72
Views
0
CrossRef citations to date
0
Altmetric
Review

Patient profiling in diabetes and role of canagliflozin

Pages 367-377 | Published online: 22 Dec 2022

Abstract

Background

Physicians attempt to achieve glycemic goals in patients with type 2 diabetes mellitus (T2DM) through various means, including glucose-lowering medications. There is interindividual variability in response to medications, which can be partially explained by the presence of genetic polymorphisms that affect drug metabolism. Pharmacogenomics studies the hereditary basis of interpatient variations in drug response and aims to identify subgroups of patients whose drug management could be tailored accordingly. The aim of this review is to explore patient profiling in the management of T2DM with a focus on the sodium glucose transporter inhibitor canagliflozin.

Methods

The PubMed database was searched using the terms “pharmacogenomics” and “diabetes” through May 31, 2014. Published articles and abstracts presented at national/international meetings were considered.

Results and conclusion

Genome-wide association studies have opened the door for patient profiling and research into genetic variants in multifactorial T2DM. Clinically, it may be possible to tailor the type of medication used based on the presence or absence of the various genetic variants. However, the polymorphisms studied may only explain some of the variability in response to T2DM drugs and needs further validation to ensure its authenticity. There are still unidentified factors which appear to play a role in the interindividual variability seen in clinical practice. The potential exists for pharmacogenomics to promote efficacious, safe, and cost-effective individualized diabetes management. Pharmacogenomics is still in its early stages, and the idea of defining patients genetically to predict individual responses to drugs and obtain safe and effective T2DM management is promising, in spite of existing barriers. Currently, clinical profiling of patients with T2DM and using an individualized approach with most drugs, including canagliflozin, based on comorbid conditions still remains the most accepted approach for the management of T2DM.

Introduction

The primary goal of type 2 diabetes mellitus (T2DM) management is to prevent long-term complication. Even with various means, including glucose-lowering medications, there are still barriers to achieving and maintaining glycemic goals.Citation1 In the 2012 position statement, the American Diabetes Association emphasized that individualizing the management with consideration to different clinical factors is important.Citation2 However, it is difficult for the guidelines to take into account individual variation in responses to pharmacological and lifestyle interventions. Physicians attempt to achieve glycemic goals through an algorithmic approach based on the patient’s responses to various antihyperglycemic agents. The wide interpatient variability seen in clinical practice can be partially explained by the presence of genetic polymorphisms which affect the absorption, distribution, metabolism, and excretion of drugs.

Pharmacogenomics studies the hereditary basis of interpatient variations in drug response and aims to identify subgroups of patients whose drug management could be tailored accordingly. This can be an important step in true individualization of therapy, which is lacking at present. The aim of this review is to explore patient profiling in the management of T2DM with a focus on the sodium glucose transporter inhibitor canagliflozin.

Genetic variants associated with response to oral antihyperglycemic agents metformin

The American Diabetes Association recommends metformin as the first line drug to be used in patients with T2DM.Citation3 Response to metformin can vary significantly at the individual level and cannot be predicted by any features found clinically.Citation4 In fact, in 2,064 T2DM patients, the glycemic response to metformin was similar in nonobese and obese patients.Citation4 Metformin is hydrophilic and needs active transportation into the tissues.Citation5 The main transporters are the PMAT encoded by the gene SLC29A4, OCT1 encoded by gene SLC22A1, OCT2 encoded by gene SLC22A2 and OCT3 encoded by gene SLC22A3.Citation6Citation8 In the renal cell, there are two other transporters called MATE1 and MATE2 encoded by genes SLC47A1 and SLC47A2, respectively.Citation8

In a retrospective study in 24 responders and nine nonresponders of metformin in T2DM patients, the single nucleotide polymorphisms (SNPs) of SLC22A1, −43T>G in intron 1, was a negative predictor of metformin response while the variant 408 Met >Val (1222A>G) in exon 7 was a positive predictor, with a predictive accuracy of 55.5% (P<0.05).Citation9 In the Rotterdam study, with 102 T2DM patients, where associations between eleven SNPs in the SLC22A1 gene and change in the A1c were analyzed, the SNP rs622342 was associated with the glucose-lowering effect of metformin.Citation7 For each minor C allele at rs622342, the A1c levels were reduced by 0.28% (95% confidence interval [CI] 0.09–0.47, P=0.005).Citation7

A genome-wide association study on glycemic response to metformin in 1,024 Scottish patients with T2DM was done, and replication was attempted in two cohorts of 1,783 Scottish patients and 1,113 patients from the United Kingdom Prospective Diabetes Study (UKPDS).Citation10

In 1,024 Scottish observational genetic cohort on metformin therapy, the minor C allele of the most strongly associated SNP, rs11212617, had a frequency of 44% and was associated with treatment success (A1c <7%), with an allelic odds ratio (OR) of 1.64 (95% CI 1.37–1.99, P=1.9×10−7), and it was associated with lower treatment A1c (per allele β −0.18% [95% CI −0.26 to −0.1], P=1.8×10−5). In the two replication cohorts, the minor C allele of rs11212617 was associated with treatment success: allelic OR 1.21, 95% CI 1.05–1.38, P=0.007 and allelic OR 1.37, 95% CI 1.1–1.72, P=0.006, respectively.

The meta-analysis of all 3,920 patients identified a SNP, rs11212617 of the ATM gene, known to be involved in DNA repair and cell cycle control, which was associated with treatment success (OR −1.35, 95% CI 1.22–1.49, P=2.9×10−9), and each copy of the minor C allele of rs11212617 was associated with 0.11% (P=6.6×10−7) lower absolute treatment A1c.Citation10

Further meta-analysis of 2,264 patients on metformin monotherapy showed that the OR for treatment success was 1.42 (95% CI 1.26–1.62, P=4×10−8).Citation10 In a subgroup analysis of 284 patients on metformin from the UKPDS study, 19% of patients who had two copies of the C allele at rs11212617 had a 3.3-fold greater likelihood of achieving an A1c <7%, which increased the variance in the response to metformin by 2.5% from 27.5% to 30% (P=0.007).Citation10

In the South Danish Diabetes Study, 27 SNPs and one deletion of the metformin transporter genes were tested in 371 T2DM patients.Citation11 The mean trough steady-state plasma concentration of metformin was 576 ng/mL, with a wide, 80-fold range (54–4,133 ng/mL), indicating that it was a true interindividual variability. Patients heterozygous for the minor alleles rs72552763, rs12208357, rs34130495, and rs34059508 in SLC22A1 showed a significantly lower trough concentration of metformin, which was additive with increasing number of reduced function SNPs.Citation11 A large difference in the levels of OCT1/OCT3 genetic variants expression in the liver, kidney, and intestines may contribute to reduced intestinal absorption, variations in hepatic uptake, increased renal clearance, and reduced clinical effect of metformin.Citation11 The variant rs3413095 in SLC22A1 was associated with a 1.1% (95% CI 0.4–1.8, P=0.003) higher A1c compared to patients who were homozygous for the wild-type allele at 6 months. The presence of SNP rs34399035 of SLC47A2 in the heterozygous patients was thought to be associated with a long-term decrease in A1c 1.1% (95% CI −0.1–2.2, P=0.06) compared with the wild-type patients over 24 months.Citation11

