0006F PDF
0006F PDF
0006F PDF
Antidiabetic Agents in Patients with Chronic Kidney Disease and End-Stage Renal
Disease on Dialysis: Metabolism and Clinical Practice
1
Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine,
Tokyo, Japan; 2Division of General Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
Abstract: Numerous drugs with different mechanisms of action and different pharmacologic profiles are being used with the aim of im-
proving glycemic control in patients with type 2 diabetes. Therapeutic options for patients with type 2 diabetes and chronic kidney dis-
ease (CKD) are limited because a reduced glomerular filtration rate results in the accumulation of certain drugs and/or their metabolites.
Conventional oral hypoglycemic agents, such as sulfonylurea (SU), are not suitable due to the risk of prolonged hypoglycemia; further-
more, metformin is contraindicated for moderate to advanced CKD. Therefore, in order to achieve good glycemic control, insulin injec-
tion therapy remains the mainstay of treatment in diabetic patients with moderate to advanced CKD, particularly in those receiving dialy-
sis therapy. However, some agents have been used even in patients with CKD. Repaglinide and mitiglinide are rapid- and short-acting in-
sulinotropic SU receptor ligands. They are rarely accompanied by hypoglycemia, and are attractive therapeutic options even in the dialy-
sis population. In addition, alpha-glucosidase inhibitors are rarely accompanied by hypoglycemia and are administered without dose ad-
justments in dialysis patients. However, the National Kidney Foundation Kidney Disease Outcomes Quality Initiative guidelines recom-
mended that alpha-glucosidase inhibitors should be avoided in patients with advanced stage CKD and on dialysis. Furthermore,
mitiglinide is not currently used in the US. Thus, recommended oral antidiabetic agents differ between countries. Moreover, dipeptidyl
peptidase-4 inhibitors and incretin mimetics are new antihyperglycemic agents, which may be used more frequently in the future in pa-
tients with type 2 diabetes and CKD. Here, we describe the pharmacokinetics, metabolism, clinical efficacy, and safety of oral antidia-
betic agents for patients with CKD, including those receiving dialysis.
Keywords: Antidiabetic agent, chronic kidney disease, clinical practice, DPP-4 inhibitor, end-stage renal disease, metabolism, type 2 diabe-
tes mellitus.
INTRODUCTION agents, such as sulfonylurea (SU), are not suitable due to the risk of
The number of patients with primary diabetic nephropathy un- prolonged hypoglycemia; furthermore, metformin is contraindi-
dergoing dialysis continues to increase. The prognosis is worse for cated. Therefore, in order to achieve good glycemic control, insulin
diabetic patients undergoing dialysis than for patients with primary injection therapy remains the mainstay of treatment in diabetic pa-
glomerular nephropathy, and diabetic patients with chronic kidney tients with moderate to advanced CKD, particularly in those receiv-
disease (CKD) who are undergoing dialysis also have an increased ing dialysis therapy. However, some agents have been used in pa-
risk of cardiovascular morbidity and a complicated disease course. tients with CKD and were found to be effective and safe even in
Recent clinical evidence has suggested that strict glycemic control those on dialysis. Therefore, some agents may be useful therapeutic
reduces the risk of cardiovascular disease, which is the main cause options for the management of diabetes.
of death in diabetic patients [1, 2]. Thus, good glycemic control can Here we review the effect and metabolism of antidiabetic
decrease cardiovascular and other diabetes-related causes of death, agents in kidney disease, with a focus primarily on reports pub-
leading to better survival rates in diabetic patients [3-5]. Diabetic lished since 1990. An electronic search of the MEDLINE biblio-
patients on long-term dialysis therapy, however, no longer need to graphic database was performed through PubMed
achieve good glycemic control to prevent deterioration of kidney (www.ncbi.nlm.nih.gov/sites/entrez) by using relevant key search
function. Nevertheless, good control may still prevent or slow the terms including pharmacokinetics, diabetes mellitus, renal failure,
progression of retinopathy, neuropathy, and possibly macrovascular renal impairment, kidney disease, drug metabolism, and antidia-
disease [6]. Survival improves with better glycemic control in pa- betic drug. Reference lists of original studies, narrative reviews, and
tients on peritoneal dialysis [7] and hemodialysis therapy [8]. After previous systematic reviews were also examined. Finally, select
adjustment for age and sex, hemoglobin A1c was found to be a information from drug manufacturers was also reviewed. Here, we
significant predictor of survival (hazard ratio, 1.133 per 1.0% in- describe the pharmacokinetics, metabolism, clinical efficacy, and
crease in HbA1c; 95% confidence interval, 1.028-1.249; P = 0.012) safety of oral antidiabetic agents for diabetic patients with CKD,
[8]. including those on dialysis.
Numerous drugs with different mechanisms of action and dif-
Classification of Antidiabetic Agents and their Pharmacother-
ferent pharmacologic profiles are being used with the aim of im-
apy
proving glycemic control in patients with type 2 diabetes. Thera-
peutic options for patients with diabetes with CKD and end-stage As shown in Table 1, several pharmacologic agents other than
renal disease (ESRD) are limited because a reduced glomerular insulin preparations are available for the management of type 2
filtration rate (GFR) results in the accumulation of certain drugs diabetes. These agents all lower blood glucose levels, but there is an
and/or their metabolites [9]. Conventional oral hypoglycemic increasing awareness that they may modify the natural history of
diabetes in different ways. Insulin secretagogues can be classified
as sulphonylureas (SUs) and meglitinides. Alpha-glucosidase in-
*Address correspondence to this author at the Division of Nephrology, hibitors are modifiers of glucose absorption, and thiazolidinediones
Hypertension and Endocrinology, Department of Internal Medicine, Nihon are insulin sensitizers. Incretin-related therapies include dipeptidyl
University School of Medicine, 30-1, Oyaguchi Kami-chou, Itabashi-ku, peptidase-4 (DPP-4) inhibitors and incretin mimetics. DPP-4 inhibi-
Tokyo 173-8610, Japan; Tel: +81-3-3972-8111; Fax: +81-3-3972-1098; tors are oral antidiabetic agents, whereas incretin mimetics are used
E-mail: abe.masanori@nihon-u.ac.jp
by subcutaneous injection.
Table 1. Dosing Adjustment by CKD Stage for Drugs Used to Treat Hyperglycemia
Dosing Recommendation
Dosing Recommendation
Class Drug CKD Stage 3,4, or
Dialysis
Kidney Transplant
Characteristics of the Pharmacokinetics in Chronic Kidney Dis- nant route of elimination, the pharmacokinetics and/or pharma-
ease codynamics may be altered in patients with renal impairment.
Many drugs bind to serum proteins, primarily albumin. How- Therefore, caution will be needed to avoid hypoglycemia when
ever, as the plasma concentration of albumin in patients with renal using SU agents in patients with advanced stage CKD and those on
impairment is commonly decreased, the concentrations of unbound dialysis. The main clinically relevant pharmacokinetics of glipizide,
drugs are increased. For drugs with a low hepatic extraction rate gliclazide, and glimepiride are summarized in Table 2.
and high protein binding rate, the overall rate of drug elimination is
Meglitinides
related to unbound plasma concentration and intrinsic clearance of
the liver. Assuming that the intrinsic capacity of the liver remains Three insulin secretagogues, nateglinide, repaglinide, and
unchanged, total clearance will increase and half-life will tend to mitiglinide, are currently in clinical use because of their rapid onset
decrease for drugs such as phenytoin and warfarin. Furthermore, of action resulting in improvement in hyperglycemia. The main
kidney disease differentially affects uptake and efflux transporters differences between these three meglitinides, which are of potential
and metabolic enzymes in the liver and gastrointestinal tract, interest for use in the management of patients with type 2 diabetes,
changes likely to be caused by uremic toxins. A change in systemic are summarized in Table 3. After oral administration, repaglinide is
bioavailability might be caused by reduced gastrointestinal absorp- almost completely metabolized in the liver by CYP enzymes, prin-
tion, which has been observed in patients with severe renal impair- cipally CYP2C8 (70%) and CYP3A4 (30%) [22, 23]. Repaglinide
ment for cyclosporine, ibuprofen, and levocabastine [10-12]. In undergoes oxidative biotransformation by CYP3A4 to metabolites
addition, it was reported that reductions in the expression and func- M2 (dicarboxylic acid) and M1 (aromatic amine) or direct conjuga-
tion of intestinal P-glycoprotein (P-gp) and multidrug resistance- tion with glucuronic acid to form M7 (acyl glucuronide), all of
associated protein-2 increased expression of hepatic P-gp and which are inactive metabolites. Repaglinide undergoes hepatic
multidrug resistance-associated protein-2 and decreased expression OATP1B1-mediated uptake, followed by CYP2C8- and CYP3A4-
of hepatic organic anion-transporting polypeptide (OATP) in ure- mediated metabolism, and then biliary excretion which is likely
mia [13, 14]. Numerous reports of known OATP, P-gp, and/or cy- mediated, at least partially, by P-gp, with <8% of the parent drug
tochrome P450 (CYP) substrates exhibiting altered pharmacokinet- being excreted unchanged in the urine [24]. The drug exhibits a
ics in patients with kidney disease suggest that uremia affects these nearly 4-fold increase in half-life after 1 week of treatment, and
elimination pathways and illustrates the clinical importance of al- significantly increases the area under the curves (AUCs) after both
tered nonrenal clearance [15, 16]. Therefore, caution is required single and multiple dosing in patients with advanced CKD (creatin-
since uremia differentially affects these pathways in the liver and ine clearance (CrCl) <30 mL/min) when compared to subjects with
gastrointestinal tract and leads to a decreased protein binding rate. normal renal function (CrCl >80 mL/min) [24]. No difference in
maximal plasma concentrations was observed, thereby suggesting
Sulphonylureas that bioavailability of the orally administered drug is not affected
Insulin secretagogues increase endogenous insulin levels. These by kidney disease and that metabolism and transport in the gastroin-
agents work by binding to SU receptors or nearby sites, resulting in testinal tract are therefore unchanged [25]. These data suggest that
closure of ATP-sensitive potassium channels of the pancreatic beta hepatic clearance of repaglinide, mediated by OATP1B1, CYP2C8,
cell, depolarization of the cell membrane, calcium influx, and sub- CYP3A4, and/or possibly P-gp, is decreased in subjects with ad-
sequently insulin release. The University Diabetes Group Project vanced CKD [25]. Thus, uremia leads to decreased activity of he-
raised concerns that first-generation secretagogues may be associ- patic and intestinal CYPs, including CYP3A, secondary to reduced
ated with increased cardiovascular risk and mortality [17]. The protein and gene expression.
