−1.89, 0.69)−0.02 (−0.24, 0.20)0.03 (−0.09, 0.15)−3.89 (−5.02, −2.75) *1.29 (0.70, 2.36)
Empagliflozin−0.79 (−0.86, −0.72) *−1.84 (−1.97, −1.72) *0.20 (0.06, 0.34) *0.08 (0.07, 0.10) *−3.29 (−4.34, −2.25) *0.31 (0.14, 0.71) *
Canagliflozin−0.99 (−1.06, −0.92) *−2.09 (−2.31, −1.87) *0.30 (0.19, 0.40) *0.10 (0.07, 0.12) *−5.36 (−6.59, −4.14) *1.57 (0.79, 3.10)
GLP−1RAs−0.99 (−1.20, −0.78) *−1.64 (−2.00, −1.28) *−1.05 (−1.81, −0.29) *−0.42 (−0.61, −0.22) *0.03 (−0.01, 0.06)−2.98 (−4.67, −1.30) *1.57 (0.82, 3.02)
Exenatide twice-daily−0.64 (−0.82, −0.47) *−1.13 (−1.49, −0.77) *−1.65 (−2.26, −1.04) *−0.28 (−0.50, −0.06) *0.01 (−0.01, 0.03)−3.19 (−5.47, −0.90) *3.36 (0.84, 13.52)
Liraglutide−1.17 (−1.47, −0.87) *−1.97 (−2.47, −1.47) *−0.80 (−1.66, 0.07)−0.56 (−0.78, −0.34) *0.13 (0.05, 0.21) *−2.74 (−5.24, −0.25) *1.37 (0.58, 3.25)
Lixisenatide−0.60 (−0.83, −0.37) *−0.97 (−1.47, −0.48) *1.04 (0.25, 4.29)
Glucose-Lowering DrugDeathTotal
Vascular Events
Myocardial InfarctionHeart FailureStrokeDiabetic NephropathyAE-Induced Discontinuations
Metformin0.88 (0.46, 1.69)0.91 (0.22, 3.73)0.98 (0.10, 9.30)0.98 (0.10, 9.30)0.98 (0.10, 9.30)1.59 (0.20, 12.85)1.03 (0.74, 1.43)
SUs1.10 (0.27, 4.52)0.46 (0.03, 7.00)1.41 (0.06, 33.26)0.16 (0.01, 3.70)2.25 (0.74, 6.81)
 Glyburide1.09 (0.07, 16.30)2.24 (0.31, 16.50)
 Glimepiride1.16 (0.15, 9.23)0.46 (0.03, 7.00)1.41 (0.06, 33.26)0.16 (0.01, 3.70)0.99 (0.10, 9.40)
 Gliclazide1.00 (0.02, 48.82)1.00 (0.02, 48.82)
 Glipizide1.02 (0.02, 50.81)4.64 (0.74, 28.95)
TZDs0.95 (0.48, 1.90)1.44 (0.38, 5.39)0.82 (0.13, 5.06)0.97 (0.19, 4.92)0.96 (0.13, 7.21)0.97 (0.02, 48.35)1.25 (0.81, 1.95)
 Rosiglitazone0.94 (0.29, 3.00)1.00 (0.02, 46.40)1.00 (0.02, 46.40)1.00 (0.02, 46.40)0.97 (0.43, 2.23)
 Pioglitazone0.96 (0.41, 2.26)1.51 (0.37, 6.17)0.78 (0.10, 6.12)0.96 (0.16, 5.78)0.96 (0.13, 7.21)0.97 (0.02, 48.35)1.38 (0.82, 2.33)
NIDEs0.96 (0.20, 4.68)0.97 (0.24, 3.81)
 Repaglinide0.97 (0.14, 6.77)0.97 (0.14, 6.77)
 Nateglinide0.94 (0.06, 14.46)0.96 (0.14, 6.67)
AGIs1.07 (0.41, 2.78)1.55 (0.19, 12.51)1.92 (0.16, 22.74)2.57 (1.64, 4.03) *
 Acarbose0.97 (0.28, 3.31)2.15 (1.23, 3.75) *
 Voglibose0.93 (0.10, 8.78)0.90 (0.02, 45.04)0.92 (0.19, 4.46)
 Miglitol1.60 (0.20, 12.92)1.92 (0.16, 22.74)1.92 (0.16, 22.74)5.37 (2.11, 13.69) *
DPP-4is0.89 (0.51, 1.58)0.82 (0.47, 1.41)0.47 (0.19, 1.16)1.00 (0.14, 7.05)0.99 (0.14, 7.00)0.98 (0.14, 6.94)0.92 (0.74, 1.14)