In 52 men with heart disease and metabolic syndrome or T2DM who were compared to 46 healthy controls, metformin exhibited a high therapeutic efficacy in patients who carried the Pro/Pro allele of the SNP Pro12Ala polymorphism of the PPARγ2 gene.Citation12 These patients had significant reduction in weight, waist circumference, body mass index (BMI), and concentrations of total cholesterol and inflammatory factors like interleukin (IL)-1beta, IL-6, IL-8, and tumor necrosis factor-alpha.Citation12

Sulphonylureas and meglitinides

Sulphonylureas (SU), one of the oldest oral agents used in T2DM, stimulates insulin secretion by inhibiting adenosine triphosphate (ATP)-sensitive potassium (KATP) channels.Citation3 Meglitinides act similarly but produce a less sustained rise in insulin secretion. SU have been well studied as they have implication in the treatment of monogenic diabetes.Citation13 Gene mutations in the inwardly rectifying potassium channel (KCNJ11) and the ABCC8 modify the insulin response to SU therapy and may be responsible for the interindividual variability and adverse effects seen in T2DM patients.Citation13

SU given to 101 T2DM patients uncontrolled on metformin monotherapy showed a higher decrease in A1c at 6 months in K allele carriers of the KCNJ11 gene variant E23K when compared with EE homozygotes (1.04%±0.10% versus 0.79%±0.12%, P=0.036, respectively).Citation14 K allele carriers of the E23K variant of the KCNJ11 geneCitation15 and the Gly972Arg polymorphism of the insulin receptor substrate 1 (IRS1)Citation16 were more susceptible to secondary failure, defined as requiring insulin secondary to plasma glucose levels >300 mg/dL. In 525 T2DM patients, the frequency of carriers of the K allele of the SNP E23K of the KCNJ11 gene (E23K heterozygotes and K23K homozygotes) was higher (66.8%) in patients who failed SU therapy compared to 58% in patients who did not fail, and they had a significant relative risk of secondary failure of 1.45 (95% CI 1.01–2.09, P=0.04) compared with E23E homozygotes, which did not change, even after adjustment for clinical factors like age, sex, fasting plasma glucose (FPG), A1c, BMI, age at diagnosis, and duration of diabetes (OR 1.69, 95% CI 1.02–2.78, P=0.04).Citation15 In 477 Caucasian T2DM patients, the Gly972Arg polymorphism of the IRS1 gene was found in 8.7% of well-controlled T2DM patients on oral therapy versus 16.7% in patients with secondary failure to SU.Citation16 This SNP was associated with relative risk of 2.7 (1.02–7.28, P=0.045) to 2.0 (1.38 –3.86, P=0.038) of secondary SU failure, which persisted even after adjustment for various clinical factors like age, sex, FPG, A1c, BMI, age at diagnosis, and duration of diabetes.Citation16

KATP channels containing the K23/A1369 haplotype of the genes KCNJ11 and ABCC8 were significantly less sensitive to inhibition by tolbutamide, chlorpropamide, and glimepiride and more sensitive to inhibition by mitiglinide and gliclazide when compared to the SNP E23/S1369.Citation17 Nateglinide, glipizide, and glibenclamide showed similar inhibitory profiles in KATP channels containing either haplotype.Citation17

Carriers of the T-allele (TT + TG) of the SNP rs163184 (T>G) of another diabetes susceptibility gene expressed in β-cells (KCNQ1) achieved significantly lower FPG (6.95 mmol/L [125 mg/dL]) 6 months after therapy with SU in 87 T2DM patients uncontrolled on metformin monotherapy when compared to the GG genotype (7.50 mmol/L [135 mg/dL]).Citation18 In 367 Chinese T2DM patients on repaglinide, those with the rs2237892 risk C allele of the gene KCNQ1 had a significantly lower fasting insulin (FI) and homeostasis model assessment of insulin resistance (HOMA-IR) levels than carriers of the T allele.Citation19 Patients with the rs2237895 risk C allele had significantly higher FPG, postprandial plasma glucose (PPPG), and HOMA-IR values when compared to patients with the A allele. Thus, response to repaglinide treatment may be better in patients with the rs2237892 T allele and rs2237895 C allele compared to those with the rs2237892 CC and rs2237895 AA genotypes.Citation19 Similarly, in 209 newly diagnosed T2DM Chinese patients given repaglinide for 48 weeks, the presence of the rs2237892 TT homozygotes showed a significantly lower PPPG (P=0.03) when compared to the C allele carriers.Citation20

To assess which other genetic polymorphisms are associated with metabolism of repaglinide, 368 Chinese patients with T2DM were compared to 132 healthy controls.Citation21 The frequency of allele T45 of the gene NeuroD1/BETA2 was significantly higher in T2DM patients (13.45%) than in the controls (6.8%, P<0.01, OR =2.342 [95% CI 1.365–4.019], P=0.002). Patients with the A45T polymorphism had higher FI and postprandial serum insulin (PPI) compared to the subjects with the T allele. These T allele subjects, after repaglinide therapy, were shown to have attenuated FPG and PPPG levels.Citation21 In R121W R allele carriers of the PAX4 gene, the RR genotype had higher PPI levels and better lowering of both FPG and PPPG levels when compared to the R/W genotypes (−6.53±6.52 versus −2.95±1.17 mmol l (−1), P<0.05).Citation21 In 443 Chinese T2DM patients on repaglinide for 8 weeks and 229 healthy volunteers, rs13266634 risk C allele of the gene SLC30A8 was found in a significantly higher frequency in T2DM patients than in controls (P<0.05).Citation22 The rs13266634 CT + TT genotypes had higher FI and PPI levels compared with CC genotype carriers. T2DM patients with rs16889462 GA genotype of the gene SLC30A8 showed a higher reduction in FPG, PPPG, and A1c with repaglinide therapy compared with subjects with the GG genotype.Citation22

Thiazolidinediones

Thiazolidinediones act as insulin-sensitizers through activation of the transcription factor peroxisome proliferator-activated receptor-gamma 2 (PPARγ2), which plays a pivotal role in adipocyte differentiation and expression of adipocyte-specific genes.Citation3 The efficacy of rosiglitazone (4 mg/day for 12 weeks) in relation to PPARγ2 genetic variations was studied in 198 T2DM patients.Citation23 The presence of the Ala12 allele lead to significantly lower FPG and A1c (2.8 mmol/L [50.6 mg/dL] and 1.41%) when compared to those without the allele (1.3 mmol/L [24.3 mg/dL] and 0.57% reduction, P=0.26 and P=0.01, respectively).Citation23 In addition, there was a significant difference in the response rate to rosiglitazone therapy between the Pro12Pro and the Pro12Ala variant groups (43.72% versus 86.67%).Citation23

