Vous êtes sur la page 1sur 6

IMPROVING OUTCOMES FOR DIABETIC PATIENTS ON DIALYSIS

Selection and Dosing of Medications for Management of Diabetes in Patients with Advanced Kidney Disease
James B. Reilly and Jeffrey S. Berns
Renal-Electrolyte and Hypertension Division, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania

ABSTRACT Diabetes mellitus is a leading cause of kidney disease worldwide. A large and expanding array of treatments for diabetes is available to improve glycemic control, including newer classes of drugs, such as thiazolidinediones and incretin-based therapies. The presence of impaired kidney function with reduced glomerular ltration rate should inuence choices, dosing, and monitoring of hypoglycemic agents, as some agents require a dosing adjustment in patients with kidney disease and some are entirely contraindicated. This article reviews the clinical use of insulin and other antidiabetic therapies, focusing on pharmacokinetic properties and dosing in patients with advanced kidney disease.

Diabetes mellitus remains a leading cause of kidney disease and end-stage renal disease (ESRD) in the United States and around the world. Avoidance of hyperglycemia has been shown in many studies to slow progression of diabetic chronic kidney disease (CKD) and also to reduce microvascular and macrovascular morbidity and mortality (13). The importance of tight glucose control and identifying optimal targets in patients who already have advanced kidney disease is somewhat controversial, but observational data show that poor glycemic control is associated with unfavorable outcomes in patients with kidney disease (4,5). Treatment of hyperglycemia remains greatly important to the health of patients with kidney disease, and the arsenal of therapy available to physicians is expanding constantly. However, effective, safe glycemic control requires adjustment of medication regimens appropriate to the patients level of kidney function, as the pharmacokinetic and safety proles of many of these therapies change as kidney function declines over time. This review will describe the clinical use and pharmacokinetic proles of major diabetic therapies and identify preferred choices, dosing, and monitoring of agents for patients both with severe CKD and dialysis patients. Special attention will be paid to the newer agents, including thiazoladinediones (TZDs), meglinitide analogs, glucagon-like peptide-1 (GLP-1) analogs, and dipeptidyl peptidase-4 (DPP-4) inhibitors.
Address correspondence to: Jeffrey S. Berns, MD, Hospital of the University of Pennsylvania, 1 Founders Pavilion, 3400 Spruce Street, Philadelphia, PA 19104, Tel.: +1-215-662-7934, Fax: +1-215-615-1688, or e-mail: bernsj@uphs.upenn.edu. Seminars in DialysisVol 23, No 2 (MarchApril) 2010 pp. 163168 DOI: 10.1111/j.1525-139X.2010.00703.x 2010 Wiley Periodicals, Inc. 163

Insulins In contrast to endogenously secreted insulin, which is degraded by the liver, exogenous insulin is primarily eliminated by the kidneys. Insulin is freely ltered at the glomerulus, and then extensively reabsorbed in the proximal tubule. Filtered insulin is taken up across the apical membrane of the proximal tubular epithelial cells, and then enzymatically degraded into peptide fragments of various sizes, which are subsequently reabsorbed. There is also uptake and degradation of insulin by peritubular endothelial cells, with total renal clearance exceeding GFR (6). As renal failure progresses, peritubular insulin uptake increases, compensating for the decline in degradation of ltered insulin until the GFR decreases to less than approximately 20 ml min. With lower levels of GFR insulin clearance decreases further and overall requirements for exogenous insulin often decline (710). If this is not anticipated, then the risk of symptomatic hypoglycemia can increase. The pharmacokinetics of various insulin preparations has not been well studied in patients with varying degrees of renal dysfunction, and there are no absolute guidelines dening appropriate dosing adjustments of insulin that should be made based on the level of GFR. It is generally recommended that when the GFR decreases to between 10 and 50 ml min, the insulin dosage should be reduced by 25%, and when the GFR decreases to <10 ml min, the insulin dosage should be reduced by 50% from previous amounts (11,12). Once patients start dialysis, they often require less exogenous insulin as peripheral insulin resistance improves with initiation of dialysis (10,11). This may be counterbalanced by increased food intake as the anorexia of advanced CKD resolves with dialysis, so careful monitoring of blood glucose levels is important.