 Sitagliptin0.82 (0.30, 2.20)0.68 (0.28, 1.66)0.56 (0.09, 3.53)1.03 (0.06, 16.28)0.34 (0.01, 8.36)0.89 (0.62, 1.28)
 Saxagliptin1.44 (0.49, 4.22)0.71 (0.29, 1.73)0.33 (0.08, 1.30)0.33 (0.01, 8.15)1.28 (0.68, 2.42)
 Vildagliptin0.48 (0.10, 2.36)1.08 (0.73, 1.60)
 Linagliptin0.85 (0.16, 4.50)0.95 (0.21, 4.25)0.31 (0.04, 2.52)0.55 (0.30, 0.99) *
 Alogliptin0.79 (0.18, 3.45)2.19 (0.39, 12.23)1.88 (0.16, 22.23)0.97 (0.06, 15.40)1.87 (0.16, 22.19)1.87 (0.16, 22.19)0.82 (0.42, 1.60)
SGLT2is0.81 (0.41, 1.60)1.00 (0.45, 2.21)0.79 (0.17, 3.64)1.00 (0.10, 9.50)0.58 (0.12, 2.88)1.53 (0.56, 4.18)0.89 (0.63, 1.24)
 Dapagliflozin1.07 (0.41, 2.80)1.19 (0.38, 3.68)1.19 (0.38, 3.68)1.66 (0.84, 3.27)
 Empagliflozin0.53 (0.15, 1.81)0.81 (0.21, 3.09)0.64 (0.08, 5.18)0.34 (0.04, 3.27)3.95 (0.44, 35.38)0.56 (0.36, 0.87) *
 Canagliflozin0.77 (0.17, 3.55)1.00 (0.10, 9.50)1.00 (0.10, 9.50)1.00 (0.10, 9.50)1.00 (0.10, 9.50)1.78 (0.78, 4.07)
GLP-1RAs0.89 (0.33, 2.43)0.39 (0.10, 1.57)0.67 (0.04, 10.13)0.67 (0.04, 10.13)0.67 (0.04, 10.13)0.80 (0.05, 12.41)1.23 (0.60, 2.54)
 Exenatide twice-daily0.96 (0.14, 6.73)0.86 (0.02, 40.01)0.86 (0.02, 40.01)0.86 (0.02, 40.01)0.86 (0.02, 40.01)0.86 (0.02, 40.01)2.63 (0.49, 14.04)
 Liraglutide0.86 (0.27, 2.79)0.35 (0.08, 1.55)0.51 (0.01, 25.66)0.51 (0.01, 25.66)0.51 (0.01, 25.66)0.75 (0.02, 37.38)0.63 (0.25, 1.61)
 Lixisenatide4.01 (0.85, 18.83)
For HbA1c, FPG, BMI, TC, HDL-C and SBP, data are WMD (95% CIs); for hypoglycemia, death, AE-induced discontinuations, total vascular events, myocardial infarction, heart failure, stroke, and diabetic nephropathy, data are RR (95% CIs). * Statistically significant differences. Table 1 represents a summary from Supplementary Figures S2–S103. Abbreviations: AE, adverse event; AGIs, α-glucosidase inhibitors; BMI, body mass index; CI, confidence interval; DPP-4is, dipeptidyl peptidase-4 inhibitors; FPG, fasting plasma glucose; GLP-1RAs, glucagon-like peptide-1 receptor agonists; HbA1c, hemoglobin Alc; HDL-C, high-density lipoprotein-cholesterol; LI, lifestyle intervention; NIDEs, glinides; PBO, placebo; RR, risk ratio; SBP, systolic blood pressure; SGLT2is, sodium-glucose cotransporter 2 inhibitors; SUs, sulfonylureas; TC, total cholesterol; TZDs, thiazolidinediones; WMD, weighted mean difference.
Table 2. Treatment effects of glucose-lowering drugs compared with each other.
Table 2. Treatment effects of glucose-lowering drugs compared with each other.