In 250 Chinese T2DM patients on pioglitazone (30 mg/day for 24 weeks), the Ala12Ala and Pro12Ala genotypes and Ala allele of the PPARγ gene were significantly more frequent in pioglitazone responders than in nonresponders (26.0% versus 13.5%, P=0.025 and 15.6% versus 7.3%, P=0.008).Citation24 The decrease in FPG (2.8 mmol/L [50.4 mg/dL] versus 2.4 mmol/L [43.3 mg/dL], P<0.001) and A1c (0.57% versus 0.35%, P=0.004) levels was significantly greater in subjects with the Pro12Ala and Ala12Ala carriers than in Pro12Pro carriers. On further analysis, only the PPARγ Pro12Ala polymorphism was found to be associated with the response to pioglitazone.Citation24 This finding was not replicated in 113 T2DM patients from Germany, where both Pro12Ala and the Pro12Pro variants were not associated with response to pioglitazone.Citation25 In 101 Iranian T2DM patients, even though the Pro12Ala genotype showed a better therapeutic response compared to the Pro12Pro genotype, the difference between groups was not statistical significance.Citation26 However, a recent meta-analysis showed that patients with PPARγ gene Pro12Ala polymorphism had a more favorable change in FPG with pioglitazone (P=0.018) when compared with the Pro12Pro genotype.Citation27

In 197 Chinese Han T2DM patients of whom 67 patients were given pioglitazone (30 mg/day for 12 weeks), those patients with PPARγ2 gene polymorphism rs1801282 CG showed significantly higher levels of PPPG and serum triglyceride compared with those with rs1801282 CC genotype.Citation28 In patients with protein tyrosine phosphatase receptor type D gene polymorphism rs17584499, the CC genotype had better PPPG levels compared with CT + TT genotypes.Citation28 In 241 Chinese patients with T2DM of whom 41 patients were given rosiglitazone (4 mg daily for 12 weeks), the presence of the SNPs Thr394Thr and Gly482Ser of the PPARγ coactivator-1 gene increased the therapeutic efficacy of rosiglitazone when compared to controls.Citation29

Other genes related to leptin, resistin, and adiponectin have been studied. In 245 Chinese patients with T2DM and 122 healthy volunteers, the frequency of the SNPs of the leptin gene G-2548A and TNF-alpha gene G-308A were the same.Citation30 At baseline in 42 T2DM patients on rosiglitazone (4 mg daily for 8 weeks), those with the G allele of leptin G-2548A had a higher FPG levels compared to the AA genotypes (P<0.05). The effect on FI and PPI were enhanced with rosiglitazone therapy in patients with AA genotype of leptin G-2548A when compared with GG + GA genotype. There was an attenuated effect on FI in patients with GA + AA genotype of TNF-alpha G-308A compared with GG genotype (P<0.05).Citation30

A prospective study of 121 T2DM patients and retrospective data from 63 T2DM patients treated with pioglitazone for 12 weeks revealed that the G/G but not C/G genotype of the resistin gene promoter SNP at −420, rs1862513 had lower FPG (P=0.020) and better HOMA-IR (P=0.012) when compared to C/C genotype.Citation31 Patients on rosiglitazone monotherapy (4 mg daily for 12 weeks) with SNP rs11377CC homozygote genotype and 45TG + GG heterozygote genotype of the adiponectin common polymorphisms 45T/G and −11377C/G had a better response in FPG, PPPG, HOMA-IR, and serum adiponectin levels (P=0.000) compared with 11377CG + GG heterozygote and 45TT homozygote genotypes.Citation32 This was contradicted in another study of Chinese T2DM patients, where the 11377CG genotype had a better response in FPG and A1c when compared to the 11377 CC polymorphism.Citation33 The presence of the two SNPs, 45T/G and 795G/A, in 101 Iranian patients with T2DM on pioglitazone for 12 weeks were not significantly associated with the response to pioglitazone.Citation34

Genetic polymorphisms have been studied in other genes such as KCNQ1, UCP2, ADRB3, and LPL. The presence of rs2237897 allele of the KCNQ1 gene was associated with significant decrease in PPPG levels in 209 Chinese T2DM patients given rosiglitazone for 48 weeks.Citation20 The frequency of the allele UCP2 −866G/A and ADRB3 Trp64Arg genetic polymorphisms were the same in 472 Chinese T2DM patients and 321 healthy volunteers.Citation35 At baseline, the A allele carriers of UCP2 had significantly lower PPI (61.5±34.3 versus 41.6±28.7 mU l (−1), P<0.01) and low-density lipoprotein-cholesterol (LDL-C) (3.4±1.1 versus 2.7±1.1 mmol l (−1), P<0.05) compared with GG genotypes. After rosiglitazone 4 mg/day for 12 weeks there was improvement in PPI (−3.82±13.2 versus −42.1±30.7 mU l (−1), P<0.01) (9.45−51.31 versus 0.48–11.88 mU1[–1]) and greater attenuation of A1c (−1.85±1.62 versus −0.61±0.80%, P<0.05) (0.14–1.37 versus 1.10–2.38%) compared with GG genotypes.Citation35 Patients with ADRB3 Trp64Arg when compared with ADRB3 Trp64Trp polymorphism had a better response in serum triglyceride, LDL-C, and adiponectin levels.Citation35

In 113 T2DM patients on pioglitazone (30 mg/day for 10 weeks), the presence of the SNP S447S genotype of the LPL gene had a significantly better response in FPG and A1c reduction, along with beneficial effects on lipid profile and blood pressure (BP) when compared to the S447X genotype.Citation36

Glucagon-like peptide-1 analogs and dipeptidyl peptidase-4 inhibitors incretins

Glucagon-like peptide-1 (GLP-1) analogs and dipeptidyl peptidase-4 (DPP-4) inhibitors, through their receptors on pancreatic α/β cells, stimulate the synthesis of insulin and suppress glucagon levels.Citation37 The speculation is that any of the genetic variants which affect insulin synthesis/secretion may also affect the incretin response.Citation38

The effect of 21 SNPs in GLP-1 receptor gene on insulin secretion was studied in 88 healthy young individuals, using a hyperglycemic clamp with GLP-1 being infused for the last 2 hours.Citation37 Two of 21 SNPs (rs6923761 and rs3765467) were associated with altered response of the β-cell to the GLP-1 infusion, which may indicate that variations in the GLP-1 receptor may alter insulin secretion in response to exogenous GLP-1.Citation37 However, this needs to be confirmed in T2DM patients with larger studies.