164

Reilly and Berns It is recommended that metformin be stopped 23 days before any anticipated surgery or administration of iodinated radiographic contrast media because of the risk of acute kidney injury, with resumption of the drug only after kidney function has clearly remained or returned to a normal baseline (20). Because of the risk of lactic acidosis, metformin should not be used in patients with serum creatinine levels 1.4 mg dl in women and 1.5 mg dl in men. Hemodialysis is effective in clearing metformin from the circulation (16) and has been used successfully for the management of metformin-associated lactic acidosis (21,22). There are also limited reports of use of continuous venovenous hemoltration for management of metformin-associated lactic acidosis (23,24). Alpha-Glucosidase Inhibitors Biguanides The alpha-glucosidase inhibitors acarbose and miglitol decrease postprandial hyperglycemia by blocking the intestinal breakdown of oligosaccharides into simpler sugars. Acarbose is only minimally absorbed from the gastrointestinal tract, where it is largely broken down by intestinal ora and intestinal enzymes into at least 13 metabolites, at least one of which has some biologic activity. Some of the metabolites are partially absorbed, although <2% of a dose of acarbose and the active metabolites appears in the urine. The elimination half-life is approximately 2 hours. In patients with reduced renal function, the plasma levels of the drug and its metabolites can increase several-fold, but the clinical signicance of this is not known. Information about the long-term use of acarbose in patients with reduced kidney function is sparse and its use in patients with later stage 3 and stages 4 and 5 CKD is not recommended (11). Miglitol has greater systemic absorption than acarbose, has minimal protein binding and is not metabolized. However, it does undergo renal excretion and accumulates in patients with impaired kidney function (11). Both acarbose and miglitol may reduce the absorption of other oral hypoglycemic agents (25). Miglitol is not recommended for patients with impaired kidney function (26). Sulfonylureas Sulfonylurea medications have long been a staple in the treatment of type 2 diabetes mellitus. They act primarily by stimulating insulin secretion of the pancreatic beta cells through interaction with a receptor that is a subunit of an adenosine triphosphate (ATP)-sensitive potassium channel, although it has been postulated that they may have additional extrapancreatic effects as well. These medications are well absorbed after oral administration and are highly protein bound in plasma. This protein binding can be reduced by other medications with higher protein-binding afnities, thus increasing free sulfonylurea plasma levels (26). The rst-generation sulfonylureas included acetohexamide, chlorpropamide, tolazamide, and tolbutamide. These drugs are now rarely used in the United

Since a reduced GFR increases the half-life and maximal serum concentration of regular human insulin, but not of rapid-acting analogs, rapid-acting insulin analogs are preferred over regular human insulin in advanced CKD, as the risk of hypoglycemia may be less. Caution is recommended when using long-acting insulin regimens but use of long-acting basal insulin like glargine is generally acceptable in patients with kidney disease and is preferred to use of NPH insulin (13). Insulin detemir binds to serum albumin after injection so its use may be less predictable in patients with nephrotic syndrome and hypoalbuminemia. Clearly it is important to monitor the patients blood glucose levels closely at multiple times of the day, particularly as the level of kidney function declines over time, and alter insulin regimens as required to avoid hypoglycemia.

Metformin and phenformin comprise the class of oral hypoglycemic known as the biguanides. Metformin has been widely prescribed for the treatment of type 2 diabetes mellitus for many years. Because of its association with a severe, and sometimes fatal, lactic acidosis, phenformin was removed from the market many years ago and its use will not be discussed further in this review. While metformin has been demonstrated to act as an agonist for the insulin receptor tyrosine kinase in some animal models, in humans it primarily inhibits hepatic gluconeogenesis by interfering with mitochondrial oxidation (14). Metformin also stimulates insulin-mediated glucose utilization by peripheral tissues. Used as monotherapy, or in combination with other diabetic medications, it has also been shown to have other physiologic cardiovascular effects beyond those related to control of blood glucose levels. Metformin is not protein bound and is eliminated unchanged by the kidneys through both glomerular ltration and tubular secretion, with an elimination half-life that has been reported to range from 1.5 to 8.7 hours (11,15). Approximately 90% of a dose is excreted within 1224 hours (16,17). Elimination of metformin is reduced in patients with reduced GFR. Cimetidine inhibits tubular secretion and overall clearance of metformin (16,18), and coadministration with metformin is not recommended. The most common side effects of metformin are gastrointestinal disturbances, including nausea, metallic taste, and diarrhea. A rare, but serious side effect of metformin is lactic acidosis (19). The estimated incidence of metformin-induced lactic acidosis, which has a mortality of approximately 3050%, is approximately 0.03 cases per 1000 patient-years, much less than that seen with phenformin (14). As GFR declines, accumulation of metformin is thought to increase the risk for development of lactic acidosis. Additional risk factors for metformin-induced lactic acidosis include congestive heart failure, advanced age, alcohol use, administration of iodinated radiographic contrast media (potentially causing acute kidney injury), and other conditions associated with hypoxemia or hypotension (15).