United Kingdom Prospective Diabetes Study (UKPDS) [18] and In vitro data have demonstrated that nateglinide is predomi-
more recently the ADVANCE study [19] demonstrated that newer nantly metabolized by CYP isoenzymes CYP2C9 (70%) and
agents (glipizide [18] and gliclazide [19]) do not appear to confer CYP3A4 (30%) [24]. However, the metabolites of nateglinide have
such a risk and are associated with benefits when used to achieve insulin secretion properties. Therefore, the half-life of this drug is
tighter glycemic control. As these agents increase endogenous insu- prolonged in patients with renal impairment and this can result in
lin levels, they are associated with an increased risk of hypoglyce- hypoglycemic episodes. On the other hand, the metabolism of
mia. This risk is mitigated when shorter-acting agents are used. mitiglinide differs from that of repaglinide and nateglinide [26].
However, many of the first-generation SU agents (i.e., acetohex- Although mitiglinide is metabolized in the kidney and liver, and
amide, chlorpropamide, tolazamide, and tolbutamide) are contrain- predominantly metabolized to the glucuronic acid conjugate (74%)
dicated in the dialysis population. The second-generation SU and hydroxide (<25%), these metabolites have little insulin secre-
glimepiride is contraindicated in dialysis patients, but low-dose tory activity [27]. Mitiglinide selectively inhibits the ATP-
initiation can be used in patients with CKD [20]. Glipizide and dependent K+ channel (Kir6.2/SUR1) of pancreatic beta cells with a
gliclazide are the preferred agents and no dose adjustment has been higher affinity for Kir6.2/SUR1 than for the other two insuli-
necessary in a dialysis population in US [6]. Glipizide is eliminated notropic SU receptor ligands (mitiglinide > repaglinide > nateg-
primarily by hepatic biotransformation; <10% of a dose is excreted linide) [28]. These results suggest that mitiglinide acts specifically
as unchanged drug in urine and feces, while approximately 90% is on pancreatic beta cells to induce secretion of insulin and has few
excreted as biotransformation products in urine (80%) and feces unwanted effects on the cardiovascular system. As mitiglinide is
(10%) [21]. The major metabolites of glipizide are products of aro- rarely accompanied by hypoglycemia, it is an attractive therapeutic
matic hydroxylation that have no hypoglycemic activity. A minor option even in the dialysis population in Japan, although it is not
metabolite which accounts for <2% of a dose, an acetylamino-ethyl available in US. Therefore, it is considered that the likelihood of
benzene derivative, is reported to have one-tenth to one-third of the hypoglycemia is low with mitiglinide, even in patients with renal
hypoglycemic activity compared to the parent compound. Glipizide impairment, compared with nateglinide. The time to maximum
is 98-99% bound to serum proteins, primarily to albumin. The mean concentration (Tmax) and the half-life of mitiglinide in patients re-
terminal elimination half-life of glipizide ranges from 2 to 5 h after ceiving dialysis are approximately 0.41 and 11.7 h, respectively.
single or multiple doses in patients with type 2 diabetes [21]. There Because the half-life of mitiglinide is 1.48 and 3.22 h in patients
is only limited information regarding the effects of renal impair- with normal renal function and stage 3 CKD, respectively, care
ment on the disposition of glipizide. However, as glipizide is highly must be taken to avoid excessive blood glucose reduction when
protein bound and hepatic biotransformation is the predomi- mitiglinide is given to patients with renal impairment.
60 Current Drug Metabolism, 2011, Vol. 12, No. 1 Abe et al.
Table 2. Main Clinically Relevant Pharmacokinetic Differences of Glipizide, Gliclazide, and Glimepiride
Table 3. Main Clinically Relevant Pharmacokinetic Differences between the Three Meglitinides
Administration With each meal preprandially With each meal preprandially With each meal preprandially
In our previous study, there were a few cases of hypoglycemia also effectively improved fasting plasma glucose in dialysis pa-
even though the dose of mitiglinide was low compared with other tients.
clinical studies in patients with normal renal function [29-31]. The
daily dose of mitiglinide (23 mg) was adequate, as evidenced by the Alpha-Glucosidase Inhibitors
fact that it was able to induce significant reductions in glycemic The enzyme alpha-glucosidase is located in the gut and hydro-
parameters such as fasting plasma glucose, hemoglobin A1c, gly- lyzes oligosaccharides, trisaccharides, and disaccharides into
cated albumin, and homeostasis model assessment for insulin resis- glucose in the brush border of the small intestine. The antihyper-
tance (HOMA-IR) levels [32]. This suggests that appropriate blood glycemic action of alpha-glucosidase inhibitors results from the
glucose levels can be maintained even at a low dose of mitiglinide, reversible inhibition of membrane-bound intestinal alpha-glucoside
not only during the postprandial period but also before meals, due hydrolase enzymes. Alpha-glucosidase inhibitors decrease the rate
to the prolonged half-life of mitiglinide in patients on dialysis com- of breakdown of complex carbohydrates so that less glucose is ab-
pared with the half-life in those with normal renal function. We sorbed and postprandial hyperglycemia is lowered. In contrast to
noted that mitiglinide significantly improved glycemic control, SUs, alpha-glucosidase inhibitors do not enhance insulin secretion.
triglyceride levels, and interdialytic weight gain even when admin- The side effects are mainly gastrointestinal and include flatulence
istered for only a short duration [33]. Thus, we considered that and diarrhea. An example of an alpha-glucosidase inhibitor is
mitiglinide not only improved hemoglobin A1c and glycated albu- acarbose, which has been shown to reduce hyperglycemia and has
min, the overall index of glycemic control in type 2 diabetes, but also been found to have beneficial effects by reducing the risk of
Antidiabetic Agents in CKD and ESRD Current Drug Metabolism, 2011, Vol. 12, No. 1 61
cardiovascular disease and slowing the progression to diabetes in glycemic control, it is not associated with weight gain [39]. The
patients with hyperglycemia and normal renal function [34, 35]. In UKPDS demonstrated that when used as monotherapy in newly
addition, alpha-glucosidase inhibitors rarely induce hypoglycemia diagnosed, obese diabetic participants, the relative risk of myocar-
and are administered without dose adjustment in the dialysis popu- dial infarction was reduced by 30% [40]. The most common side
lation in Japan [36]. However, the National Kidney Foundation effect of metformin is gastrointestinal disturbance; however, it has
Kidney Disease Outcomes Quality Initiative (NKF-K/DOQI) guide- mild anorectic effects and has been shown to have beneficial car-
lines recommend that alpha-glucosidase inhibitors including acar- diovascular effects [41-43]. Metformin is absorbed via the small
bose and miglitol are to be avoided [6]. The main differences be- intestine and the absolute bioavailability is approximately 50-60%.
tween three alpha-glucosidase inhibitors, which are of potential Intravenous single-dose studies in normal subjects demonstrate that
interest for use in the management of patients with type 2 diabetes, metformin is excreted unchanged in the urine and does not undergo
are summarized in Table 4. hepatic metabolism (no metabolites have been identified in hu-
Acarbose is metabolized exclusively within the gastrointestinal mans) or biliary excretion [44]. Renal clearance of metformin is
tract, principally by intestinal bacteria, but also by digestive en- approximately 3.5-fold greater than creatinine clearance, which
zymes. Within 96 h of ingestion, 51% of an oral dose was excreted indicates that tubular secretion via human organic cation transporter
in the faces as unabsorbed drug-related radioactivity [37]. Because 2 is the major route of metformin elimination [45]. Following oral
acarbose acts locally within the gastrointestinal tract, low systemic administration, approximately 90% of the absorbed drug is elimi-
bioavailability of the parent compound is therapeutically desirable. nated via the renal route within the first 24 h, with a plasma elimi-
A fraction of these metabolites (approximately 34% of the dose) nation half-life of approximately 6.2 h. In blood, the elimination
was absorbed and subsequently excreted in the urine. The major half-life is approximately 17.6 h, suggesting that erythrocyte mass
metabolites have been identified as 4-methylpyrogallol derivatives may be a compartment of distribution. Single-dose and steady-state
(i.e., sulfate, methyl, and glucuronide conjugates). In addition, one pharmacokinetics of metformin were compared between patients
metabolite (formed by cleavage of a glucose molecule from acar- with normal renal function (CrCl >90 mL/min), mild impaired renal
bose) also has alpha-glucosidase inhibitory activity [37]. This me- function (CrCl 61-90 mL/min), moderate impaired renal function
tabolite, together with the parent compound, recovered from the (CrCl 31-60 mL/min), and severe impaired renal function (CrCl 10-
urine, accounts for <2% of the total administered dose. Although 30 mL/min). In patients with moderate to severe impaired renal
<2% of an oral dose of acarbose was absorbed as active drug, pa- function, C max and AUC were increased 173% and 390%, respec-
tients with severe renal impairment (CrCl <25 mL/min) attained tively, compared to the patients with normal renal function. In pa-
increases about 5-fold higher for peak plasma concentration of tients with decreased renal function, based on measured CrCl, the
acarbose and 6-fold higher for AUC values than subjects with nor- plasma half-life of metformin is prolonged and renal clearance is
mal renal function [37]. Because long-term clinical trials in diabetic decreased in proportion to the decrease in CrCl [45]. Therefore,
patients with significant renal dysfunction have not been conducted, metformin should be avoided in patients with moderate to severe
treatment of these patients with acarbose is not recommended [37]. CKD including those on dialysis since the risk of metformin accu-
mulation and lactic acidosis increases in line with the degree of
Miglitol, another alpha-glucosidase inhibitor, is not metabolized impairment of renal function. Furthermore, the fixed combination
in humans or in any other animal species studied thus far [38]. No of repaglinide and metformin (PrandiMetR , Novo Nordisk,
metabolites have been detected in plasma, urine, or feces, indicating Bagsvaerd, Denmark) should be avoided in patients with impaired
a lack of either systemic or presystemic metabolism. Miglitol is renal function. Although the plasma protein-binding capacity of
eliminated by renal excretion as unchanged drug [38]. Patients with metformin is low (1.1-2.8%), metformin is dialyzable with a clear-
CrCl <25 mL/min taking the miglitol 25 mg 3 times daily exhibited ance of up to 170 mL/min [46]. Therefore, hemodialysis may be
a greater than 2-fold increase in miglitol plasma levels when com- useful for the removal of accumulated drug from patients in whom
pared to subjects with CrCl >60 mL/min [38]. Dose adjustment to metformin overdose is suspected.