HbA1c, % (Left Lower Half) FPG, mmol/L (Right Upper Half)
Metformin1.05 (0.45, 1.66) *0.26 (−0.23, 0.75)−0.90 (−1.38, −0.42) *−0.46 (−1.12, 0.20)−0.71 (−1.10, −0.32) *−0.07 (−0.51, 0.37)−0.01 (−0.53, 0.51)
0.43 (0.12, 0.74) *SUs−0.79 (−1.36, −0.22) *−1.95 (−2.51, −1.39) *−1.51 (−2.23, −0.79) *−1.76 (−2.24, −1.28) *−1.12 (−1.65, −0.59) *−1.06 (−1.66, −0.46) *
−0.07 (−0.32, 0.18)−0.50 (−0.78, −0.22) *TZDs−1.16 (−1.59, −0.73) *−0.72 (−1.35, −0.09) *−0.97 (−1.30, −0.64) *−0.33 (−0.72, 0.06)−0.27 (−0.75, 0.21)
−0.52 (−0.84, −0.20) *−0.95 (−1.29, −0.61) *−0.45 (−0.74, −0.16) *NIDEs0.44 (−0.18, 1.06)0.19 (−0.12, 0.50)0.83 (0.46, 1.20) *0.89 (0.43, 1.36) *
−0.34 (−0.60, −0.08) *−0.77 (−1.06, −0.48) *−0.27 (−0.50, −0.04) *0.18 (−0.12, 0.48)AGIs−0.25 (−0.80, 0.30)0.39 (−0.20, 0.98)0.45 (−0.20, 1.10)
−0.33 (−0.54, −0.12) *−0.76 (−1.00, −0.52) *−0.26 (−0.42, −0.10) *0.19 (−0.06, 0.44)0.01 (−0.17, 0.19)DPP−4is0.64 (0.40, 0.88) *0.70 (0.33, 1.07) *
−0.16 (−0.37, 0.05)−0.59 (−0.84, −0.34) *−0.09 (−0.26, 0.08)0.36 (0.10, 0.62) *0.18 (−0.01, 0.37)0.17 (0.08, 0.26) *SGLT2is0.06 (−0.37, 0.49)
0.03 (−0.26, 0.32)−0.40 (−0.72, −0.09) *0.10 (−0.16, 0.36)0.55 (0.23, 0.87) *0.37 (0.10, 0.64) *0.36 (0.14, 0.58) *0.19 (−0.03, 0.41)GLP−1RAs
BMI, kg/m2 (Left Lower Half) TC, mmol/L (Right Upper Half)
Metformin0.10 (−0.72, 0.92)−0.31 (−0.57, −0.05) *−0.51 (−1.13, 0.11)−0.01 (−0.34, 0.32)−0.26 (−0.44, −0.08) *−0.52 (−0.71, −0.33) *0.12 (−0.14, 0.38)
−2.50 (−3.96, −1.04) *SUs−0.41 (−1.24, 0.42)−0.61 (−1.61, 0.39)−0.11 (−0.96, 0.74)−0.36 (−1.16, 0.44)−0.62 (−1.43, 0.19)0.02 (−0.80, 0.84)
−1.91 (−2.95, −0.87) *0.59 (−0.56, 1.74)TZDs−0.20 (−0.83, 0.43)0.30 (−0.06, 0.66)0.05 (−0.17, 0.27)−0.21 (−0.43, 0.01)0.43 (0.15, 0.71) *
−1.36 (−3.04, 0.32)1.14 (−0.61, 2.89)0.55 (−0.86, 1.96)NIDEs0.50 (−0.17, 1.17)0.25 (−0.35, 0.85)−0.01 (−0.62, 0.60)0.63 (−0.00, 1.26)
−0.79 (−2.03, 0.45)1.71 (0.38, 3.05) *1.12 (0.27, 1.97) *0.57 (−1.00, 2.14)AGIs−0.25 (−0.55, 0.05)−0.51 (−0.81, −0.21) *0.13 (−0.22, 0.48)
−1.75 (−2.84, −0.66) *0.75 (−0.44, 1.94)0.16 (−0.44, 0.76)−0.39 (−1.84, 1.06)−0.96 (−1.87, −0.05) *DPP−4is−0.26 (−0.37, −0.15) *0.38 (0.17, 0.59) *
−0.68 (−2.30, 0.94)1.82 (0.13, 3.51) *1.23 (−0.11, 2.57)0.68 (−1.20, 2.56)0.11 (−1.39, 1.61)1.07 (−0.31, 2.45)SGLT2is0.64 (0.43, 0.86) *
−0.23 (−1.47, 1.01)2.27 (0.94, 3.60) *1.68 (0.84, 2.52) *1.13 (−0.43, 2.69)0.56 (−0.52, 1.64)1.52 (0.62, 2.42) *0.45 (−1.05, 1.95)GLP−1RAs
HDL-C, mmol/L (Left Lower Half) SBP, mmHg (Right Upper Half)
Metformin−3.34 (−10.15, 3.47)−2.28 (−6.17, 1.61)4.48 (−2.27, 11.23)−0.10 (−4.12, 3.92)−1.52 (−4.07, 1.03)2.68 (0.30, 5.06) *1.48 (−1.36, 4.32)
0.05 (−0.13, 0.23)SUs1.06 (−6.08, 8.20)7.82 (−1.20, 16.84)3.24 (−3.97, 10.45)1.82 (−4.69, 8.33)6.02 (−0.42, 12.46)4.82 (−1.81, 11.45)