DIABETES MEDICATIONS IN CKD

165

States, and so we will not discuss them further. Second-generation sulfonylureas currently available in the United States include glipizide, glyburide, and glimepiride. The elimination half-life with chronic use of these newer agents ranges from 7 to 14 hours, with effective durations of action of up to 24 hours. Glipizide is hepatically metabolized into several inactive metabolites (11,17,27). Less than 10% of a dose of glipizide is excreted unchanged in the urine, with approximately 60% excreted as metabolites. Glipizide clearance and elimination half-life, which is 24 hours in normal individuals, are unaffected by reduced GFR and therefore dose adjustment in patients with CKD is not necessary (28). Several of the metabolites of glipizide accumulate in patients with reduced GFR, including one that may have a small amount of activity, although this does not appear to cause an increased risk of hypoglycemia (28). Because of its favorable pharmacokinetics in patients with reduced GFR, glipizide should be the sulfonylurea of choice in patients with advanced kidney dysfunction. Caution should be used with the extended release form of the drug, as few data are available regarding its safety in patients with CKD. Glyburide undergoes extensive hepatic metabolism to several metabolites that undergo equal biliary and renal excretion (11,17,29). Approximately 50% of a dose appears in the feces, with most of the remainder excreted as metabolites in the urine. Less than 5% of the parent compound is recovered in the urine. The clearance of glyburide does not correlate with the level of kidney function, and the parent drug does not appear to accumulate in patients with reduced GFR (30), although the elimination half-life and insulin area under the curve (AUC) may be slightly greater in hemodialysis patients than normal controls (31). Some of glyburides metabolites have hypoglycemic activity (30,32), which can increase the likelihood of hypoglycemia in patients with renal failure (3335). Therefore, glyburide should be used with great caution in these patients, especially considering that glipizide has a more favorable pharmacokinetic prole in patients with CKD. Glimepiride is also hepatically metabolized, with two major metabolites, one of which has been shown to have some pharmacologic activity. The elimination half-life is approximately 58 hours. Approximately 60% of a dose appears in the urine, with the remainder appearing in feces. Virtually all of the urinary excretion is as metabolites, with little or no parent drug excreted in the urine (36). Glimepiride does not accumulate in patients with reduced GFR, but urinary excretion of its metabolites is reduced. Prolonged hypoglycemia has been reported with the use of glimepiride in patients with reduced GFR (11,35,37), so the drug should be used very cautiously if at all in such patients. Thiazolidinediones The TZDs rosiglitazone and pioglitazone, as well as troglitazone, an earlier drug removed from the market for hepatotoxicity, act on the peroxisome proliferator-activated receptor-c (PPAR-c) and decrease

insulin resistance. They also increase the peripheral utilization of insulin and decrease hepatic gluconeogenesis (26). Because PPAR-c is expressed in multiple areas of the kidney, there have been many postulated potential benets of these agents in the CKD and ESRD populations, related mainly to their hemodynamic, antiproliferative, and anti-inammatory effects in diseases such as diabetic nephropathy (38). Despite much early promise, the long-term benets of these drugs are still controversial, although their use remains widespread, including in CKD and ESRD patients. Both rosiglitazone and pioglitazone are virtually completely hepatically metabolized, each forming several metabolites. Both drugs are also highly protein bound, primarily to albumin. Rosiglitazone has an elimination half-life of 34 hours. There are two major metabolites, neither of which is biologically active, and <1% of the parent drug appears in the urine unchanged (11). Pioglitazone has an elimination half-life of approximately 37 hours and six metabolites, of which three are active (11). Neither parent compound nor their active metabolites accumulate in patients with reduced GFR (3941). In fact, with both drugs, the AUC is less in patients with severe renal failure, perhaps because of reduced protein binding. Hemodialysis does not affect the pharmacokinetics of these drugs. In continuous ambulatory peritoneal dialysis (CAPD) patients, the half life of rosiglitazone is increased, but the AUC and average maximum plasma concentration (Cmax) is unchanged compared with normal controls (42). It is therefore not necessary to reduce the dosages of pioglitazone and rosiglitazone in patients with reduced GFR (11,26). Although there are no clinically signicant dosing issues for thiazolidinediones in patients with moderate or severe kidney dysfunction, their safety and tolerability proles raise some issues for CKD and dialysis patients. In particular, the side effect most worrisome to nephrologists is uid retention, which most often manifests as edema and congestive heart failure (43). In a subset of patients, expansion of plasma volume occurs through mechanisms that are likely multifactorial. Increased sodium reabsorption in the collecting duct is considered to be the primary renal site of increased sodium reabsorption with TZDs, with experimental evidence both for and against a role of TZDs on function of the distal nephron renal epithelial sodium channel (ENaC) as well as transport proteins in the proximal tubule (4448). TZDs additionally increase sympathetic nervous activity and alter endothelial permeability (49,50) which may also contribute signicantly to uid retention. To what extent this particular side effect of the TZDs is clinically important in patients with advanced CKD, and especially in dialysis patients is unclear, although several studies have suggested that some hemodialysis and peritoneal dialysis patients may experience weight gain with TZD therapy (5153). Effects of TZDs on cardiovascular morbidity and mortality have been somewhat controversial, with oftenconicting study results. A 2007 meta-analysis found that use of rosiglitazone was associated with a signicant increase in risk of myocardial infarction and a trend toward greater risk of cardiovascular death (54). This