In 1,578 German subjects at high risk to develop diabetes, the effect of KCNQ1 gene SNPs rs151290, rs2237892, rs2237895, and rs2237897 on insulin secretion in response to oral glucose tolerance test (OGTT), intravenous GTT (IVGTT), and to hyperglycemic clamp combined with GLP-1 and arginine administration was studied.Citation38 Insulin secretion was lower in homozygous major allele carriers of SNPs, rs151290, rs2237892, and rs2237897 while the β-cell function was improved in homozygous major allele carriers of rs2237895 (AA). However, during the hyperglycemic clamp combined with GLP-1 administration, there were no associations found between the KCNQ1 variants and glucose, GLP-1, and insulin secretion.Citation38 In a subset of patients, there was a significant increase in OGTT-stimulated gastric inhibitory polypeptide (GIP) levels (P=0.019) with the SNP rs151290, even after adjustment for sex, age, and BMI. This may suggest that an altered incretin secretion after food intake provides a potential link between KCNQ1 gene variants and impaired β-cell function. This effect was not found with other variants, rs2237892, rs2237895, and rs2237897, and the authors concluded that this could be secondary to limited sample size or that these KCNQ1 variants regulate insulin secretion differently than rs151290.Citation38

The transcription factor T-cell factor 7-like-2 (TCF7L2) is an essential factor for GLP-1 secretion from intestinal L-cells.Citation39 The interaction between TCF7L2 and GLP-1/GIP receptors is important for β-cell function.Citation40 When islet cells of patients with T2DM were treated with small interfering RNA to TCF7L2, the expression of the GLP-1 and GIP receptors were decreased, and there was impaired insulin secretion when stimulated by glucose and GLP-1/GIP.Citation39 Of the five SNPs in the TCF7L2 gene which were genotyped in 1,110 nondiabetic German participants, there was a significant reduction in GLP-1-induced insulin secretion in carriers of rs7903146 and rs12255372 (P<0.02), which may be the result of a functional defect in GLP-1 signaling in β-cells rather than a reduction in GLP-1 secretion.Citation40 In the presence of TCF7L2 gene variants (TT or TC at rs7903146) in eight subjects with high risk to develop T2DM, the β-cell response to OGTT was 50% lower (47±4 versus 95±15×109 min (−1), P=0.01), and the incretin effect was also reduced by 30% (32%±4% versus 46%±4%, P=0.02) when compared to ten matched subjects with wild-type genotype, while the response was similar in both groups to IVGTT.Citation41 The lower incretin effect occurred in spite of having the same level of GLP-1/GIP secretion to oral glucose in both groups, indicating that it may be secondary to the effect of TCF7L2 gene variants on the sensitivity of the β-cell to incretins.Citation41

The WFS1 gene plays a significant role in the maintenance of homeostasis of the endoplasmic reticulum in pancreatic β-cells.Citation42 In 1,578 subjects with high risk to develop T2DM, the SNP rs10010131 of the WFS1 gene was associated with reduced OGTT and GLP-1 infusion combined with a hyperglycemic clamp-derived insulin secretion (P=0.03), with no difference between groups in IVGTT and hyperglycemic clamp.Citation42

Gene variants of JAZF1, CDC123/CAMK1D, TSPAN8/LGR5, THADA, ADAMTS9, NOTCH2/ADAMS30, DCD VEGFA, BCL11A, HNF1B, WFS1, and MTNR1B were evaluated in a study with 180 subjects with normal glucose tolerance and 156 subjects with impaired glucose tolerance.Citation43 Certain gene variants in CDC123/CAMK1D, ADAMTS9, BCL11A, and MTNR1B affected various aspects of the insulin response to glucose (all P<6.9×10−3) while the THADA gene variant was associated with lower β-cell response to GLP-1 and arginine (both P<1.6×10−3). In carriers of MTNR1B, the trend seen toward an increased insulin response to GLP-1 (P=0.03), if confirmed, may be important clinically.Citation43

The SNPs of the gene ABCB1, CGC/CGC, CGC/TTT, and TTT/TTT, did not influence sitagliptin pharmacokinetics in ten healthy volunteers.Citation44 Various SNPs of the DPP-4 enzyme genes, g-234A/C, rs13015258, IVS8-128A/G, rs17848920, IVS8 + 46C/T, rs10930040, IVS11-143A/G, rs2302873, G645G, rs17848910, IVS22 + 4C/T, and rs2268891, studied to assess the efficacy of vildagliptin, showed no significant associations, and the authors hypothesized that it could be secondary to a small sample size.Citation45 DPP-4 gene variants could affect the efficacy of the DPP-4 inhibitors; however, there is no study currently proving that a patient with a specific variant in a specific gene shows a better response to a particular DPP-4 inhibitor. The presence or absence of genetic variants regarding clinical response to incretin therapy, if predictive, could lead to cost savings.

Sodium glucose transport 2 inhibitors

Sodium glucose transport 2 (SGLT2) inhibitors block the reabsorption of filtered glucose, leading to glucosuria, with improvements in glycemic control and weight loss. The SGLT2 gene (SLC5) has been mapped to chromosome 16 p11.2,Citation46 and up to 50 different mutations of this gene have been reported and lead to familial renal glucosuria.Citation47,Citation48 Genotyping for four common genetic variations in the SLC5 gene in 1,013 nondiabetic German subjects showed that the SNP rs9934336 G allele was found with increased 30-minute plasma glucose, 120-minute insulin concentrations, and 120-minute area under the curve glucose during OGTT.Citation49 In 2,590 nondiabetic subjects from Berlin, there was a nominal association with the 120-minute insulin concentrations, suggesting a possible role in the regulation of glucose homeostasis for SLC5 gene variants.Citation49 So far, there have been no definitive studies of patients with T2DM regarding the genetic variants and SNPs associated with response to the SGLT2 inhibitors.

Canagliflozin: role of clinical profiling – efficacy and adverse events data

Effect on glycemic parameters and weight

In multiple Phase III studies lasting from 26–52 weeks, a total of 4,978 patients who were either treatment-naïve or on mono/dual oral agents were randomized to canagliflozin 100 and 300 mg daily versus placebo, glimiperide, and sitagliptin.Citation50Citation55 The placebo-corrected reduction in A1c, FPG, and PPPG with canagliflozin 100 and 300 mg were significant. The A1c reductions with canagliflozin 100 and 300 mg ranged from 0.63–0.91% versus 0.97–1.16%, respectively.Citation50Citation52 The FBG and PPBG reductions with cangliflozin 100 mg and 300 mg ranged from 1.2–2.0 mmol/L (21.6–36 mg/dL) and 2.7 mmol/L (48.6 mg/dL) versus 1.92–2.4 mmol/L (34.5–43.2 mg/dL) and 3.6 mmol/L (63.8 mg/dL), respectively.Citation50Citation52 Weight loss of 1.4–3.0 kg was found with canagliflozin 100 mg and 2.0–3.8 kg with canagliflozin 300 mg. The decrease in A1c was similar between treatment-naïve patients and those already on oral antihyperglycemic therapy.Citation50Citation52