−0.07 (−0.13, −0.01) *−0.12 (−0.31, 0.07)TZDs6.76 (−0.33, 13.85)2.18 (−2.39, 6.75)0.76 (−2.58, 4.10)4.96 (1.74, 8.18) *3.76 (0.19, 7.33) *
−0.03 (−0.32, 0.26)−0.08 (−0.42, 0.26)0.04 (−0.25, 0.33)NIDEs−4.58 (−11.74, 2.58)−6.00 (−12.45, 0.45)−1.80 (−8.18, 4.58)−3.00 (−9.57, 3.57)
0.02 (−0.13, 0.17)−0.03 (−0.26, 0.20)0.09 (−0.06, 0.24)0.05 (−0.27, 0.37)AGIs−1.42 (−4.91, 2.07)2.78 (−0.59, 6.15)1.58 (−2.13, 5.29)
0.02 (−0.03, 0.07)−0.03 (−0.21, 0.15)0.09 (0.03, 0.15) *0.05 (−0.24, 0.34)0.00 (−0.15, 0.15)DPP−4is4.20 (2.90, 5.50) *3.00 (0.98, 5.02) *
−0.04 (−0.08, 0.00)−0.09 (−0.27, 0.09)0.03 (−0.02, 0.08)−0.01 (−0.30, 0.28)−0.06 (−0.21, 0.09)−0.06 (−0.10, −0.02) *SGLT2is−1.20 (−3.01, 0.61)
0.02 (−0.03, 0.07)−0.03 (−0.21, 0.15)0.09 (0.03, 0.15) *0.05 (−0.24, 0.34)0.00 (−0.15, 0.15)0.00 (−0.05, 0.05)0.06 (0.02, 0.10) *GLP-1RAs
Hypoglycemia (Left Lower Half) Death (Right Upper Half)
Metformin0.80 (0.17, 3.80)0.93 (0.36, 2.39)0.92 (0.17, 5.09)0.83 (0.26, 2.63)0.99 (0.42, 2.35)1.09 (0.42, 2.79)0.99 (0.30, 3.29)
0.28 (0.10, 0.81) *SUs1.15 (0.24, 5.55)1.15 (0.14, 9.57)1.03 (0.19, 5.67)1.23 (0.27, 5.65)1.36 (0.28, 6.51)1.24 (0.22, 7.01)
3.13 (1.32, 7.42) *11.08 (3.33, 36.87) *TZDs0.99 (0.18, 5.58)0.89 (0.28, 2.90)1.07 (0.44, 2.61)1.18 (0.45, 3.09)1.07 (0.32, 3.62)
1.12 (0.26, 4.90)3.97 (0.73, 21.66)0.36 (0.07, 1.76)NIDEs0.90 (0.14, 5.71)1.08 (0.20, 5.78)1.19 (0.21, 6.64)1.08 (0.17, 7.05)
1.79 (0.89, 3.57)6.33 (2.14, 18.73) *0.57 (0.23, 1.42)1.60 (0.36, 7.16)AGIs1.20 (0.39, 3.64)1.32 (0.41, 4.27)1.20 (0.30, 4.82)
1.73 (1.02, 2.95) *6.14 (2.28, 16.51) *0.55 (0.25, 1.22)1.55 (0.37, 6.49)0.97 (0.53, 1.76)DPP-4is1.10 (0.46, 2.67)1.01 (0.32, 3.19)
1.78 (0.93, 3.38)6.30 (2.20, 18.05) *0.57 (0.24, 1.36)1.59 (0.36, 6.96)0.99 (0.49, 2.00)1.03 (0.60, 1.76)SGLT2is0.91 (0.27, 3.07)
0.97 (0.44, 2.15)3.45 (1.09, 10.91) *0.31 (0.12, 0.84) *0.87 (0.19, 4.10)0.55 (0.24, 1.26)0.56 (0.28, 1.15)0.55 (0.25, 1.22)GLP-1RAs
Total Vascular Events (Left Lower Half) AE-Induced Discontinuations (Right Upper Half)
Metformin0.46 (0.14, 1.46)0.82 (0.48, 1.42)1.07 (0.26, 4.37)0.40 (0.23, 0.70) *1.12 (0.76, 1.66)1.16 (0.73, 1.85)0.84 (0.38, 1.85)
1.99 (0.09, 42.75)SUs1.79 (0.54, 5.91)2.33 (0.40, 13.59)0.88 (0.27, 2.90)2.45 (0.79, 7.58)2.53 (0.80, 8.06)1.82 (0.49, 6.85)
0.63 (0.09, 4.37)0.32 (0.02, 6.59)TZDs1.30 (0.31, 5.48)0.49 (0.26, 0.92) *1.37 (0.84, 2.23)1.41 (0.81, 2.46)1.02 (0.44, 2.37)
NIDEs0.38 (0.09, 1.59)1.05 (0.26, 4.22)1.09 (0.27, 4.46)0.78 (0.17, 3.69)
0.59 (0.05, 7.30)0.30 (0.01, 9.16)0.93 (0.08, 10.98)AGIs2.80 (1.70, 4.61) *2.89 (1.65, 5.07) *2.08 (0.89, 4.88)
1.12 (0.25, 5.08)0.56 (0.04, 9.07)1.76 (0.42, 7.38)1.90 (0.22, 16.48)DPP-4is1.03 (0.69, 1.54)0.75 (0.35, 1.58)