166

Reilly and Berns inhibitors are the most recently approved diabetic treatments. Incretin hormones, such as GLP-1, are released after meals and serve to stimulate pancreatic insulin secretion, temper glucagon release, and slow gastric emptying (6769). Exenatide, the rst GLP-1 analog used in clinical practice, is an injectable therapy derived from the saliva of the Gila monster that has been shown to be effective in conjunction with more conventional oral therapies to improve glycemic control, particularly postprandial hyperglycemia (70,71). Exenatide reaches peak concentrations within 2 hours of administration, and is predominantly eliminated by the kidneys, with a half life of 2.5 hours in patients with normal renal function (72,73). The elimination half-life increases to 3.2 hours in patients with moderately reduced GFR (3150 ml min) and up to 6 hours in patients with GFR < 30 ml min or ESRD, associated with an increase in AUC with potentially toxic blood levels detected (72). For this reason, it is recommended that treatment be started with a 50% dose reduction (5 mcg twice daily) in patients with creatinine clearance 3050 ml min, titrating to full dose of 10 mcg twice daily only if tolerated and necessary for glycemic control. Exenatide is not recommended if creatinine clearance is <30 ml min. Liraglutide, a new injectable GLP-1 analog approved in Europe but currently pending FDA approval in the United States, may be safer in severe CKD and ESRD, as reduced GFR does not alter its metabolism or excretion (70,74,75). DPP-4 Inhibitors Two DPP-4 inhibitors, sitagliptin and saxagliptin, are currently FDA approved for treatment of patients with Type 2 diabetes mellitus. DPP-4 is an enzyme responsible for degradation of GLP-1, and antagonism of this enzyme has been postulated to potentiate incretin effects and reduce hyperglycemia, especially after meals. Sitagliptin is an oral, once-daily DPP-4 inhibitor that is primarily cleared by the kidneys. In normal, healthy subjects, 7580% of the drug is excreted unchanged in the urine (76). In patients with moderate kidney disease (creatinine clearances 30 50 ml min), the AUC increases more than twofold, and in patients with more severe kidney disease, including ESRD, the AUC increases by approximately fourfold. The drug is not effectively dialyzed (77). Based on these ndings, a 50% dose reduction (50 mg daily rather than the usual dose of 100 mg) is recommended in patients with moderate kidney disease, and a 75% dose reduction is recommended in patients with more advanced kidney disease and those on dialysis (77,78). Saxagliptin is another orally administered, once-daily DPP-4 recently approved by the FDA (79). Although the drug is primarily dependent upon the cytochrome P450 3A4 5 (CYP3A4 5) system for its metabolism, studies in animals indicate that renal metabolism and excretion contribute signicantly to elimination of saxagliptin (80). The major metabolite of saxagliptin is

association has not been consistently supported in later studies, however, and there are few data specically addressing use in patients with kidney disease. Recent studies in CKD and ESRD patients have likewise been contradictory. A cohort study of over 2300 hemodialysis patients found that use of rosiglitazone was associated with signicantly higher risk for cardiovascular and allcause mortality compared to other agents (55). However, another cohort study of over 5000 incident dialysis patients with diabetes found that TZD use was associated with better overall survival in non-insulin-dependent diabetic dialysis patients (56). Further study of TZD use in patients with CKD and ESRD, ideally in a randomized trial, is warranted. In the meantime, TZDs can be used safely without need for dose adjustment in patients with kidney disease, with close monitoring for clinical evidence of volume overload and CHF. These agents should be used with great caution in patients with increased risk for CHF, and are contraindicated in patients with NYHA Class III or IV heart failure, in accordance with consensus guidelines from the American Heart Association and the American Diabetes Association and a black box warning in the package labeling (57). Meglinitides Repaglinide and nateglinide are insulin secretagogues indicated for the treatment of type 2 diabetes mellitus. They act by stimulating insulin secretion of the pancreatic beta cell through effects on beta-cell K+-ATP channels, but through a different mechanism than the sulfonylureas (58). These drugs have shorter onset and duration of action compared with the sulfonylureas. Repaglinide is rapidly absorbed following oral administration, and therefore should be taken just before meals. It is primarily hepatically metabolized, with an elimination half-life of 0.61.8 hours (58,59). Less than 10% of a dose appears in the urine, but drug concentration, AUC, and elimination half-life are increased somewhat in patients with reduced GFR (11,60,61), although dosage reductions are not necessary for such patients (62). Nateglinide is also rapidly absorbed and eliminated primarily by hepatic metabolism, with an elimination half-life of approximately 1.21.8 hours. Several of its metabolites are active. Eighty-three percent of the drug appears in the urine, primarily as metabolites, with only approximately 16% as parent drug (58,63). Although reduced GFR does not signicantly increase elimination half-life or plasma concentration of nateglinide (64), plasma protein binding is reduced and accumulation of an active metabolite can occur; this has been associated with development of hypoglycemia (65,66). Therefore, nateglinide should be used very cautiously if at all in patients with advanced kidney disease. GLP-1 Analogues Incretin-based therapies with glucagon-like peptide1(GLP-1) analogues and dipeptidyl peptidase-4 (DPP-4)