The mean A1c and FPG reduction with the canagliflozin 100 mg dose was noninferior to glimiperide, 0.82% and 0.81% and 1.4 and 1.0 mmol/L (25.2 and 18 mg/dL), respectively, while the canagliflozin 300 mg group showed superiority (0.93% and 1.5 mmol/L [27 mg/dL] reduction, respectively).Citation53 The A1c reduction was sustained for 52 weeks in the canagliflozin groups while it started to rise after week 18 in the glimiperide group. A mean weight reduction of 4.2–4.7 kg was noted with canagliflozin compared to 1.0 kg weight gain with glimiperide and was predominantly from fat mass as shown by a reduced percent total fat,Citation53,Citation56 and accounted for two-thirds of the overall reduction, with slightly greater reductions in visceral fat.Citation56 In the first study, where canagliflozin was compared to sitagliptin, there was a mean reduction in A1c and FPG in the canagliflozin (1.03% and 1.7 mmol/L [30.6 mg/dL]) versus sitagliptin (0.66% and 0.3 mmol/L [5.4 mg/dL]).Citation56 As with SU, there was an increase in A1c after week 12 in the sitagliptin group. A mean weight reduction of 2.5 kg was noted with canagliflozin compared to 0.3 kg weight gain with sitagliptin. The A1c goals of <7% were reached in 47.6% with canagliflozin and 35.3% with sitagliptin.Citation54 In the second study at week 52, the mean A1c reduction in canagliflozin 100 and 300 mg versus sitagliptin was comparable. Body weight decreased in canagliflozin 100 and 300 mg (3.8%–4.2%) versus sitagliptin (1.3%).Citation55

Data of 2,313 patients (1,868 <65 years and 445 >65 years) from four Phase III studies showed that canagliflozin was efficacious in attaining a reduction in A1c, FPG, body weight, and systolic BP (SBP) in both groups when compared to placebo.Citation57 However, data extraction from 15 clinical trials showed that in elderly T2DM patients, the A1c reduction was lower with canagliflozin when compared to younger population, while the reduction in SBP and weight was the same.Citation58 In multiple Phase III studies, there was a significant increase in β-cell glucose sensitivity compared to placebo with canagliflozin, and it was noninferior to sitagliptin. This indicates that canagliflozin given for 26–52 weeks improved measures of β-cell function.Citation59

Effect on BP, pulse rate, and lipids

Canagliflozin showed a significant reduction in the placebo-corrected mean SBP and diastolic BP in both dose groups, ranging from 1.62–10.7 mmHg and 1.6–7.0 mmHg, respectively.Citation50Citation52 Canagliflozin showed a mean decrease in SBP (3.3–5.1 mmHg) versus an increase in the glimiperide (0.2 mmHg) and sitagliptin (0.9 mmHg) groups.Citation53,Citation54 Both canagliflozin groups showed a placebo-corrected significant increase in high-density lipoprotein-cholesterol (HDL-C) levels (2.3%–10.7%),Citation50,Citation51 with triglycerides showing a minimal decrease (0.6%–12.8%),Citation50,Citation51 and LDL-C showed an increase (1.9%–10.9%) in most studies.Citation50Citation53 When canagliflozin 100 and 300 mg was compared to glimiperide, there was an increase in HDL-C of 7.9%, 9.0%, and 0.3%, respectively, while the triglycerides showed a decrease of 3.7% in the canagliflozin 100 mg group compared to an increase of 2.3% and 9.5% in the canagliflozin 300 mg and glimiperide groups, respectively.Citation53 Similarly, HDL-C increased in the pooled canagliflozin groups (7.6%) compared to sitagliptin (0.6%) while triglycerides showed an increase in both canagliflozin 300 mg (9.6%) and sitagliptin (11.9%).Citation54 An increase in LDL-C was also seen in both canagliflozin 300 mg (11.7%) and sitagliptin (5.2%) groups.Citation54

Adverse events

In Phase III studies, the incidence of adverse events (AEs) were similar between canagliflozin (50%–77%) versus placebo (53%–66%)Citation51,Citation52 versus glimiperide (68%) and sitagliptin (77.5%).Citation53,Citation54 Mild to moderate hypoglycemia episodes were higher in the pooled canagliflozin (0%–28%) versus placebo (2%),Citation50Citation52 while it was lower (4.6%–5.6%) when compared to glimiperide (34.2%)Citation53 and similar to sitagliptin.Citation55 There were no severe hypoglycemic events reported in most studies.Citation50,Citation52Citation54 Only small changes from baseline for serum creatinine, blood urea nitrogen, and serum urate were seen with canagliflozin relative to placebo.Citation50,Citation53 Small increases in hemoglobin were observed with canagliflozin (3.6% and 3.9%), whereas a slight decrease was observed with placebo (0.2%).Citation50 Increased urinary frequency was low in the pooled canagliflozin groups (2.4%–5.3%) versus placebo (0%–0.5%).Citation51Citation54 The incidence of serious AEs were similar in the pooled canagliflozin groups (2.1%–4.1%) versus placebo (1%–5.8%).Citation50Citation52 Two deaths occurred during the treatment period, one each in the placebo and canagliflozin 100 mg group, and were deemed not to be drug related.Citation50 The incidence of serious AEs when compared to glimiperide and sitagliptin were similar.Citation53,Citation54 There were no cardiovascular (CV) AEs reported in any study.Citation50Citation54 AEs secondary to reduced intravascular volume such as postural dizziness/orthostatic hypotension were low (≤3%).Citation50,Citation52

The incidence of overall AEs were the same in patients <65 and ≥65 years of age.Citation57 Serious AEs and AE-related discontinuations were higher in canagliflozin 100 mg in the ≥65 year-old patient group. The incidence of genital mycotic infections and osmotic diuresis-related AEs were higher with canagliflozin in both age groups. Incidences of urinary tract infections (UTIs), renal-related AEs, AEs related to volume depletion, lipid parameters, and documented hypoglycemia episodes were similar in both groups, indicating that it was generally well tolerated in older T2DM patients.Citation57

When T2DM patients on hydrochlorothiazide (HCTZ) were given canagliflozin, the change in 24-hour urine volume from baseline was −0.1 L with canagliflozin alone and 0.4 L with HCTZ alone and with canagliflozin + HCTZ.Citation60 Most AEs were mild and higher with canagliflozin + HCTZ (69%) than with canagliflozin (47%) or HCTZ (50%) alone. There were minimal changes in serum electrolytes with canagliflozin + HCTZ compared with individual treatments. Canagliflozin coadministered with HCTZ was generally well tolerated.Citation60 Similarly, 36 T2DM patents on angiotensin-converting enzyme inhibitors or angiotensin receptor blockers given canagliflozin showed a modest increase in urine volume initially which was attenuated at the end of the study.Citation61 Measures of volume status which were modestly increased at the beginning remained elevated till the end.Citation61

In pooled data from Phase III studies, the incidence of female and male genital mycotic infections in 2,313 and 9,439 T2DM patients exposed to canagliflozin for 24 and 68 weeks, respectively, were studied.Citation62 In the first group, the incidence of genital mycotic infections was higher in females compared to males for both canagliflozin 100 (10.4% and 4.2%) and canagliflozin 300 mg (11.4% and 3.7%) versus placebo (3.2% and 0.6%). In the second group, with 68 weeks exposure to canagliflozin, the incidence for canagliflozin 100 and 300 mg versus placebo was females: 14.7%, 13.9%, 3.1% versus males: 7.3%, 9.3%, 1.6%, respectively. Most were mild to moderate in intensity, occurred within the first 4 months in females and first year in males, none were serious, few led to discontinuation, few patients had >1 event (females, 2.3%; males, 0.9%), were not dose dependent, and responded to standard treatments.Citation62

Clinical profiling of canagliflozin

Based on the available data regarding the mechanism of canagliflozin, its efficacy, and safety, the clinician may attempt to find the ideal T2DM patient for whom it may lead to certain benefits. As it acts independent of β-cell function or mass and insulin sensitivity, canagliflozin can be used at any stage of T2DM, any age, irrespective of the duration of diabetes and in combination with all other oral agents and insulin. Ideally, patients who need an A1c lowering from 0.5% to 1.17% may benefit from its use.Citation50Citation54 It may be used as a second line agent in obese T2DM patients who need weight loss with fat mass reductions. In addition, the use of canagliflozin in patients on oral SU and/or insulin therapy may help to attenuate the weight gain associated with these agents. Additional benefits seen are BP reductions due to a diuretic effect, which appear to be safe, and may allow more T2DM patients to reach the goal BP with or without reduction in dose of antihypertensive medications.