0.91 (0.18, 4.61)0.46 (0.03, 7.86)1.44 (0.31, 6.74)1.56 (0.17, 14.53)0.82 (0.31, 2.15)SGLT2is0.72 (0.33, 1.60)
2.33 (0.32, 16.87)1.17 (0.06, 24.97)3.67 (0.54, 25.02)3.96 (0.32, 48.69)2.08 (0.47, 9.30)2.55 (0.51, 12.66)GLP-1RAs
Treatment estimates are WMD (95% CIs) (or RR (95% CIs)) of the column-defining treatment compared with the row-defining treatment for HbA1c, BMI (or hypoglycemia, total vascular events) (left lower half), and WMD > 0 (or RR > 1) favor the row-defining treatment. Treatment estimates are WMD (95% CIs) of the column-defining treatment compared with the row-defining treatment for HDL-C (left lower half), and WMD > 0 favor the column-defining treatment. Treatment estimates are WMD (95% CIs) (or RR (95% CIs)) of the row-defining treatment compared with the column-defining treatment for FPG, TC, SBP (or death, AE-induced discontinuations) (right upper half), and WMD > 0 (or RR > 1) favor the column-defining treatment. * Statistically significant differences. Abbreviations: AE, adverse event; AGIs, α-glucosidase inhibitors; BMI, body mass index; CI, confidence interval; DPP-4is, dipeptidyl peptidase-4 inhibitors; FPG, fasting plasma glucose; GLP-1RAs, glucagon-like peptide-1 receptor agonists; HbA1c, hemoglobin Alc; HDL-C, high-density lipoprotein-cholesterol; NIDEs, glinides; RR, risk ratio; SBP, systolic blood pressure; SGLT2is, sodium-glucose cotransporter 2 inhibitors; SUs, sulfonylureas; TC, total cholesterol; TZDs, thiazolidinediones; WMD, weighted mean difference.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gu, S.; Hu, X.; Shi, L.; Zhen, X.; Sun, X.; Huang, M.; Gu, Y.; Dong, H. Choice of Glucose-Lowering Drugs as Initial Monotherapy for Type 2 Diabetes Patients with Contraindications or Intolerance to Metformin: A Systematic Review and Meta-Analysis. J. Clin. Med. 2022, 11, 7094. https://doi.org/10.3390/jcm11237094

AMA Style

Gu S, Hu X, Shi L, Zhen X, Sun X, Huang M, Gu Y, Dong H. Choice of Glucose-Lowering Drugs as Initial Monotherapy for Type 2 Diabetes Patients with Contraindications or Intolerance to Metformin: A Systematic Review and Meta-Analysis. Journal of Clinical Medicine. 2022; 11(23):7094. https://doi.org/10.3390/jcm11237094

Chicago/Turabian Style

Gu, Shuyan, Xiaoqian Hu, Lizheng Shi, Xuemei Zhen, Xueshan Sun, Minzhuo Huang, Yuxuan Gu, and Hengjin Dong. 2022. "Choice of Glucose-Lowering Drugs as Initial Monotherapy for Type 2 Diabetes Patients with Contraindications or Intolerance to Metformin: A Systematic Review and Meta-Analysis" Journal of Clinical Medicine 11, no. 23: 7094. https://doi.org/10.3390/jcm11237094

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

Article Metrics

Back to TopTop