DIABETES MEDICATIONS IN CKD

167

also a biologically active DPP-4 inhibitor. In humans, approximately 25% of a single dose is excreted unchanged in the urine and more than one-third is excreted as the active metabolite. Renal clearance of saxagliptin exceeds GFR in normal subjects, suggesting a component of renal tubular secretion. In a small single-dose study, reduced GFR was associated with an increase in AUC for both saxagliptin and its active metabolite (81). The manufacturer recommends a dose of 2.5 mg once daily for patients with creatinine clearance 50 ml min, including those on dialysis. Saxagliptin and its active metabolite are partially removed by hemodialysis (81). Another DPP-4 inhibitor, vildaglipitin, has been recently reported to be associated with an increased risk of angioedema in patients also receiving ACE inhibitors, suggesting the need for some caution in such patients (82,83). Conclusion In the past several years, a great number of novel diabetic therapies in different categories have been introduced, with the promise of more to come. Given the concurrence of kidney disease in many diabetics, available therapies may be dictated by the patients baseline kidney function and how it evolves over time. Some agents that are used as rst line treatment in patients with normal kidney function, such as metformin, are completely contraindicated in renal failure, while others can be used safely, some with dose adjustments and some at full strength. Most of the commonly used classes of diabetic therapies contain at least one medication that is safe in kidney disease, and these medications should be utilized preferentially in patients who have kidney disease or those who are likely to develop kidney disease in the future. New medications should be evaluated with an eye to potential efcacy and safety in patients with reduced GFR as they are introduced into clinical practice. References
1. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The Diabetes Control and Complications Trial Research Group. N Engl J Med 329:977986, 1993 2. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes Study (UKPDS) Group. Lancet 352:837853, 1998 3. Ohkubo Y, Kishikawa H, Araki E, Miyata T, Isami S, Motoyoshi S, Kojima Y, Furuyoshi N, Shichiri M: Intensive insulin therapy prevents the progression of diabetic microvascular complications in Japanese patients with noninsulin-dependent diabetes mellitus: a randomized prospective 6-year study. Diabetes Res Clin Pract 28:103117, 1995 4. Oomichi T, Emoto M, Tabata T, Morioka T, Tsujimoto Y, Tahara H, Shoji T, Nishizawa Y: Impact of glycemic control on survival of diabetic patients on chronic regular hemodialysis: a 7-year observational study. Diabetes Care 29:14961500, 2006 5. Morioka T, Emoto M, Tabata T, Shoji T, Tahara H, Kishimoto H, Ishimura E, Nishizawa Y: Glycemic control is a predictor of survival for diabetic patients on hemodialysis. Diabetes Care 24:909913, 2001 6. Rabkin R, Ryan MP, Duckworth WC: The renal metabolism of insulin. Diabetologia 27:351357, 1984 7. Rubenstein AH, Mako ME, Horwitz DL: Insulin and the kidney. Nephron 15:306326, 1975