It is unclear at this point if there is a clinical implication of increase in LDL-C levels associated with canagliflozin therapy on CV risk. At this time, it would be prudent to keep a close watch on the LDL-C levels and adjust statin therapy accordingly to maintain LDL goals and await the results of long-term CV studies like the Canagliflozin cardioVascular Assessment Study.

It may thus be appropriate at the present time to be cautious about starting canagliflozin therapy in patients with a heavy preexisting CV burden. Even though UTIs and genital infections were mild to moderate in nature and the same in the <65 and >65 year age groups, it may be prudent to avoid using it in patients with preexisting repeated episodes of candidiasis and/or UTIs, and the consequences of treatment of these AEs may discourage the use of canagliflozin in the elderly patient. Increased urinary frequency in the less mobile elderly may lead to discomfort.

Discussion

The importance of genetic variants was shown in the monogenic forms of diabetes. The clinical implication of finding these single gene diabetes disorders is that the physicians can be certain that oral antihyperglycemic agents rather that insulin therapy can be used successfully to treat such patients. However, as monogenic diabetes is found only in a small percentage of patients, a similar discovery of genetic variants, which can guide and individualize diabetes therapy in the more common T2DM that is multifactorial in origin, would be clinically significant. Genome-wide association studies have opened the door for patient profiling and research into genetic variants in multifactorial T2DM.

Another push for individualized management is that drug costs account for a large portion of the health expenditure in the US. The response to drugs is not always the same even in T2DM patients with similar phenotypes. At present, most physicians follow an algorithmic approach to T2DM treatment. A prior knowledge of the drug response in a particular patient based on pharmacogenomics can lead to a more efficient use of the appropriate drugs. It would guide physicians in providing patients with the most effective treatment strategy given their individual genetic background and avoid drug side effects in the process.

It appeared that oral antihyperglycemic therapy was more effective in the presence of certain genotypes. Clinically, it may be possible to tailor the type of medication used based on the presence or absence of the various genetic variants and thus identify patients who will have the highest benefit from these agents. It appears that most of the information on genetic variants and response to meglitinides and thiazolidinedione therapy comes from the Chinese and Japanese populations, which raises the question about its applicability to other ethnic groups.

A number of the genes are involved in drug metabolism in T2DM patients, and some of the information available in the literature needs further validation to ensure its authenticity. There are several studies whose findings have not been replicated and, at present, cannot confirm association. The polymorphisms studied may only explain some of the variability in response to T2DM drug therapy. There are still unidentified factors including racial differences which appear to play a role in the interindividual variability seen in clinical practice. For example, the SNP CYP2C8*3 was found in higher frequency in Caucasians (10%–23%),Citation63 followed by Hispanics, and is rare among people with African and Asian ancestry.Citation63,Citation64 The present studies have been done in certain ethnic groups, and further well-designed studies are needed to cover other ethnic groups found in the US and the rest of the world. Further studies to detect other genetic factors which may better explain the variability in drug treatment response is needed.

Advances in T2DM genetics may in the future improve disease prediction, prevention, and treatment. A barrier to the implementation of routine genetic testing may be the acceptance by physicians and patients. A recent survey of a sample of 304 nationally representative US primary care physicians and endocrinologists, 152 nondiabetic primary care patients, and 89 patients enrolled in a diabetes pharmacogenetics study was done to assess their reactions to genetic testing for T2DM.Citation65 Both physicians (79%) and patients (80%) favored genetic testing for diabetes risk prediction. About 71% of patients versus 23% of physicians indicated that a “high risk” result would be very likely to improve motivation to adopt preventive lifestyle changes. In addition, more patients favored genetic testing to guide therapy (78%) versus physicians (48%) and reported that genetic testing would make them “much more motivated” to adhere to medications (72%). Both physicians and patients have high expectations that genetic testing would improve patient motivation to control T2DM and can possibly motivate behavior change compared with standard risk information.Citation65 Regarding canagliflozin, until we have well-performed genetic studies, the decision to use canagliflozin or any other SGLT2 inhibitor should be individualized based on the health and presence of comorbid conditions on a case by case basis, especially in the older age patient groups. Adequate education should be provided to all patients prior to starting canagliflozin.

Conclusion

The potential exists for pharmacogenomics to promote efficacious, safe, and cost-effective, individualized diabetes management. However, pharmacogenomics is still in its early stages, and the idea of defining patients genetically to predict individual responses to drugs and obtain safe and effective T2DM management is promising in spite of existing barriers. Currently, clinical profiling of patients with T2DM and using an individualized approach with most drugs, including canagliflozin, based on comorbid conditions still remains the most accepted approach for the management of T2DM.

Disclosure

The author reports no conflicts of interest in this work.