correct for the increased plasma concentrations is not feasible be-
cause miglitol acts locally. However, treatment of patients with Thiazolidinedione
CrCl <25 mL/min with miglitol is not recommended because the
Diabetes leads to ESRD and predisposes patients to a higher
safety of miglitol in these patients has not yet been elucidated [38].
cardiovascular risk and chronic inflammatory conditions via multi-
There has been speculation that alpha-glucosidase inhibitors are
factorial mechanisms, including insulin resistance [47, 48]. Insulin
more effective in Japanese diabetics, as the Japanese diet has his-
resistance, as assessed by HOMA-IR, is an independent predictor of
torically consisted of mainly carbohydrates, in particular rice. None
cardiovascular mortality in patients with ESRD [49]. Rosiglitazone
of the earlier studies, however, evaluated the efficacy of a alpha-
and pioglitazone are potent peroxisome proliferator-activated recep-
glucosidase inhibitor alone for patients with CKD and on dialysis.
tor gamma (PPAR) agonists belonging to a class of oral antidia-
Our group previously compared the effects of voglibose monother-
betic agents known as thiazolidinediones. PPAR agonists are insu-
apy with the effects of an add-on therapy, pioglitazone combined
lin sensitizers that reduce insulin resistance, increase glucose uptake
with voglibose, in type 2 diabetics on dialysis [36]. We found that
in muscles and adipose tissue, and decrease hepatic glucose produc-
the alpha-glucosidase inhibitor monotherapy reduced hemoglobin
tion [50, 51].
A1c levels approximately 0.4% in patients on dialysis. Furthermore,
our results suggested that voglibose is effective for maintaining the The two available glitazones (rosiglitazone and pioglitazone)
glycemic state, as it neither worsened nor improved the glycemic have an adequate oral bioavailability and are extensively metabo-
control in patients with a high level of insulin resistance during the lized by the liver. The main differences between thiazolidinediones
treatment period. Thus, we demonstrated that combination therapy of potential interest for use in the management of patients with type
with pioglitazone is more effective than voglibose monotherapy in 2 diabetes are summarized in Table 5. Rosiglitazone is mainly me-
achieving good glycemic control in type 2 diabetic patients with tabolized by CYP2C8 into inactive metabolites, and <1% of the
insulin resistance. parent drug appears in the urine in unchanged form [52, 53]. The
half-life of rosiglitazone is similar in patients with ESRD and in
Biguanide healthy individuals, and can therefore be administered to ESRD
With the exception of insulin, metformin is the most studied patients without dose adjustment or risk of causing hypoglycemia
therapy for type 2 diabetes. Metformin does not cause increased [54-56]. Pioglitazone is metabolized by CYP3A4 and CYP2C8/9
insulin levels, but rather decreases hepatic glucose output by sup- [57]. Metabolites of pioglitazone are more active than those of
pressing fasting gluconeogenesis. Although effective at improving rosiglitazone and are excreted predominantly in bile. The
62 Current Drug Metabolism, 2011, Vol. 12, No. 1 Abe et al.
Table 4. Main Clinically Relevant Pharmacokinetic Differences between the Three alpha-glucosidase Inhibitors
Administration With each meal preprandially With each meal preprandially With each meal preprandially
Table 5. Main Clinically Relevant Pharmacokinetic Differences between the Two Thiazolidinediones
pioglitazone metabolites, M-III and M-IV, do not accumulate in administered irrespective of the time of dialysis. Because of the
CKD. The pharmacokinetic profile of pioglitazone was found to be high molecular weight (392 Da), high protein-binding capacity
similar in healthy subjects and patients with moderately or severely (>98%), and predominant hepatic metabolism of pioglitazone, its
impaired renal function who did not require dialysis [57], while for pharmacokinetic profile is similar in patients with normal renal
those who did require dialysis [58], pioglitazone was found to have function and CKD, and in those undergoing dialysis therapy. How-
a Tmax of 1.8 h and a half-life of 5.4 h [57]. Therefore, a post- ever, thiazolidinediones have the potential to cause or exacerbate
dialysis supplementary dose is not required, and pioglitazone can be heart failure and should be used with caution in patients with he-
Antidiabetic Agents in CKD and ESRD Current Drug Metabolism, 2011, Vol. 12, No. 1 63
patic injury. Furthermore, it was reported that rosiglitazone is asso- without diabetes apparently improved the insulin sensitivity and
ciated with a significant increase in the risk of myocardial infarc- reduced the systolic and diastolic blood pressure due to the vasodi-
tion [59, 60]. In contrast, pioglitazone improved the cardiovascular latory effects of thiazolidinediones. Nonetheless, thiazolidinediones
outcome in type 2 diabetes patients who were not dependent on must be used with caution as they tend to increase fluid retention
dialysis therapy [61]. Therefore, further investigation is required to and can be associated with congestive heart failure.
determine whether these two drugs belonging to the thiazolidinedi-
one group are associated with cardiovascular events in type 2 diabe- DPP-4 Inhibitors
tes patients receiving dialysis. New compounds targeting the so-called incretin hormone sys-
Rosiglitazone and pioglitazone have been reported to (1) reduce tem may offer some advantages regarding many adverse events
insulin requirements, (2) play various roles in lipid metabolism, associated with other oral glucose-lowering agents. The intestinal
fibrinolysis, coagulation, and platelet aggregation, (3) ameliorate hormone glucagon-like peptide-1 (GLP-1) stimulates insulin secre-
albuminuria, (4) protect against impairment of endothelial function, tion in a glucose-dependent manner. However, its meal-induced
and (5) have an anti-inflammatory effect [62-65]. When used for secretion is generally decreased in patients with type 2 diabetes, and
the clinical management of type 2 diabetes and ESRD, thiazolidin- this may contribute to the amplification of postprandial hypergly-
ediones are primarily metabolized in the liver and will not accumu- cemia. GLP-1 is rapidly inactivated by the enzyme dipeptidylpepti-
late in patients with CKD. They might also improve uremia- dase-4 (DPP-4) [77]. Therefore, an effective way to potentiate post-
associated insulin resistance and confer benefits at the metabolic, prandial GLP-1 response is the use of selective DPP-4 inhibitors
inflammatory, vascular, and hemodynamic levels [65]. Agrawal et [78, 79]. The main differences between DPP-4 inhibitors of poten-
al. studied the effects of an add-on therapy of rosiglitazone admin- tial interest for use in the management of patients with type 2 diabe-
istered in combination with an SU (glyburide, gliclazide, or glipiz- tes are summarized in Table 6 [80].
ide) over a 6-month period in type 2 diabetics with mild to moder- Sitagliptin
ate renal impairment after SU monotherapy had failed to confer Sitagliptin is a highly selective, oral, once-daily, DPP-4 inhibi-
adequate glycemic control. Their results showed similar efficacy in tor approved for the treatment of patients with type 2 diabetes [81].
patients with normal renal function and in those with mild to mod- DPP-4 inhibitors slow the degradation and the inactivation of the
erate renal impairment [66]. Insulin resistance may lead to hyper- incretins, GLP-1 and glucose-dependent insulinotropic polypeptide
triglyceridemia through two independent mechanisms: first, by [77]. These two incretins regulate glucose homeostasis by stimulat-
enhancing hepatic synthesis of very low-density lipoprotein and ing insulin release, while GLP-1 also suppresses glucagon release.
triglycerides due to compensatory hyperinsulinemia, and second, by Both of these effects of incretins are glucose dependent [82].
decreased removal of triglycerides due to impaired regulation of CYP3A4 is the major CYP isozyme responsible for the limited
lipoprotein lipase activity by insulin [67]. It has been reported that oxidative metabolism of sitagliptin, with some minor contribution
pioglitazone administration is associated with mean decreases in from CYP2C8. However, the contribution of the CYP system in
triglyceride levels and mean increases in high-density lipoprotein sitagliptin metabolism is so small that clinically relevant drug-drug
(HDL)-cholesterol without consistent changes in the mean levels of interactions via CYP3A4 or CYP2C8 would probably be negligible
total cholesterol or low-density lipoprotein (LDL)-cholesterol in [83]. This is in contrast to reports of other glucose-lowering agents
non-uremic patients [68]. such as sulfonylureas, meglitinides (repaglinide), and thiazolidin-
Thiazolidinediones are known to reduce HOMA-IR and levels ediones (rosiglitazone). Sitagliptin is primarily renally eliminated
of high-sensitivity C-reactive protein (hs-CRP) and tumor necrosis with approximately 80% of the oral dose excreted unchanged in the
factor-alpha (TNF-), and increase adiponectin levels in patients urine [84, 85]. Excretion is thought to be via active secretion and
not undergoing dialysis. In patients undergoing peritoneal dialysis, glomerular filtration [85, 86]. Following single oral doses of sita-
thiazolidinediones have been reported to reduce hs-CRP levels, but gliptin, plasma sitagliptin exposure increases with decreasing renal
levels of interleukin-6 (IL-6) and TNF- were not reduced [47, 63]. function, as determined by 24-h CrCl. Relative to subjects with
In a short-term study of dialysis patients, thiazolidinediones were normal or mildly impaired renal function, patients with moderate
reported to reduce the levels of hs-CRP but not adiponectin [69]. It renal insufficiency (CrCl 30-50 mL/min), severe renal insufficiency
has been reported that pioglitazone treatment reduced the levels of (CrCl <30 mL/min, not on dialysis) or ESRD on dialysis had ap-
hs-CRP, IL-6, and TNF- and increased the high-molecular weight proximately 2.3-fold, 3.8-fold, or 4.5-fold higher plasma sitagliptin
adiponectin level, even in hemodialysis patients [70]. Furthermore, exposures, respectively, and the Cmax increased by 1.4-fold to 1.8-
the dosage of erythropoiesis-stimulating agents (ESA) was signifi- fold [87]. Tmax was significantly increased in patients with ESRD,
cantly reduced during pioglitazone treatment with improvement in and the terminal half-life increased with decreasing renal function.