8. Rave K, Heise T, Pfutzner A, Heinemann L, Sawicki PT: Impact of diabetic nephropathy on pharmacodynamic and pharmacokinetic properties of insulin in type 1 diabetic patients. Diabetes Care 24:886890, 2001 9. Biesenbach G, Raml A, Schmekal B, Eichbauer-Sturm G: Decreased insulin requirement in relation to GFR in nephropathic Type 1 and insulin-treated Type 2 diabetic patients. Diabet Med 20:642645, 2003 10. Mak RH: Impact of end-stage renal disease and dialysis on glycemic control. Semin Dial 13:48, 2000 11. Charpentier G, Riveline JP, Varroud-Vial M: Management of drugs affecting blood glucose in diabetic patients with renal failure. Diabetes Metab 26(Suppl. 4):7385, 2000 12. Aronoff G, Berns JS, Brier ME, Kasbekar N, Mueller BA, Pasko DA, Smoyer WE: Drug Prescribing in Renal Failure: Dosing Guidelines for Adults and Children, 5th edn. Philadelphia: American College of Physicians, 2007 13. Lubowsky ND, Siegel R, Pittas AG: Management of glycemia in patients with diabetes mellitus and CKD. Am J Kidney Dis 50:865879, 2007 14. Kirpichnikov D, McFarlane SI, Sowers JR: Metformin: an update. Ann Intern Med 137:2533, 2002 15. Bailey CJ, Turner RC: Metformin. N Engl J Med 334:574579, 1996 16. Scheen AJ: Clinical pharmacokinetics of metformin. Clin Pharmacokinet 30:359371, 1996 17. Harrower AD: Pharmacokinetics of oral antihyperglycaemic agents in patients with renal insufciency. Clin Pharmacokinet 31:111119, 1996 18. Somogyi A, Stockley C, Keal J, Rolan P, Bochner F: Reduction of metformin renal tubular secretion by cimetidine in man. Br J Clin Pharmacol 23:545551, 1987 19. Misbin RI, Green L, Stadel BV, Gueriguian JL, Gubbi A, Fleming GA: Lactic acidosis in patients with diabetes treated with metformin. N Engl J Med 338:265266, 1998 20. Heupler FA Jr: Guidelines for performing angiography in patients taking metformin. Members of the Laboratory Performance Standards Committee of the Society for Cardiac Angiography and Interventions. Cathet Cardiovasc Diagn 43:121123, 1998 21. Lalau JD, Andrejak M, Moriniere P, Coevoet B, Debussche X, Westeel PF, Fournier A, Quichaud J: Hemodialysis in the treatment of lactic acidosis in diabetics treated by metformin: a study of metformin elimination. Int J Clin Pharmacol Ther Toxicol 27:285288, 1989 22. Seidowsky A, Nseir S, Houdret N, Fourrier F: Metformin-associated lactic acidosis: a prognostic and therapeutic study. Crit Care Med 37:21912196, 2009 23. Arroyo AM, Walroth TA, Mowry JB, Kao LW: The MALAdy of metformin poisoning: is CVVH the cure? Am J Ther, in press 24. Bruijstens LA, van Luin M, Buscher-Jungerhans PM, Bosch FH: Reality of severe metformin-induced lactic acidosis in the absence of chronic renal impairment. Neth J Med 66:185190, 2008 25. Scheen AJ, Lefebvre PJ: Potential pharmacokinetics interference between alpha-glucosidase inhibitors and other oral antidiabetic agents. Diabetes Care 25:247248, 2002 26. Snyder RW, Berns JS: Use of insulin and oral hypoglycemic medications in patients with diabetes mellitus and advanced kidney disease. Semin Dial 17:365370, 2004 27. Gerich JE: Oral hypoglycemic agents. N Engl J Med 321:12311245, 1989 28. Balant L, Zahnd G, Gorgia A, Schwarz R, Fabre J: Pharmacokinetics of glipizide in man: inuence of renal insufciency. Diabetologia 9 (Suppl 1): 331338, 1973 29. Feldman JM: Glyburide: a second-generation sulfonylurea hypoglycemic agent. History, chemistry, metabolism, pharmacokinetics, clinical use and adverse effects. Pharmacotherapy 5:4362, 1985 30. Pearson JG, Antal EJ, Raehl CL, Gorsch HK, Craig WA, Albert KS, Welling PG: Pharmacokinetic disposition of 14C-glyburide in patients with varying renal function. Clin Pharmacol Ther 39:318324, 1986 31. Brier ME, Bays H, Sloan R, Stalker DJ, Welshman I, Aronoff GR: Pharmacokinetics of oral glyburide in subjects with non-insulin-dependent diabetes mellitus and renal failure. Am J Kidney Dis 29:907911, 1997 32. Jonsson A, Rydberg T, Sterner G, Melander A: Pharmacokinetics of glibenclamide and its metabolites in diabetic patients with impaired renal function. Eur J Clin Pharmacol 53:429435, 1998 33. Rydberg T, Jonsson A, Roder M, Melander A: Hypoglycemic activity of glyburide (glibenclamide) metabolites in humans. Diabetes Care 17:1026 1030, 1994 34. Krepinsky J, Ingram AJ, Clase CM: Prolonged sulfonylurea-induced hypoglycemia in diabetic patients with end-stage renal disease. Am J Kidney Dis 35:500505, 2000 35. Holstein A, Plaschke A, Hammer C, Egberts EH: Characteristics and time course of severe glimepiride- versus glibenclamide-induced hypoglycaemia. Eur J Clin Pharmacol 59:9197, 2003 36. Rosenkranz B, Profozic V, Metelko Z, Mrzljak V, Lange C, Malerczyk V: Pharmacokinetics and safety of glimepiride at clinically effective doses in diabetic patients with renal impairment. Diabetologia 39:16171624, 1996 37. Holstein A, Egberts EH: Risk of hypoglycaemia with oral antidiabetic agents in patients with Type 2 diabetes. Exp Clin Endocrinol Diabetes 111:405414, 2003 38. Iglesias P, Diez JJ: Peroxisome proliferator-activated receptor gamma agonists in renal disease. Eur J Endocrinol 154:613621, 2006