References

  • AliMKMcKeever BullardKImperatoreGBarkerLGreggEWCenters for Disease Control and Prevention (CDC)Characteristics associated with poor glycemic control among adults with self-reported diagnosed diabetes – National Health and Nutrition Examination Survey, United States, 2007–2010MMWR Morb Mortal Wkly RepSuppl61323722695461
  • InzucchiSEBergenstalRMBuseJBAmerican Diabetes Association (ADA)European Association for the Study of Diabetes (EASD)Management of hyperglycemia in type 2 diabetes: a patient-centered approach: position statement of the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD)Diabetes Care20123561364137922517736
  • InzucchiSEOral antihyperglycemic therapy for type 2 diabetes: scientific reviewJAMA2002287336037211790216
  • DonnellyLADoneyASHattersleyATMorrisADPearsonERThe effect of obesity on glycaemic response to metformin or sulphonylureas in Type 2 diabetesDiabet Med200623212813316433709
  • GrahamGGPuntJAroraMClinical pharmacokinetics of metforminClin Pharmacokinet2011502819821241070
  • OtsukaMMatsumotoTMorimotoRAriokaSOmoteHMoriyamaYA human transporter protein that mediates the final excretion step for toxic organic cationsProc Natl Acad Sci U S A200510250179231792816330770
  • BeckerMLVisserLEvan SchaikRHHofmanAUitterlindenAGStrickerBHGenetic variation in the organic cation transporter 1 is associated with metformin response in patients with diabetes mellitusPharmacogenomics J20099424224719381165
  • TaniharaYMasudaSSatoTKatsuraTOgawaOInuiKSubstrate specificity of MATE1 and MATE2-K, human multidrug and toxin extrusions/H(+)-organic cation antiportersBiochem Pharmacol200774235937117509534
  • ShikataEYamamotoRTakaneHHuman organic cation transporter (OCT1 and OCT2) gene polymorphisms and therapeutic effects of metforminJ Hum Genet200752211712217111267
  • ZhouKBellenguezCSpencerCCGoDARTS and UKPDS Diabetes Pharmacogenetics Study GroupWellcome Trust Case Control Consortium 2MAGIC investigatorsCommon variants near ATM are associated with glycemic response to metformin in type 2 diabetesNat Genet201143211712021186350
  • ChristensenMMBrasch-AndersenCGreenHThe pharmacogenetics of metformin and its impact on plasma metformin steady-state levels and glycosylated hemoglobin A1cPharmacogenet Genomics2011211283785021989078
  • LavrenkoAVShlykovaOAKutsenkoLAMamontovaTVKaĭdashevIPPharmacogenetic features of the effect of metformin in patients with coronary heart disease in the presence of metabolic syndrome and type 2 diabetes mellitus in terms of PPAR-gamma2 gene polymorphismTer Arkh20128493540 Russian23091851
  • PearsonERLiddellWGShepherdMCorrallRJHattersleyATSensitivity to sulphonylureas in patients with hepatocyte nuclear factor-1alpha gene mutations: evidence for pharmacogenetics in diabetesDiabet Med200017754354510972586
  • JavorskyMKlimcakovaLSchronerZKCNJ11 gene E23K variant and therapeutic response to sulfonylureasEur J Intern Med201223324524922385882
  • SestiGLarattaECardelliniMThe E23K variant of KCNJ11 encoding the pancreatic beta-cell adenosine 5′-triphosphate-sensitive potassium channel subunit Kir6.2 is associated with an increased risk of secondary failure to sulfonylurea in patients with type 2 diabetesJ Clin Endocrinol Metab20069162334233916595597
  • SestiGMariniMACardelliniMThe Arg972 variant in insulin receptor substrate-1 is associated with an increased risk of secondary failure to sulfonylurea in patients with type 2 diabetesDiabetes Care20042761394139815161794
  • LangVYFatehiMLightPEPharmacogenomic analysis of ATP-sensitive potassium channels coexpressing the common type 2 diabetes risk variants E23K and S1369APharmacogenet Genomics201222320621422209866
  • SchronerZDobrikovaMKlimcakovaLVariation in KCNQ1 is associated with therapeutic response to sulphonylureasMed Sci Monit2011177CR392CR39621709633
  • DaiXPHuangQYinJYKCNQ1 gene polymorphisms are associated with the therapeutic efficacy of repaglinide in Chinese type 2 diabetic patientsClin Exp Pharmacol Physiol201239546246822414228
  • YuWHuCZhangREffects of KCNQ1 polymorphisms on the therapeutic efficacy of oral antidiabetic drugs in Chinese patients with type 2 diabetesClin Pharmacol Ther201189343744221289621
  • GongZCHuangQDaiXPNeuroD1 A45T and PAX4 R121W polymorphisms are associated with plasma glucose level of repaglinide monotherapy in Chinese patients with type 2 diabetesBr J Clin Pharmacol201274350150922296034
  • HuangQYinJYDaiXPAssociation analysis of SLC30A8 rs13266634 and rs16889462 polymorphisms with type 2 diabetes mellitus and repaglinide response in Chinese patientsEur J Clin Pharmacol201066121207121520809084
  • KangESParkSYKimHJEffects of Pro12Ala polymorphism of peroxisome proliferator-activated receptor gamma2 gene on rosiglitazone response in type 2 diabetesClin Pharmacol Ther200578220220816084854
  • HsiehMCLinKDTienKJCommon polymorphisms of the peroxisome proliferator-activated receptor-gamma (Pro12Ala) and peroxisome proliferator-activated receptor-gamma coactivator-1 (Gly482Ser) and the response to pioglitazone in Chinese patients with type 2 diabetes mellitusMetabolism20105981139114420045142
  • BlüherMLübbenGPaschkeRAnalysis of the relationship between the Pro12Ala variant in the PPAR-gamma2 gene and the response rate to therapy with pioglitazone in patients with type 2 diabetesDiabetes Care200326382583112610044
  • NamvaranFAzarpiraNRahimi-MoghaddamPDabbaghmaneshMHPolymorphism of peroxisome proliferator-activated receptor γ (PPARγ) Pro12Ala in the Iranian population: relation with insulin resistance and response to treatment with pioglitazone in type 2 diabetesEur J Pharmacol20116711–31621968139
  • Kawaguchi-SuzukiMFryeRFCurrent clinical evidence on pioglitazone pharmacogenomicsFront Pharmacol2013414724324437
  • PeiQHuangQYangGPPPAR-γ2 and PTPRD gene polymorphisms influence type 2 diabetes patients’ response to pioglitazone in ChinaActa Pharmacol Sin201334225526123147557
  • ZhangKHHuangQDaiXPEffects of the peroxisome proliferator activated receptor-γ coactivator-1α (PGC-1α) Thr394Thr and Gly482Ser polymorphisms on rosiglitazone response in Chinese patients with type 2 diabetes mellitusJ Clin Pharmacol20105091022103020498286
  • LiuHLLinYGWuJImpact of genetic polymorphisms of leptin and TNF-alpha on rosiglitazone response in Chinese patients with type 2 diabetesEur J Clin Pharmacol200864766367118438653
  • MakinoHShimizuIMuraoSA pilot study suggests that the G/G genotype of resistin single nucleotide polymorphism at −420 may be an independent predictor of a reduction in fasting plasma glucose and insulin resistance by pioglitazone in type 2 diabetesEndocr J20095691049105819738363
  • SunHGongZCYinJYThe association of adiponectin allele 45T/G and −11377C/G polymorphisms with Type 2 diabetes and rosiglitazone response in Chinese patientsBr J Clin Pharmacol200865691792618429970
  • LiZPengXWuYXiaYLiuXZhangQThe influence of adiponectin gene polymorphism on the pioglitazone response in the Chinese with type 2 diabetesDiabetes Obes Metab200810979480218494805