insulin resistance and a decrease in the levels of inflammatory cy- Compared with values in subjects with normal renal function (13.1
tokines. Therefore, it was suggested that ESA responsiveness is h), the terminal half-life values of sitagliptin in those with mild,
enhanced with improvement in insulin resistance by long-term pio- moderate, and severe renal impairment, and ESRD were raised to
glitazone treatment [70]. 16.1, 19.1, 22.5, and 28.4 h, respectively. The fraction of dose re-
Potential side effects of PPAR treatment include fluid reten- moved by dialysis was low with 13.5% and 3.5% for dialysis initi-
tion, hemodilution, and weight gain. Thiazolidinedione-induced ated at 4 and 48 h post dose, respectively. Plasma protein binding of
weight gain is mediated through an increase in subcutaneous adi- 38% was not altered in uremic plasma from patients with renal
pose tissue accumulation and possibly a vasodilatory effect leading impairment. Based on these findings, to achieve plasma sitagliptin
to total-body fluid retention [71-73]. In any case, the higher risk of concentrations comparable to those in patients with normal renal
heart failure in patients receiving combination treatments of insulin function [88], sitagliptin dose adjustments are recommended for
and thiazolidinediones (2-3% in the combination group versus 1% patients with type 2 diabetes and moderate to severe renal insuffi-
in the monotherapy group) underscores the need for caution when ciency, as well as for those with ESRD requiring dialysis. Chan et
administering these therapies [74]. Pioglitazone administration is al. reported that sitagliptin dose-adjustment treatment reduced he-
associated with reductions in both systolic and diastolic blood pres- moglobin A1c by 0.7% at 54 weeks in patients with moderate to
sure [75]. This is consistent with the findings of a study of the ef- severe renal insufficiency, including those on peritoneal dialysis
fects of rosiglitazone on insulin resistance and blood pressure in [88].
patients with essential hypertension but without diabetes [76]; a Vildagliptin
decline in systolic blood pressure was correlated with an improve- Because vildagliptin is not a CYP enzyme substrate, and does
ment in insulin sensitivity. Rosiglitazone treatment in hypertensives not inhibit or induce CYP enzymes, it is unlikely to interact with
64 Current Drug Metabolism, 2011, Vol. 12, No. 1 Abe et al.
Table 6. Main Clinically Relevant Pharmacokinetic Differences Between the Five Dipeptidylpeptidase-4 Inhibitors
co-medications that are substrates, inhibitors, or inducers of these urine [93]. In patients with type 2 diabetes, alogliptin was also pri-
enzymes [89, 90]. Vildagliptin does not affect the metabolic clear- marily excreted renally (mean fraction of drug excreted in urine
ance of co-medications metabolized by CYP1A2, CYP2C8, from 0-72 h after dosing, 60.8-63.4%), with a renal clearance rate
CYP2C9, CYP2C19, CYP2D6, CYP2E1, or CYP3A4/5 [80]. of 165-254 mL/min; this is slightly higher than the normal GFR,
Based on an in vitro recombinant DPP-4 assay, the inhibitory con- suggesting the occurrence of some active renal secretion. Metabo-
centration 50% (IC50) for vildagliptin is 2 nM. In humans, the effi- lism of alogliptin constituted a minor pathway; although N-
cacy of vildagliptin against the DPP-4 enzyme also shows a low in demethylated metabolite (M-I) is pharmacologically active and has
vivo IC50 (4.5 nM), which suggests a higher potency than that re- exhibited a similar potency and degree of selectivity for DPP-4 as
ported for sitagliptin (IC50 26 nM) [85, 91]. Vildagliptin is rapidly the parent compound in vitro, the AUC and Cmax of M-I was <1%
absorbed in fasting humans after a single oral dose of 100 mg; ab- of the AUC and Cmax of parent drug, confirming that it is a minor
sorption is high (>85.4%) and dose recovery complete. The major metabolite in humans [94]. The results of a single-dose (50 mg)
circulating components in the plasma were unchanged drug and pharmacokinetic study in patients with renal impairment showed an
main metabolite (M20.7) which was not biologically active. Elimi- increase in alogliptin exposure compared with healthy volunteers:
nation of vildagliptin mainly involves renal excretion of unchanged approximately 1.7-, 2.1-, 3.2-, and 3.8-fold increase in patients with
parent drug and cyano group hydrolysis with little CYP involve- mild, moderate, and severe renal impairment, and in patients with
ment, suggesting a low potential for drug-drug interaction when co- ESRD, respectively [80, 95]. Based on these findings, to achieve
administered with CYP inhibitors/inducers. Moreover, vildagliptin plasma alogliptin concentrations comparable to those in patients
is unlikely to inhibit the metabolic clearance of co-medications with normal renal function, alogliptin dose adjustments are recom-
metabolized by CYP enzymes [92]. mended for patients with type 2 diabetes and moderate to severe
In patients with mild, moderate, and severe renal impairment, renal insufficiency, including those with ESRD requiring dialysis.
and ESRD patients on hemodialysis, systemic exposure to vilda- Saxagliptin
gliptin was increased (Cmax 8-66%; AUC 32-134%) compared to The metabolism of saxagliptin is primarily mediated by
subjects with normal renal function. However, changes in exposure CYP3A4/5. The major metabolite of saxagliptin is also a selective,
to vildagliptin did not correlate with the severity of renal function. reversible, competitive DPP-4 inhibitor, which is 50% less potent
In contrast, exposure of the main metabolite (M20.7) increased with than saxagliptin [96]. The fact that the metabolite of saxagliptin is
increasing severity of renal function (AUC 1.6- to 6.7-fold), but this pharmacologically active is in contrast to the other DPP-4 inhibitors
effect has no clinically relevant consequences because the metabo- whose metabolites are inactive. Saxagliptin is cleared by both me-
lite is pharmacologically inactive. The elimination half-life of vil- tabolism and renal excretion. The effects of renal impairment and
dagliptin was not affected by renal function. Half of all patients in dialysis on the pharmacokinetics of saxagliptin have been deter-
the global development program exposed to the marketed doses of mined [80]. The degree of renal impairment did not affect the Cmax
vildagliptin as monotherapy had either mild or moderate renal im- of saxagliptin or its major metabolite. In subjects with mild renal
pairment at study baseline. Vildagliptin was effective and well tol- impairment, AUC from time 0 to infinity (AUC ) values of saxa-
erated in this population [80]. According to the labeling, no dosage gliptin and its major metabolite were 1.2- and 1.7-fold higher, re-
adjustment of vildagliptin is required in patients with mild renal spectively, than mean AUC values in controls. Corresponding
impairment. In clinical practice, special precautions are advised for values were 1.4- and 2.9-fold higher in subjects with moderate renal
the use of vildagliptin in patients with moderate to severe renal impairment, and 2.1- and 4.5-fold higher in those with severe im-
impairment, including those on dialysis. pairment. A 4-h dialysis session removed 23% of the saxagliptin
Alogliptin dose. AUC values for saxagliptin and, to a greater extent, its major
Alogliptin was rapidly absorbed (median Tmax, 1-2 h) and metabolite were correlated with the degree of renal impairment,
slowly eliminated (mean T1/2, 12.4-21.4 h), primarily via urinary whereas Cmax values were not well correlated. Kidney function
excretion (mean fraction of drug excreted unchanged in urine from should be assessed before initiating saxagliptin therapy and patients
0-72 h after dosing, 60-71%) in healthy subjects. The metabolites with moderate to severe kidney impairment should receive no >2.5
M-I (N-demethylated) and M-II (N-acetylated) accounted for <2% mg/day [97]. Saxagliptin can be taken after dialysis in patients with
and <6%, respectively, of alogliptin concentrations in plasma and ESRD.
Antidiabetic Agents in CKD and ESRD Current Drug Metabolism, 2011, Vol. 12, No. 1 65
cytes, has been suggested as a better indicator of glycemic control, [14] Nolin, T.D.; Naud, J.; Leblond, F.A.; Pichette, V. Emerging evi-
and glycated albumin levels may become the target of glycemic dence of impacy of kidney disease on drug metabolism and trans-
control in diabetic dialysis patients. However, there is no clear con- port. Clin. Pharmacol. Ther., 2008, 83(6) 898-903.
sensus on the target hemoglobin A1c and glycated albumin levels [15] Nolin, T.D.; Frye, R.F.; Matzke, G.R. Hepatic drug metabolism and
for a good prognosis of diabetic dialysis patients. This will need to transport in patients with kidney disease. Am. J. Kidney. Dis., 2003,
42(5), 906-925.
be clarified by long-term studies.
[16] Sun, H.; Frassetto, L.; Benet, L.Z. Effects of renal failure on drug
transport and metabolism. Pharmacol. Ther., 2006, 109(1-2), 1-11.