168

Reilly and Berns


62. 63. with type 2 diabetes and renal impairment. Eur J Clin Pharmacol 57:147 152, 2001 Hasslacher C: Safety and efcacy of repaglinide in type 2 diabetic patients with and without impaired renal function. Diabetes Care 26:886891, 2003 Weaver ML, Orwig BA, Rodriguez LC, Graham ED, Chin JA, Shapiro MJ, McLeod JF, Mangold JB: Pharmacokinetics and metabolism of nateglinide in humans. Drug Metab Dispos 29:415421, 2001 Devineni D, Walter YH, Smith HT, Lee JS, Prasad P, McLeod JF: Pharmacokinetics of nateglinide in renally impaired diabetic patients. J Clin Pharmacol 43:163170, 2003 Inoue T, Shibahara N, Miyagawa K, Itahana R, Izumi M, Nakanishi T, Takamitsu Y: Pharmacokinetics of nateglinide and its metabolites in subjects with type 2 diabetes mellitus and renal failure. Clin Nephrol 60:9095, 2003 Nagai T, Imamura M, Iizuka K, Mori M: Hypoglycemia due to nateglinide administration in diabetic patient with chronic renal failure. Diabetes Res Clin Pract 59:191194, 2003 Unger RH: Reinventing type 2 diabetes: pathogenesis, treatment, and prevention. JAMA 299:11851187, 2008 Gromada J, Brock B, Schmitz O, Rorsman P: Glucagon-like peptide-1: regulation of insulin secretion and therapeutic potential. Basic Clin Pharmacol Toxicol 95:252262, 2004 Gromada J, Rorsman P: New insights into the regulation of glucagon secretion by glucagon-like peptide-1. Horm Metab Res 36:822829, 2004 Kendall DM, Cuddihy RM, Bergenstal RM: Clinical application of incretin-based therapy: therapeutic potential, patient selection and clinical use. Am J Med 122:S37S50, 2009 Bray GM: Exenatide. Am J Health Syst Pharm 63:411418, 2006 Linnebjerg H, Kothare PA, Park S, Mace K, Reddy S, Mitchell M, Lins R: Effect of renal impairment on the pharmacokinetics of exenatide. Br J Clin Pharmacol 64:317327, 2007 Copley K, McCowen K, Hiles R, Nielsen LL, Young A, Parkes DG: Investigation of exenatide elimination and its in vivo and in vitro degradation. Curr Drug Metab 7:367374, 2006 Buse JB, Rosenstock J, Sesti G, Schmidt WE, Montanya E, Brett JH, Zychma M, Blonde L: Liraglutide once a day versus exenatide twice a day for type 2 diabetes: a 26-week randomised, parallel-group, multinational, open-label trial (LEAD-6). Lancet 374:3947, 2009 Elbrond B, Jakobsen G, Larsen S, Agerso H, Jensen LB, Rolan P, Sturis J, Hatorp V, Zdravkovic M: Pharmacokinetics, pharmacodynamics, safety, and tolerability of a single-dose of NN2211, a long-acting glucagon-like peptide 1 derivative, in healthy male subjects. Diabetes Care 25:13981404, 2002 Herman GA, Stevens C, Van Dyck K, Bergman A, Yi B, De Smet M, Snyder K, Hilliard D, Tanen M, Tanaka W, Wang AQ, Zeng W, Musson D, Winchell G, Davies MJ, Ramael S, Gottesdiener KM, Wagner JA: Pharmacokinetics and pharmacodynamics of sitagliptin, an inhibitor of dipeptidyl peptidase IV, in healthy subjects: results from two randomized, double-blind, placebo-controlled studies with single oral doses. Clin Pharmacol Ther 78:675688, 2005 Bergman AJ, Cote J, Yi B, Marbury T, Swan SK, Smith W, Gottesdiener K, Wagner J, Herman GA: Effect of renal insufciency on the pharmacokinetics of sitagliptin, a dipeptidyl peptidase-4 inhibitor. Diabetes Care 30:1862 1864, 2007 Chan JC, Scott R, Arjona Ferreira JC, Sheng D, Gonzalez E, Davies MJ, Stein PP, Kaufman KD, Amatruda JM, Williams-Herman D: Safety and efcacy of sitagliptin in patients with type 2 diabetes and chronic renal insufciency. Diabetes Obes Metab 10:545555, 2008 Tahrani AA, Piya MK, Barnett AH: Saxagliptin: a new DPP-4 inhibitor for the treatment of type 2 diabetes mellitus. Adv Ther 26:249262, 2009 Fura A, Khanna A, Vyas V, Koplowitz B, Chang SY, Caporuscio C, Boulton DW, Christopher LJ, Chadwick KD, Hamann LG, Humphreys WG, Kirby M: Pharmacokinetics of the dipeptidyl peptidase 4 inhibitor saxagliptin in rats, dogs, and monkeys and clinical projections. Drug Metab Dispos 37:11641171, 2009 Product package insert. Available at http://packageinserts.bms.com/pi/ pi_onglyza.pdf, accessed September 11, 2009 Brown NJ, Byiers S, Carr D, Maldonado M, Warner BA: Dipeptidyl peptidase-IV inhibitor use associated with increased risk of ACE inhibitorassociated angioedema. Hypertension 54:516523, 2009 Grouzmann E, Livio F, Buclin T: Angiotensin-converting enzyme and dipeptidyl peptidase IV inhibitors. An increased risk of angioedema. Hypertension 54:468470, 2009