  • NamvaranFRahimi-MoghaddamPAzarpiraNDabbaghmaneshMHPolymorphism of adiponectin (45T/G) and adiponectin receptor-2 (795G/A) in an Iranian population: relation with insulin resistance and response to treatment with pioglitazone in patients with type 2 diabetes mellitusMol Biol Rep20123955511551822187345
  • YangMHuangQWuJEffects of UCP2 -866 G/A and ADRB3 Trp64Arg on rosiglitazone response in Chinese patients with Type 2 diabetesBr J Clin Pharmacol2009681142219659999
  • WangGWangXZhangQMaZResponse to pioglitazone treatment is associated with the lipoprotein lipase S447X variant in subjects with type 2 diabetes mellitusInt J Clin Pract200761455255717394430
  • SathananthanAManCDMichelettoFCommon genetic variation in GLP1R and insulin secretion in response to exogenous GLP-1 in nondiabetic subjects: a pilot studyDiabetes Care20103392074207620805279
  • MüssigKStaigerHMachicaoFAssociation of type 2 diabetes candidate polymorphisms in KCNQ1 with incretin and insulin secretionDiabetes20095871715172019366866
  • ShuLMatveyenkoAVKerr-ConteJChoJHMcIntoshCHMaedlerKDecreased TCF7L2 protein levels in type 2 diabetes mellitus correlate with downregulation of GIP- and GLP-1 receptors and impaired beta-cell functionHum Mol Genet200918132388239919386626
  • SchäferSATschritterOMachicaoFImpaired glucagon-like peptide-1-induced insulin secretion in carriers of transcription factor 7-like 2 (TCF7L2) gene polymorphismsDiabetologia200750122443245017661009
  • VillarealDTRobertsonHBellGITCF7L2 variant rs7903146 affects the risk of type 2 diabetes by modulating incretin actionDiabetes201059247948519934000
  • SchäferSAMüssigKStaigerHA common genetic variant in WFS1 determines impaired glucagon-like peptide-1-induced insulin secretionDiabetologia20095261075108219330314
  • Simonis-BikAMNijpelsGvan HaeftenTWGene variants in the novel type 2 diabetes loci CDC123/CAMK1D, THADA, ADAMTS9, BCL11A, and MTNR1B affect different aspects of pancreatic beta-cell functionDiabetes201059129330119833888
  • AquilanteCLWempeMFSidhomMSKosmiskiLAPredhommeJAEffect of ABCB1 polymorphisms and atorvastatin on sitagliptin pharmacokinetics in healthy volunteersEur J Clin Pharmacol20136971401140923407853
  • KwonOChoeEYChoiYDiscovery of dipeptidyl peptidase-4 gene variants and the associations with efficacy of vildagliptin in patients with type 2 diabetes – a pilot studyJ Diabetes Metab2013S13006
  • WellsRGMohandasTKHedigerMALocalization of the Na+/glucose cotransporter gene SGLT2 to human chromosome 16 close to the centromereGenomics19931737877898244402
  • YuLLvJCZhouXJZhuLHouPZhangHAbnormal expression and dysfunction of novel SGLT2 mutations identified in familial renal glucosuria patientsHum Genet2011129333534421165652
  • LeeYWClinical and genetic analysis in a patient with primary renal glucosuria: Identification of a novel mutation in the SLC5A2 geneExp Ther Med2013661532153424255686
  • EnigkUBreitfeldJSchleinitzDRole of genetic variation in the human sodium-glucose cotransporter 2 gene (SGLT2) in glucose homeostasisPharmacogenomics20111281119112621830867
  • StenlöfKCefaluWTKimKAEfficacy and safety of canagliflozin monotherapy in subjects with type 2 diabetes mellitus inadequately controlled with diet and exerciseDiabetes Obes Metab201315437238223279307
  • WildingJPMathieuCVercruysseFUsiskinKDengLCanovatchelWCanagliflozin (CANA), a sodium glucose co-transporter 2 inhibitor, improves glycemic control and reduces body weight in subjects with type 2 diabetes (T2D) inadequately controlled with metformin (MET) and sulfonylurea (SU)Poster presented at: American Diabetes Association 72nd Annual Scientific SessionsJune 8–12, 2012Philadelphia, PA Abstract 1022P
  • ForstTGuthrieRGoldenbergREfficacy and safety of canagliflozin in subjects with type 2 diabetes on metformin and pioglitazonePoster presented at: 4th World Congress on Controversies to Consensus in Diabetes, Obesity and Hypertension (CODHy)November 8–11, 2012Barcelona, Spain Board 64
  • CefaluWTLeiterLAYoonKHEfficacy and safety of canagliflozin versus glimepiride in patients with type 2 diabetes inadequately controlled with metformin (CANTATA-SU): 52 week results from a randomised, double-blind, phase 3 non-inferiority trialLancet2013382989694195023850055
  • GrossJLSchernthanerGFuMEfficacy and safety of canagliflozin, a sodium glucose co-transporter 2 inhibitor, compared with sitagliptin in patients with type 2 diabetes on metformin plus sulfonylureaAbstract presented at: American Diabetes Association 72nd Annual Scientific SessionsJune 8–12, 2012Philadelphia, PA Abstract 50
  • Lavalle-GonzálezFJJanuszewiczADavidsonJEfficacy and safety of canagliflozin compared with placebo and sitagliptin in patients with type 2 diabetes on background metformin monotherapy: a randomised trialDiabetologia201356122582259224026211
  • ToubroSCefaluWTXieJCanagliflozin reduces body weight mainly through loss of fat mass in subjects with type 2 diabetesPoster presented at: 4th World Congress on Controversies to Consensus in Diabetes, Obesity and Hypertension (CODHy)November 8–11, 2012Barcelona, Spain Board 72
  • SinclairABodeBHarrisSEfficacy and safety of canagliflozin compared with placebo in older patients with type 2 diabetes mellitus: a pooled analysis of clinical studiesBMC Endocr Disord2014143724742013
  • ElmoreLKBaggettSKyleJASkelleyJWA review of the efficacy and safety of canagliflozin in elderly patients with type 2 diabetesConsult Pharm201429533534624849690
  • PolidoriDMariAFerranniniECanagliflozin, a sodium glucose co-transporter 2 inhibitor, improves model-based indices of beta cell function in patients with type 2 diabetesDiabetologia201457589190124585202
  • DevineniDVaccaroNPolidoriDRuschSWajsEEffects of hydrochlorothiazide on the pharmacokinetics, pharmacodynamics, and tolerability of canagliflozin, a sodium glucose co-transporter 2 inhibitor, in healthy participantsClin Ther201436569871024726680
  • ShaSPolidoriDHeiseTEffect of the sodium glucose co-transporter 2 inhibitor, canagliflozin, on plasma volume in patients with type 2 diabetes mellitusDiabetes Obes Metab2014161087109524939043
  • NyirjesyPSobelJDFungAGenital mycotic infections with canagliflozin, a sodium glucose co-transporter 2 inhibitor, in patients with type 2 diabetes mellitus: a pooled analysis of clinical studiesCurr Med Res Opin20143061109111924517339
  • AquilanteCLWempeMFSpencerSHKosmiskiLAPredhommeJASidhomMSInfluence of CYP2C8*2 on the pharmacokinetics of pioglitazone in healthy African-American volunteersPharmacotherapy20133391000100723712614
  • MartisSPeterIHulotJSKornreichRDesnickRJScottSAMulti-ethnic distribution of clinically relevant CYP2C genotypes and haplotypesPharmacogenomics J201313436937722491019
  • GrantRWHivertMPandiscioJCFlorezJCNathanDMMeigsJBThe clinical application of genetic testing in type 2 diabetes: a patient and physician surveyDiabetologia200952112299230519727660