CONCLUSION
[17] Salsburg, D.S. The UGDP study. JAMA, 1971, 218(11), 1704-
Although therapeutic possibilities with oral agents in type 2 1705.
diabetes mellitus are increasing, the presence of CKD is an impor- [18] United Kingdom Prospective Diabetes Study Group (UKPDS).
tant limitation for using the majority of the currently available oral Intensive blood-glucose control with sulphonylureas or insulin
antidiabetic drugs. In recent years, several promising drugs have compared with conventional treatment and risk of complications in
been introduced in the therapeutic armamentarium of affected pa- patients with type 2 diabetes (UKPDS 33). Lancet, 1998,
tients. However, recommended oral antidiabetic agents differ be- 352(9131), 837-853.
tween countries. Therefore, kidney function of each patient should [19] Zoungas, S.; de Galan, B.E.; Ninomiya, T.; Grobbee, D.; Hamet,
P.; Heller, S.; MacMahon, S.; Marre, M.; Neal, B.; Patel, A.;
be assessed before initiating antidiabetic therapy, and careful moni-
Woodward, M.; Chalmers, J.; ADVANCE Collaborative Group;
toring against hypoglycemia is necessary, in particular when using Cass, A.; Glasziou, P.; Harrap, S.; Lisheng, L.; Mancia, G.; Pillai,
insulin secretagogues or combination therapy. Whether the pharma- A.; Poulter, N.; Perkovic, V.; Travert, F. Combined effects of rou-
cokinetic differences outlined above will translate into clinically tine blood pressure lowering and intensive glucose control on mac-
relevant differences concerning the efficacy and safety profiles of rovascular and microvascular outcomes in patients with type 2
these drugs in patients with type 2 diabetes with CKD or ESRD diabetes: New results from the ADVANCE trial. Diabetes Care,
remains to be determined. Furthermore, the long-term efficacy and 2009, 32(11), 2068-2074.
safety of the DPP-4 inhibitors remain largely unknown. In particu- [20] Rosenkranz, B.; Profozic, V.; Metelko, Z.; Mizljak, V.; Lange, C.;
lar, clinical data on the durability of glucose control, cardiovascular Malerczyk V. Pharmacokinetics and safety of glimepiride at clini-
outcomes, diabetic complications, all-cause mortality, and long- cally effective doses in diabetic patients with renal impairment.
term safety for patients with CKD on dialysis are still needed. Diabetologia, 1996, 39(12), 1617-1624.
[21] Available from URL: http://www.pfizer.com/files/ prod-
REFERENCES ucts/uspi_glipizide.pdf (Accessed October 1, 2010)
[22] Kajosaari, L.I.; Laitila, J.; Neuvonen, P.J.; Backman, J.T. Metabo-
[1] Cao, J.J.; Hudson, M.; Jankowski, M.; Whitehouse, F.; Weaver, lism of repaglinide by CYP2C8 and CYP3A4 in vitro: effect of fi-
W.D. Relation of chronic and acute glycemic control on mortality brates and rifampicin. Basic Clin. Pharmacol. Toxicol., 2005,
in acute myocardial infarction with diabetes mellitus. Am. J. Car- 97(4), 249-256.
diol., 2005, 96(2), 183-186. [23] Kajosaari, L.I.; Niemi, M.; Backman, J.T.; Neuvonen, P.J. Te-
[2] Gaede, P.; Vedel, P.; Larsen, N.; Jensen, G..V.; Parving, H.H.; lithromycin, but not montelukast, increases the plasma concentra-
Pedersen, O. Multifactorial intervention and cardiovascular disease tions and effects of the cytochrome P450 3A4 and 2C8 substrate
in patients with type 2 diabetes. N. Engl. J. Med., 2003, 348(5), repaglinide. Clin. Pharmacol. Ther., 2006, 79(39), 231-242.
383-393. [24] Niemi, M.; Backman, J.T.; Juntti-Patinen, L.; Neuvonen, M.; Neu-
[3] Garber, A.J. Attenuating CV risk factors in patients with diabetes: vonen, P.J. Coadministration of gemfibrozil and itraconazole has
clinical evidence to clinical practice. Diabetes. Obes. Metab., 2002, only a minor effect on the pharmacokinetics of the CYP2C9 and
4, S5-12. CYP3A4 substrate nateglinide. Br. J. Clin. Pharmacol., 2005,
[4] Kassab, E.; McFarlane, S.I.; Sower, J.R. Vascular complications in 60(2), 208-217.
diabetes and their prevention. Vasc. Med. 2001, 6(4), 249-255. [25] Murbury, T.C.; Ruckle, J.L.; Hatorp, V.; Andersen, M.P.; Nielsen,
[5] Colwell, J.A. Vascular thrombosis in type II diabetes mellitus. K.K.; Huang, W.X.; Strange, P. Pharmakokinetics of repaglinide in
Diabetes, 1993, 42(1), 8-11. subjects with renal impairment. Clin. Pharmacol. Ther., 2000,
[6] Kidney Disease Outcomes Quality Initiative (KDOQI). Clinical 67(1), 7-15.
practice guidelines and clinical practice recommendations for dia- [26] Yu, L.; Lu, S.; Lin, Y.; Zeng, S. Carboxyl-glucuronidation of
betes and chronic kidney disease. Am. J. Kidney. Dis., 2007, 49(2), mitiglinide by human UDP-glucuronosyltransferases. Biochem.
S62-S73. Pharmacol., 2007, 73(11), 1842-1851.
[7] Yu, C.C.; Wu, M.S.; Wu, C.H.; Young, C.W.; Huang, J.Y.; Hong, [27] Yu, L.; Lu, S.; Lin, Y.; Zeng, S. Carboxyl-glucuronidation of
J.J.; Fan Cjiang, C.Y.; Leu, M.L.; Huang, C.C. Predialysis glyce- mitiglinide by human UDP-glucuronosyltransferases. Biochem.
mic control is an independent predictor of clinical outcome in type Pharmacol., 2007, 73, 1842-1851.
II diabetics on continuous ambulatory peritoneal dialysis. Perit. [28] Reimann, F.; Proks, P.; Ashcroft, F.M. Effects of mitiglinide (S
Dial. Int., 1997, 17(3), 262-268. 21403) on Kir6.2/SURl, Kir6.2/SUR2A and Kir6.2/SUR2B types
[8] Morioka, T.; Emoto, M.; Tabata, T.; Shoji, T.; Tahara, H.; Kishi- of ATP-sensitive potassium channel. Br. J. Pharmacol., 2001,
moto, H.; Ishimura, E.; Nishizawa, Y. Glycemic control is a predic- 132(7), 1542-1548.
tor of survival for diabetic patients on hemodialysis. Diabetes [29] Kaku, K.; Tanaka, S.; Origasa, H.; Kikuchi, M.; Akanuma, Y.
Care, 2001, 24(5), 909-913. Addition of mitiglinide to pioglitazone monotherapy improves
[9] Yale, J.F. Oral antihyperglycemic agents and renal disease: new overall glycemic control in Japanese patients with type 2 diabetes:
agents, new concepts. J. Am. Soc. Nephrol., 2005, 16, 7-10. A randomized double blind trial. Endocr. J., 2009, 56(5), 657-664.
[10] Senekjian, H.O.; Lee, C.S.; Kuo, T.H.; Au, D.S.; Krothapalli, R. [30] Kaku, K.; Tanaka, S.; Orisawa, H.; Kikuchi, M.; Akanuma, Y.
Absorption and disposition of ibuprofen in hemodialyzed uremic Effect of mitiglinide on glycemic control over 52 weeks in Japa-
patients. Eur. J. Rheumatol. Inflam. 1983, 6(2), 155-162. nese type 2 diabetic patients insufficiently controlled with pioglita-
[11] Zazgornik, J.; Huang, M.L.; Van Peer, A.; Woestenborghs, R.; zone monotherapy. Endocr. J., 2009, 56(6), 739-746.
Heykants, J.; Stephen, A. Pharmacokinetics of orally administered [31] Gao, X. The mitiglinide versus nateglinide comparison group.
levocabastine in patients with renal insufficiency. J. Clin. Pharma- Multicentre, double-blind, randomized study of mitiglinide coma-
col., 1993, 32(12), 1214-1218. pared with nateglinide in type 2 diabetes mellitus patients in China.
[12] Touchette, M.A.; Slaughter, R.L. The effect of renal failure on J. Int. Med. Res., 2009, 37(8), 812-821.
hepatic drug clearance. DICP, 1991, 25(11), 1214-1224. [32] Abe, M. ; Okada, K.; Maruyama, T.; Maruyama, N.; Maysumoto,
[13] Naud, J.; Michaud, J.; Leblond, F.A.; Bernier, G.; Bonnardeaux, K. Combination therapy with mitiglinide and voglibose improves
A.; Pichette, V. Effects of chronic renal failure on liver drug trans- glycemic control in type 2 diabetic patients on hemodialysis. Ex-
porters. Drug. Metab. Dispos., 2008, 36(1), 124-128. pert Opin. Pharmacother., 2010, 11(2), 169-176.
Antidiabetic Agents in CKD and ESRD Current Drug Metabolism, 2011, Vol. 12, No. 1 67
[33] Abe, M.; Okada, K.; Maruyama, T.; Maruyama, N.; Maysumoto, [55] Goldstein, B.J. Rosiglitazone. Int. J. Clin. Pract., 2000, 54(5), 333-
K. Efficacy and safety of mitiglinide in diabetic patients on main- 337.
tenance hemodialysis. Endcrine. J., 2010, 57(7), 581-588. [56] Thompson, C.K.; Zussman, B.; Miller, A.K.; Freed, M.I. Pharma-
[34] Chiasson, J.L.; Gomis, R.; Hanefeld, M.; Josse, R.G.; Karasik, A.; kokinetics of rosiglitazone in patients with end stage renal disease.