39. Budde K, Neumayer HH, Fritsche L, Sulowicz W, Stompor T, Eckland D: The pharmacokinetics of pioglitazone in patients with impaired renal function. Br J Clin Pharmacol 55:368374, 2003 40. Chapelsky MC, Thompson-Culkin K, Miller AK, Sack M, Blum R, Freed MI: Pharmacokinetics of rosiglitazone in patients with varying degrees of renal insufciency. J Clin Pharmacol 43:252259, 2003 41. Thompson-Culkin K, Zussman B, Miller AK, Freed MI: Pharmacokinetics of rosiglitazone in patients with end-stage renal disease. J Int Med Res 30:391399, 2002 42. Aramwit P, Supasyndh O, Sriboonruang T: Pharmacokinetics of single-dose rosiglitazone in chronic ambulatory peritoneal dialysis patients. J Clin Pharm Ther 33:685690, 2008 43. Hollenberg NK: Considerations for management of uid dynamic issues associated with thiazolidinediones. Am J Med 115(Suppl. 8A):111S115S, 2003 44. Kiryluk K, Isom R: Thiazolidinediones and uid retention. Kidney Int 72:762768, 2007 45. Vallon V, Hummler E, Rieg T, Pochynyuk O, Bugaj V, Schroth J, Dechenes G, Rossier B, Cunard R, Stockand J: Thiazolidinedione-induced uid retention is independent of collecting duct alphaENaC activity. J Am Soc Nephrol 20:721729, 2009 46. Guan Y, Hao C, Cha DR, Rao R, Lu W, Kohan DE, Magnuson MA, Redha R, Zhang Y, Breyer MD: Thiazolidinediones expand body uid volume through PPARgamma stimulation of ENaC-mediated renal salt absorption. Nat Med 11:861866, 2005 47. Panchapakesan U, Pollock C, Saad S: Review article: importance of the kidney proximal tubular cells in thiazolidinedione-mediated sodium and water uptake. Nephrology (Carlton) 14:298301, 2009 48. Staels B: Fluid retention mediated by renal PPARgamma. Cell Metab 2:77 78, 2005 49. Rennings AJ, Smits P, Stewart MW, Tack CJ: Fluid retention and vascular effects of rosiglitazone in obese, insulin-resistant, nondiabetic subjects. Diabetes Care 29:581587, 2006 50. Walker AB, Naderali EK, Chattington PD, Buckingham RE, Williams G: Differential vasoactive effects of the insulin sensitizers rosiglitazone (BRL 49653) and troglitazone on human small arteries in vitro. Diabetes 47:810 814, 1998 51. Wong TY, Szeto CC, Chow KM, Leung CB, Lam CW, Li PK: Rosiglitazone reduces insulin requirement and C-reactive protein levels in type 2 diabetic patients receiving peritoneal dialysis. Am J Kidney Dis 46:713719, 2005 52. Manley HJ, Allcock NM: Thiazolidinedione safety and efcacy in ambulatory patients receiving hemodialysis. Pharmacotherapy 23:861865, 2003 53. Chiang CK, Ho TI, Peng YS, Hsu SP, Pai MF, Yang SY, Hung KY, Wu KD: Rosiglitazone in diabetes control in hemodialysis patients with and without viral hepatitis infection: effectiveness and side effects. Diabetes Care 30:37, 2007 54. Nissen SE, Wolski K: Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N Engl J Med 356:2457 2471, 2007 55. Ramirez SP, Albert JM, Blayney MJ, Tentori F, Goodkin DA, Wolfe RA, Young EW, Bailie GR, Pisoni RL, Port FK: Rosiglitazone is associated with mortality in chronic hemodialysis patients. J Am Soc Nephrol 20:10941101, 2009 56. Brunelli SM, Thadhani R, Ikizler TA, Feldman HI: Thiazolidinedione use is associated with better survival in hemodialysis patients with non-insulin dependent diabetes. Kidney Int 75:961968, 2009 57. Nesto RW, Bell D, Bonow RO, Fonseca V, Grundy SM, Horton ES, Le Winter M, Porte D, Semenkovich CF, Smith S, Young LH, Kahn R: Thiazolidinedione use, uid retention, and congestive heart failure: a consensus statement from the American Heart Association and American Diabetes Association. Circulation 108:29412948, 2003 58. Dornhorst A: Insulinotropic meglitinide analogues. Lancet 358:17091716, 2001 59. Hatorp V: Clinical pharmacokinetics and pharmacodynamics of repaglinide. Clin Pharmacokinet 41:471483, 2002 60. Marbury TC, Ruckle JL, Hatorp V, Andersen MP, Nielsen KK, Huang WC, Strange P: Pharmacokinetics of repaglinide in subjects with renal impairment. Clin Pharmacol Ther 67:715, 2000 61. Schumacher S, Abbasi I, Weise D, Hatorp V, Sattler K, Sieber J, Hasslacher C: Single- and multiple-dose pharmacokinetics of repaglinide in patients

64.

65.

66.

67. 68.

69. 70.

71. 72.

73.

74.

75.

76.

77.

78.

79. 80.

81. 82.

83.

Vous aimerez peut-être aussi