Laakso, M. The STOP-NIDDM Trial: an international study on the J. Int. Med. Res., 2002, 30(4), 391-399.
efficacy of an alpha-glucosidase inhibitor to prevent type 2 diabetes [57] Budde, K.; Neumayer, H.H.; Fritsche, L.; Sulowicz, W.; Stompor,
in a population with impaired glucose tolerance: rationale, design, T.; Eckland, D. The pharmacokinetics of pioglitazone in patients
and preliminary screening data. Study to Prevent Non-Insulin- with impaired renal function. Br. J. Clin. Pharmacol. 2003, 55(4),
Dependent Diabetes Mellitus. Diabetes Care, 1998,21(10), 1720- 368-374.
1725. [58] Abe, M.; Kikuchi, F.; Okada, K.; Matsumoto, K. Plasma concentra-
[35] Chiasson, J.L.; Josse, R.G.; Gomis, R.; Hanefeld, M.; Karasik, A.; tion of pioglitazone in patients with type 2 diabetes on hemodialy-
Laakso, M. Acarbose treatment and the risk of cardiovascular dis- sis. Therap. Apher. Dial., 2009, 13(3), 238-239.
ease and hypertension in patients with impaired glucose tolerance: [59] Nissen, S.E.; Wolski, K. Effect of rosiglitazone on the risk of myo-
the STOP-NIDDM trial. JAMA, 2003, 290(4), 486-494. cardial infarction and death from cardiovascular causes. N. Engl. J.
[36] Abe, M.; Kikuchi, F.; Kaizu, K.; Matsumoto, K. Combination Med., 2007, 356(24), 2457-2471.
therapy of pioglitazone with vogliboseimproves glycemic control [60] Graham, D.J.; Hellstrom, R.O.; MaCurdy, T.E.; Ali, F.; Sholley,
safely and rapidly in Japanese type 2-diabetic patients on hemo- C.; Worrall, C.; Kelman, J.A. Risk of acute myocardial infarction,
dialysis. Clin. Nephrol., 2007, 68(5), 287-294. stroke, heart failure, and death in elderly medicare patients treated
[37] Available from URL: http://www.accessdata.fda.gov/ drugsat- with rosiglitazone or pioglitazone. JAMA, 2010, 304(4), 411-418.
fda_docs/label/2008/020482s023lbl.pdf (Accessed Jan 15, 2011). [61] Dormandy, J.A.; Charbonnel, B.; Eckland, D.J.; Erdmann, E.;
[38] Available from URL: http://www.pfizer.com/files/ prod- Massi-Benedetti, M.; Moules, I.K.; Skene, A.M.; Tan, M.H.; Le-
ucts/uspi_glyset.pdf (Accessed Jan 15, 2011). fèbvre, P.J.; Murray, G.D.; Standl, E.; Wilcox, R.G.; Wilhelmsen,
[39] United Kingdom Prospective Diabetes Study Group (UKPDS).13; L.; Betteridge, J.; Birkeland, K.; Golay, A.; Heine, R.J.; Korányi,
Relative efficacy of randomly allocated diet, sulphonylurea, insu- L.; Laakso, M.; Mokán, M.; Norkus, A.; Pirags, V.; Podar, T.;
lin, or metformin in patients with newly diagnosed non-insulin de- Scheen, A.; Scherbaum, W.; Schernthaner, G.; Schmitz, O.; Skrha,
pendent diabetes followed for three years. B.M.J., 1995, 310(6972), J.; Smith, U.; Taton, J. PROactive investigators. Secondary preven-
83-88. tion of macrovascular events in patients with type 2 diabetes in the
[40] United Kingdom Prospective Diabetes Study Group (UKPDS). PROactive Study (Prospective pioglitazone clinical trial in mac-
Effect of intensive blood-glucose control with metformin on com- rovascular events): a randomized controlled trial. Lancet, 2005,
plications in overweight patients with type 2 diabetes (UKPDS 34). 366(9493), 1279-1289.
Lancet, 1998, 352(9131), 854-865. [62] Martens, F.M.; Visseren, F.L.; Lemay, J.; de Koning, E.J.; Ra-
[41] Bailey, C.J.; Turner, R.C. Metformin. N. Engl. J. Med., 1996, belink, T.J. Metabolic and additional vascular effects of thia-
334(9), 574-579. zolidinediones. Drugs, 2002, 62(10), 1463-1480.
[42] Cusi, K.; Consoli, A.; DeFronzo, R.A. Metabolic effects of met- [63] Lin, S.H.; Lin, Y.F.; Kuo, S.W.; Hsu, Y.J.; Hung, Y.J. Rosiglita-
formin on glucose and lactate metabolism in noninsulin-dependent zone improves glucose metabolism in nondiabetic uremic patients
diabetes mellitus. J. Clin. Endo. Met., 1996, 81(11), 4059-4067. on CAPD. Am. J. Kid. Dis., 2003, 42(4), 774-780.
[43] Mamputu, J.C.; Wiernsperger, N.F.; Renier, G. Antiatherogenic [64] Sarafidis, P.A.; Bakris, G.L. Protection of the kidney by thia-
properties of metformin: the experimental evidence. Diabetes Me- zolidinediones: An assessment from bench to bedside. Kidney Int.,
tab., 2003, 29(4 Pt 2), 6S71-76. 2006, 70(7), 1223-1233.
[44] Sirtori CR, Franceschini G, Galli-Kiele M, Galli G, Bondioli A, [65] Iglesias, P.; Díez, J.J. Peroxisome proliferator-activated receptor
Conti F. Disposition of metformin (N,N-dimethylbiguanide) in gamma agonists in renal disease. Eur. J. Endocrinol., 2006, 154(5),
man. Clin. Pharmacol. Ther., 1978, 24(6), 683-193. 613-621.
[45] Available from URL: http://www.sanofi-aventis.ca/products/en/ [66] Agrawal, A.; Sautter, M.C.; Jones, N.P. Effects of rosiglitazone
glucophage.pdf (Accessed Jan 15, 2011). maleate when added to a sulfonylurea regimen in type 2 diabetics
[46] Lalau, J.D.; Andrejak, M.; Moriniere, P.; Coevoet, B.; Debussche, mellitus and mild to moderate renal impairment: a post hoc analy-
X.; Westeel, P.F.; Fournier, A.; Quichaud, J. Hemodialysis in the sis. Clin. Ther., 2003, 25(11), 2754-2764.
treatment of lactic acidosis in diabetics treated by metformin: a [67] Defronzo, R.A.; Ferrannini, E. Insulin resistance: a multifaceted
study of metformin ekimination. Int. J. Clin. Pharmacol. Ther. syndrome responsible for NIDDM, obesity, hypertension, dyslipi-
Toxicol., 1989, 27(6), 285-288. demia, and atherosclerotic cardiovascular disease. Diabetes Care,
[47] Wong, T.Y.; Szeto, C.C.; Chow, K.M.; Leung, C.B.; Lam, C.W.; 1991, 14(3), 173-194.
Li, P.K. Rosiglitazone reduces insulin requirement and C-reactive [68] Ginsberg, H.; Plutzky, J.; Sobel, B.E. A review of metabolic and
protein levels in type 2 diabetic patients receiving peritoneal dialy- cardiovascular effects of oral antidiabetic agents: beyond glucose-
sis. Am. J. Kidney Dis., 2005, 46(4), 713-719. level lowering. J. Cardiovasc. Risk., 1999, 6(5), 337-346.
[48] Campbell, I.W. The clinical significance of PPAR agonism. Curr. [69] Chiang, C.K.; Ho, T.I.; Peng, Y.S.; Hsu, S.P.; Pai, M.F.; Yang,
Mol. Med., 2005, 5(3), 349-363. S.Y.; Hung, K.Y.; Wu, K.D. Rosiglitazone in diabetes control in
[49] Shinohara, K.; Shoji, T.; Emoto, M.; Tahara, H.; Koyama, H.; hemodialysis patients with and without viral hepatitis infection.
Ishimura, E.; Miki, T.; Tabata, T.; Nishizawa, Y. Insulin resistance Diabetes Care, 2007, 30(1), 3-7.
as an independent predictor of cardiovascular mortality in patients [70] Abe, M.; Okada, K.; Maruyama, T.; Maruyama, N.; Soma, M.;
with end-stage renal disease. J. Am. Soc. Nephrol., 2002, 13(7), Matsumoto, K. Clinical effectiveness and safety evaluation of long-
1894-1900. term pioglitazone treatment for erythropoietin responsiveness and
[50] Ciaraldi, T.P.; Huber-Knudsen, K.; Hickman, M.; Olefsky, J.M. insulin resistance in type 2 diabetic patients on hemodialysis. Ex-
Regulation of glucose transport in culture muscle cells by novel pert Opin. Pharmacother., 2010, 11(10), 1611-1620.
hypoglycemic agents. Metabolism, 1995, 44(8), 976-982. [71] Manley, H.J.; Allcock, N.M. Thiazolidinedione safety and efficacy
[51] Spiegelman, B.M. PPAR-: adipogenic regulator and thiazolidin- in ambulatory patients receiving hemodialysis. Pharmacotherapy,
edione receptor. Diabetes, 1998, 47(4), 507-514. 2003, 23(7), 861-865.
[52] Krentz AJ, Bailey CJ. Oral antidiabetic agents: current role in type [72] Phillips, L.S.; Grunberger, G.; Miller, E.; Patwardhan, R.; Rappa-
2 diabetes mellitus. Drugs, 2005, 65(3), 385-411. port, E.; Salzman, A. Once- and twice- daily dosing with rosiglita-
[53] Snyder, R.W.; Berns, J.S. Use of insulin and oral hypoglycemic zone improves glycemic control in patients with type 2 diabetes.
medications in patients with diabetes mellitus and advanced kidney Diabetes Care, 2001, 24(2), 308-315.
disease. Semin. Dial., 2004, 17(5), 365-370. [73] Aronoff, S.; Rosenblatt, S.; Braithwaite, S.; Egen, J.W.; Mathisen,
[54] Cox, P.J.; Ryan, D.A.; Hollis, F.J.; Harris, A.M.; Miller, A.K.; A.L.; Schneider, R.L. Pioglitazone hydrochloride monotherapy im-
Vousden, M.; Cowley, H. Absorption, disposition, and metabolism proves glycemic control in the treatment of patients with type 2
of rosiglitazone, a potent thiazolidinedione insulin sensitizer, in diabetes. Diabetes Care, 2000, 23(11), 1605-1611.
humans. Drug Metab. Dispos., 2000, 28(7), 772-780.
68 Current Drug Metabolism, 2011, Vol. 12, No. 1 Abe et al.
[74] Nesto, R.W.; Bell, D.; Bonow, R.Q.; Fonseca, V.; Grandy, S.M.; [91] He, Y.L.; Serra, D.; Wang, Y.; Campestrini, J.; Riviere, G.J.; Dea-
Horton, E.S.; Le Winter, M.; Porte, D.; Semenkovich, C.F.; Smith, con, C.F.; Holst, J.J.; Schwartz, H.S.; Nielsen, J.C.; Ligueros-
S.; Young, L.H.; Kahn, R. Thiazolidinedione use, fluid retention Saylan, M. Pharmacokinetics and pharmacodynamics of vilda-
and congestive heart failure. A consensus statement from the AHA gliptin in patients with type 2 diabetes mellitus. Clin. Pharmacoki-
and ADA. Circulation, 2003, 108(23), 2941-2948. net., 2007, 46(7), 577-588.
[75] Abe, M.; Okada, K.; Kikuchi, F.; Matsumoto, K. Clinical investiga- [92] He, H.; Yin, T.H.; Smith, H.; Batard, Y.; Wang, L.; Einolf, H.; Gu,
tion of the effects of pioglitazone on the improvement of insulin re- H.; Mangold, J.B.; Fischer, V.; Howard, D. Absorption, metabo-
sistance and blood pressure in type 2-diabetic patients undergoing lism, and excretion of [14C]Vildagliptin, a novel dipeptidyl pepti-
hemodialysis. Clin. Nephrol., 2008, 70(3), 220-228. dase 4 inhibitor, in humans. Drug Metab. Dispos., 2009, 37(3),
[76] Raji, A.; Seely, E.W.; Bekins, S.A.; Williams, G.H.; Simonson, 536-544.
D.C. Rosiglitazone improves insulin sensitivity and lowers blood [93] Christopher, R.; Covington, P. ; Davenport, M. ; Fleck, P. ; Mekki,
pressure in hypertensive patients. Diabetes Care, 2003, 26(1), 172- Q.A.; Wann, E.R.; Karim, A. Pharmacokinetics, pharmacodynam-
178. ics, and tolerability of single increasing doses of the dipeptidyl pep-
[77] Drucker, D.J.; Nauck, M.A. The incretin system: glucagon-like tidase-4 inhibitor alogliptin in healthy male subjects. Clin. Ther.,
peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in 2008, 30(3), 513-527.
type 2 diabetes. Lancet, 2006, 368(9548), 1696-1705. [94] Karim, A.; Covington, P. ; Christopher, R.; Davenport, M.; Fleck,
[78] Idris, I.; Donnelly, R. Dipeptidyl peptidase-IV inhibitors: a major P.; Li, X.; Wann, E.; Mekki, Q. Pharmacokinetic of alogliptin when
new class of oral antidiabetic drug. Diabetes. Obes. Metab., 2007, administered with food, metformin, or cimetidine: a two-phase,
9(2), 153-165. crossover study in healthy subjects. Int. J. Clin. Pharmacol. Ther.,
[79] Richter, B.; Banderia-Echtler, E.; Bergerhoff, K.; Lerch. C.L. Di- 2010, 48(1), 46-58.
peptidyl peptidase-4 (DPP-4) inhibitors for type 2 diabetes melli- [95] Partley, R.E. Alogliptin: a new. highly selective dipeptidyl pepti-
tus. Cochrane. Database. Syst. Rev., 2008, 16(2), CD006739. dase-4 inhibitor for the treatment of type 2 diabetes. Expert Opin.
[80] Scheen, A.J. Pharmacokinetics of dipeptidylpeptidase-4 inhibitors. Pharmacother., 2009, 10, 503-512.
Diabetes Obes. Metab., 2010, 12(8), 648-658. [96] Fura, A.; Khanna, A.; Vyas, V.; Koplowitz, B.; Chang, S.Y.; Ca-
[81] Herman, G.A.; Stein, P.P.; Thornberry, N.A.; Wagner, J.A. Dipep- poruscio, C.; Boulton, D.W.; Christopher, L.J.; Chadwick, K.D.
tidyl peptidase-4 inhibitors for the treatment of type 2 diabetes: fo- Pharmacokinetics of the dipeptidyl peptidase 4 inhibitor saxagliptin
cus on sitagliptin. Clin. Pharmacol. Ther., 2007, 81(5), 761-767. in rats, dogs, and monkeys and clinical projections. Drug Metab.
[82] Drucker, D.J. The biology of incretin hormones. Cell Metab., 2006, Dispos., 2009; 37(6): 1164-1171.
3(3), 153-165. [97] Available from URL: http://www.ema.europa.eu/humandocs/
[83] Vincent, S.H.; Reed, J.R.; Bergman, A.J.; Elmore, C.S.; Zhu, B.; PDFs/EPAR/ onglyza/H-1039-fr1.pdf (Accessed Jan 15, 2011).
Xu, S.; Ebel, D.; Larson, P.; Zeng, W.; Chen, L.; Dilzer, S.; Lasse- [98] Fuchs, H.; Tillement, J.P.; Urien, S.; Greischel, A.; Roth, W. Con-
ter, K.; Gottesdiener, K.; Wagner, J.A.; Herman, G.A. Metabolism centrationdependent plasma protein binding of the novel dipeptidyl
and Excretion of the dipeptidyl peptidase 4 inhibitor peptidase 4 inhibitor BI 1356 due to saturable binding to its target
[14C]Sitagliptin in humans. Drug Metab. Dispos., 2007, 35(4), 533- in plasma of mice, rats and humans. J. Pharm. Pharmacol., 2009,
538. 61(1), 55-62.
[84] Herman, G.A.; Bergman, A.; Stevens, C.; Kotey, P.; Yi, B.; Zhao, [99] Fuchs, H.; Binder, R.; Greischel, A. Tissue distribution of the novel
P.; Dietrich, B.; Golor, G.; Schrodter, A.; Keymeulen, B.; Lasseter, DPP-4 inhibitor BI 1356 is dominated by saturable binding to its
K.C.; Kipnes, M.S.; Snyder, K.; Hilliard, D.; Tanen, M.; Cilissen, target in rats. Biopharm. Drug Dispos., 2009, 30, 229-240.
C.; De Smet, M.; de Lepeleire, I.; Van Dyck, K.; Wang, A.Q.; [100] Blech, S.; Ludwig-Schwellinger, E.; Graefe-Mody, E.U.; Withopf,
Zeng, W.; Davies, M.J.; Tanaka, W.; Holst, J.J.; Deacon, C.F.; Got- B.; Wagner, K. The metabolism and disposition of the oral dipepti-
tesdiener, K.M.; Wagner, J.A. Effect of single oral doses of sita- dyl peptidase-4 inhibitor, linagliptin, in humans. Drug Metab. Dis-
gliptin, a dipeptidyl peptidase-4 inhibitor, on incretin and plasma pos., 2010, 38(4), 667-678.
glucose levels following an oral glucose tolerance test in patients [101] Huttner, S.; Graefe-Mody, E.U.; Withopf, B.; Ring, A.; Dugi, K.A.
with type 2 diabetes. J. Clin. Endocrinol. Metab., 2006, 91(11), Safety, tolerability, pharmacokinetics and pharmacodynamics of
4612-4619. single oral doses of Bl 1356, an inhibitor of dipeptidylpeptidase 4,
[85] Herman, G.A.; Stevens, C.; Van Dyck, K.; Bergman, A.; Yi, B.; De in healthy male volunteers. J. Clin. Pharmacol. , 2008, 48(10),
Smet, M.; Snyder, K.; Hilliard, D.; Tanen, M.; Tanaka, W.; Wang, 1171-1178.
A.Q.; Zeng, W., Musson, D.; Winchell, G.; Davies, M.J.; Ramael, [102] Holst, J.J. Enteroglucagon. Ann. Rev. Physiol., 1997, 59, 257-271.
S.; Gottesdiener, K.M., Wagner, J.A. Pharmacokinetics and phar- [103] Meier, J.J.; Gallwitz, B.; Nauck, M.A. Glucagon-like peptide and
macodynamics of single doses of sitagliptin, an inhibitor of dipep- gastric inhibitory polypeptide: potential applications in type 2 dibe-
tidyl peptidase-IV, in healthy subjects: results from two random- tes mellitus. Bio. Drugs, 2003, 17(2), 93-102.
ized, double-blind, placebo-controlled studies with single oral [104] Nauck, M.A.; Meier, J.J.; Creutzfeldt, W. Incretins and their ana-
doses. Clin. Pharmacol Ther., 2005, 78(6), 675-688. logues as new antidiabetic agents. Drug News Perspect., 2003,
[86] Chu, X.Y.; Bleasby, K.; Yabut, J.; Cai, X.; Chan, G.H.; Hafey, 16(7), 413-422.
M.J.; Xu, S.; Bergman, A.J.; Braun, M.P.; Dean, D.C.; Evers, R. [105] Nauck, M.A.; Hompesch, M.; Filipczak, R.; Le, T.D.T.; Zdravk-
Transport of the dipeptidyl peptidase-4 inhibtor sitagliptin by hu- ovic, M.; Gumprecht, J. Five weeks of treatment with GLP-1 ana-
man organic anion transporter 3, organic anion transporting poly- logue liraglutide improves glycemic control and lowers body
peptide 4C1, and multidrug resistance P-glycoprotein. J. Pharma- weight in subjects with type 2 diabetes. Exp. Clin. Endocrinol.
col. Exp. Ther., 2007, 321(2), 673-683. Diabetes., 2006, 114(8), 417-423.
[87] Bergman, A.J.; Cote, J.; Yi, B.; Marbuy, T.; Swan, S.K.; Smith, [106] Baggio, L.L.; Drucker, D.J. Biology of incretins: GLP-1 and GIP.
W.; Gottesdiener, K.; Wagner, J.; Herman, G.A. Effect of renal in- Gastroenterology, 2007, 132(6), 2131-2157.
sufficiency on the pharmacokinetics of sitagliptin, a dipeptidyl pep- [107] Copley, K.; McCowen, K.; Hiles, R.; Nielsen, L.L.; Young, A.;
tidase-4 inhibitor. Diabetes Care, 2007, 30(7), 1862-1864. Parkes, D.G. Investigation of exenatide elimination and its in vivo
[88] Chan, J.C.; Scott, R.; Arjona Ferreira, J.C.; Sheng, D.; Gonzalez, and in vitro degradation. Curr. Drug. Metab., 2006, 7(4), 367-374.
E.; Davies, M.J.; Stein, P.P.; Kaufman, K.D.; Amatruda, J.M.; Wil- [108] Linnebjerg, H.; Kothare, P.A.; Park, S.; Mace, K.; Reddy, S.;
liams-Hermann, D. Safety and efficacy of sitagliptin in patients Mitchell, M.; Lins, R. Effect of renal impairment on the pharma-
with type 2 diabetes and chronic renal insufficiency. Diabetes cokinetics of exenatide. Br. J. Clin. Pharmacol., 2007, 64(3), 317-
Obes. Metab., 2008, 10(7), 545-555. 327.
[89] Henness, S.; Keam, S.J. Vildagliptin. Drugs, 2006, 66(15), 1989- [109] Byetta (exenatide injection) Prescribing information. Available at
2001. http://pi.lilly.com/us/byetta-pi.pdf
[90] Croxtall, J.D.; Keam, S.J. Vildagliptin: a review of its use in the [110] Knudsen, L.B.; Nielsen, P.F.; Huusfeldt, P.O.; Johansen, N.L.;
management of type 2 diabetes mellitus. Drugs, 2008, 68(16), Madsen, K.; Pedersen, F.Z.; Thogersen, H.; Wilken, M.; Agerso, H.
2387-2409. Potent derivatives of glucagon-like peptide-1 with pharmacokinetic
Antidiabetic Agents in CKD and ESRD Current Drug Metabolism, 2011, Vol. 12, No. 1 69
properties suitable for once daily administration. J. Med. Chem., [122] Lutz, T.A.; Mollet, A.; Rushing, P.A.; Riediger, T.; Scharrer, E.
2000, 43(9), 1664-1669. The anorectic effect of a chronic peripheral infusion of amylin is
[111] Agerso, H.; Jensen, L.B.; Elbrond, B.; Rolan, P.; Zdravkovic, M. abolished in area postrema/nucleus of the solitary tract (AP/NTS)
The pharmacokinetics, pharmacodinamics, safety and tolerability lesioned rats. Int. J. Obes. Relat. Metab. Disord., 2001, 258(7),
of NN2211, a new long-acting GLP-1 derivative, in healthy men. 1005-1011.
Diabetologia, 2002, 45(2), 195-202. [123] Whitehouse, F.; Kruger, D.F.; Fineman, M.; Shen, L.; Ruggles,
[112] Malm-Erjefalt, M.; Bjornsdottir, I.; Vanggaard J.; Helleberg, H.; J.A.; Maggs, D.G.; Weyer, C.; Kolterman, O.G. A randomized
Larsen, U.; Oosterhuis, B.; van Lier, J.J.; Zdravkovic, M. Olsen, study and open-label extension evaluating the long-term efficacy of
A.K. Metabolism and excretion of the once daily human GLP-1 pramlinitide as an adjunct to insulin therapy in type 1 diabetes.
analog liraglutide in healthy males subjects and its in vitro degrada- Diabetes Care, 2002, 25(4), 724-730.
tion by dipeptidyl peptidase IV and neutral endopeptidase. Drug [124] Ratner, R.E.; Dickey, R.; Fineman, M.; Maggs, D.G.; Shen, L.;
Metab. Dispos., 2010, 38(11), 1944-1953. Strobel, S.A.; Weyer, C.; Kolterman, O.G. Amylin replacement
[113] Juhl, C.B.; Hollingdal, M.; Sturis, J.; Jakobsen, G.; Agerso, H.; with pramlinitide as an adjunct to insulin therapy improves long-
Veldhuis, J.; Porksen, N.; Schmitz, O. Bedtime administration of term glycaemic and weight control in type 1 diabetes mellitus: a 1-
NN2211, a long-acting GLP-1 derivative, substantially reduces year randomized controlled trial. Diabet. Med., 2004, 21(11), 1204-
fasting and postprandial glycemia in type 2 diabetes. Diabetes, 1212.
2002, 51(2), 424-429. [125] Chapman, I.; Parker, B.; Doran, S.; Feinle-Bisset, C.; Wishart, J.;
[114] Chang, A.M.; Jakobsen, G.; Sturis, J.; Smith, M.J.; Bloem, C.I.; Strobel, S.; Wang, Y.; Burns, C.; Lush, C.; Weyer, C.; Horowitz,
Galecki, A.; Halter, J.B. The GLP-1 derivative NN2211 restores M. Effect of pramlinitide on satiety and food intake in obese sub-
beta-cell sensitivity to glucose in type 2 diabetic patients after a jects and subjects with type 2 diabetes. Diabetologia, 2005, 48(5),
single dose. Diabetes, 2003, 52(7), 1786-1791. 838-848.
[115] Degn, K.B.; Juhl, C.B.; Sturis, J.; Jakobsen, G.; Brock, B.; [126] Hollander, P.A.; Levy, P.; Fineman, M.S.; Maggs, D.G.; Shen,
Chandramouli, V.; Rungby, J.; Landau, B.R.; Schmitz, O. One L.Z.; Strobel, S.A.; Weyer, C.; Kolterman, O.G. Pramlinitide as an
week’s treatment with the long-acting GLP-1 derivative, liraglutide adjunct to insulin therapy improves long-term glycemic and weight
(NN2211), markedly improves 24-h glycaemia, alpha- and beta- control in patients with type 2 diabetes: a 1-year randomized con-
cell function and reduces endogenous glucose release in patients trolled trial. Diabetes Care, 2003, 26(3), 784-790.
with type 2 diabetes. Diabetes, 2004, 53(5), 1187-1194. [127] Riddle, M.; Brown, C.; Frias, J.; Lutz, K.; Zhang, B.; Kolterman,
[116] Vilsboll, T.; Brock, B.; Perrild, H.; Levin, K.; Lervang, H.H.; O.; Maier, H. Pramlintide improved glycemic control and reduced
Kolendorf, K.; Krarup, T.; Schmitz, O.; Zdravkovic, M.; Le-Thi, weight in patients with type 2 diabetes using basal insulin. Diabetes
T.; Madsbad, S. Liraglutide, a once-daily human GLP-1 analogue, Care, 2007, 30(11), 2794-2799.
improves pancreatic -cell function and arginine-stimulated insulin [128] Available from URL: http://documents.symlin.com/ SYM-
secretion during hyperglycemia in patients with type 2 diabetes LIN_PI.pd (Accessed Jan 15, 2011).
mellitus. Diabet. Med., 2008, 25(2), 152-156. [129] 2007 American Diabetes Association. Standards of Medical Care in
[117] Courreges, J.P.; Zdravkovic, M.; Vilsboll, T.; Zdravkovic, M.; Le- Diabetes-2008. Diabetes Care, 2008, 31, S12-S54.
thi, T.; Krarup, T.; Schmitz, O.; Verhoeven. R.; Buganova, I.; [130] Inaba, M.; Okuno, S.; Kumeda, Y.; Yamada, S.; Imanishi, Y.;
Madsbad, S. Liraglutide, a once-daily human glucagon-like pep- Tabata T.; Okamura M.; Okada, S.; Yamakawa T.; Ishimura, E.;
tide-1 analogue, shows beneficial effects on cardiovascular risk Nishizawa, Y. The Osaka CKD Expert Research Group. Glycated
biomarkers in patients with type 2 diabetes. Diabet. Med., 2008, albumin is a better glycemic indicator than glycated hemoglobin
25(9), 1129-1131. values in hemodialysis patients with diabetes: effect of anemia and
[118] Jacobsen, L.V.; Hindsberger, C.; Robson, R.; Zdravkovic, M. Ef- erythropoietin injection. J. Am. Soc. Nephrol., 2007, 18(3), 896-
fect of renal impairment on the pharmacokinetics of the GLP-1 903.
analogue liraglutide. Br. J. Clin. Pharmacol., 2009, 68(6), 898-905. [131] Peacock, T.P.; Shihabi, Z.K.; Bleyer, A.J.; Dolbare, E.L.; Byers,
[119] Young, A. Inhibition of glucagon secretion. Adv. Pharmacol., J.R.; Knovich, M.A.; Calles-Escandon, J.; Russell, G.B.; Freedman,
2005, 52, 151-171. B.I. Comparison of glycated albumin and hemoglobin A1c levels in
[120] Young, A.A.; Gedulin, B.; Vine, W.; Percy, A.; Rink, T.J. Gstric diabetic subjects on hemodialysis. Kidney Int., 2008, 73(9), 1062-
emptying is accelerated in diabetic BB rats and is slowed by subcu- 1068.
taneous injections of amylin. Diabetologia, 1995, 38(6), 642-648. [132] Abe, M.; Matsumoto, K. Glycated hemoglobin or glycated albumin
[121] Gedulin, B.R.; Rink, T.J.; Young, A.A. Doseresponse for gluca- for assessment of glycemic control in dialysis patients with diabe-
gonostatic effect of amylin in rats. Metabolism, 1997, 46(1), 67-70. tes? Nat. Clin. Pract. Nephrol., 2008, 4(9), 482-483.
Received: October 12, 2010 Revised: January 12, 2011 Accepted: January 14, 2011