What hypertension drug is the first choice for diabetic and renal failure patients?

  • PDFView PDF

What hypertension drug is the first choice for diabetic and renal failure patients?

What hypertension drug is the first choice for diabetic and renal failure patients?

Under a Creative Commons license

Open access

Diabetic kidney disease (DKD) accounts for >40% cases of chronic kidney disease (CKD) globally. Hypertension is a major risk factor for progression of DKD and the high incidence of cardiovascular disease and mortality in these people. Meticulous management of hypertension is therefore crucial to slow down the progression of DKD and reduce cardiovascular risk. Randomized controlled trial evidence differs in type 1 and type 2 diabetes and in different stages of DKD in terms of target blood pressure (BP). Renin-angiotensin blocking agents reduce progression of DKD and cardiovascular events in both type 1 and type 2 diabetes, albeit differently according to the stage of CKD. There is emerging evidence for the benefit of sodium glucose cotransporter 2, nonsteroidal selective mineralocorticoid antagonists, and endothelin-A receptor antagonists in slowing progression and reducing cardiovascular events in DKD. This UK guideline, developed jointly by diabetologists and nephrologists, has reviewed all available current evidence regarding the management of hypertension in DKD to produce a set of comprehensive individualized recommendations for BP control and the use of antihypertensive agents according to age, type of diabetes, and stage of CKD (https://ukkidney.org/sites/renal.org/files/Management-of-hypertension-and-RAAS-blockade-in-adults-with-DKD.pdf). A succinct summary of the guideline, including an infographic, is presented here.

Keywords

ACE inhibitors

angiotensin receptor blockers

chronic kidney disease

diabetes

dialysis

hypertension

Cited by (0)

© 2022 International Society of Nephrology. Published by Elsevier Inc.

Approach Considerations

Several issues are key in the medical care of patients with diabetic nephropathy. [4, 5] These include glycemic control, management of hypertension, and reducing dietary salt intake and phosphorus and potassium restriction in advanced cases.

A meta-analysis from the Cochrane Database shows a large fall in blood pressure with salt restriction, similar to that of single-drug therapy. [26] All diabetic patients should consider reducing salt intake at least to less than 5-6 g/d, in keeping with current recommendations for the general population, and may benefit from lowering salt intake to even lower levels. Reducing dietary salt intake may help slow progression of diabetic kidney disease. Renal replacement therapy may be necessary in patients with end-stage renal disease (ESRD).

A 2012 post-hoc analysis of the data merged from the Reduction of Endpoints in NIDDM with the Angiotensin II Antagonist Losartan (RENAAL) trial and the Irbesartan Diabetic Nephropathy Trial (IDNT) in 1177 patients demonstrated that a low-sodium diet (24-h urinary sodium/creatinine ratio (mmol/g) < 121) enhanced the renoprotective and cardioprotective effect of angiotensin receptor blockers (losartan or irbesartan) in type 2 diabetic patients with nephropathy. Compared with higher sodium intake groups, the patients in the low-sodium group had better renal (by 43%) and cardiovascular (by 37%) outcomes. These improved outcomes in the low-sodium group underscore the importance of recent calls for population-wide intervention to reduce dietary salt intake, particularly in patients with diabetes and nephropathy treated with angiotensin receptor blockers. [27]

Guidelines from the organization Kidney Disease: Improving Global Outcomes (KDIGO) look at diabetes management in the context of chronic kidney disease, including with regard to comprehensive care, glycemic monitoring and targets, interventions addressing lifestyle and hyperglycemia, and approaches to self-management and models for optimal care.

Glycemic Control

In persons with either type 1 or type 2 diabetes mellitus (DM), hyperglycemia has been shown to be a major determinant of the progression of diabetic nephropathy. The evidence is best reported for type 1 DM.

It has been shown that intensive therapy can partially reverse glomerular hypertrophy and hyperfiltration, delay the development of microalbuminuria, and stabilize or even reverse microalbuminuria.

Results from pancreatic transplant recipients in whom true euglycemia is restored suggest that strict glycemic and metabolic control may slow the progression rate of progressive renal injury even after overt dipstick-positive proteinuria has developed.

In the Diabetes Control and Complications Trial, reduction in microvascular complications was of a smaller magnitude in patients with type 2 DM receiving intensive insulin therapy than in patients with type 1 DM. [28] In an outcome and cost-effective analysis of the United Kingdom Prospective Diabetes Study (UKPDS), the authors concluded that intensive blood glucose control in patients with type 2 DM significantly increased treatment costs but substantially reduced the cost of complications and increased the time free of complications. [29]

Newer agents in diabetic patients with kidney disease

Dipeptidyl peptidase inhibitors

The dipeptidyl peptidase (DPP)–4 inhibitors (ie, gliptins) are a new class of antidiabetic agents that can be used in type 2 diabetes. These agents include sitagliptin, saxagliptin, linagliptin, and alogliptin, and they decrease the breakdown of the incretin hormones such as glucagonlike peptide 1 (GLP-1). GLP-1 is secreted by the GI tract in response to food intake and leads to insulin secretion in a glucose-dependent manner, while also decreasing glucagon release. GLP-1 also slows gastric emptying.

Sitagliptin was the first available DPP-4 inhibitor. Approximately 80% of sitagliptin is cleared by the kidney; therefore, the standard dose of 100 mg daily should be reduced in patients with reduced glomerular filtration rates (GFRs). With an estimated GFR (eGFR) of 30 or greater to less than 50 mL/min/1.73 m2, the recommended dose is 50 mg once daily, and with an eGFR less than 30 mL/min/1.73 m2, a dose of 25 mg once daily is advised. [30]

The starting dose for saxagliptin is 2.5-5 mg daily in patients with an eGFR greater than 50 mL/min, but dose adjustment is recommended in patients with an eGFR of 50 mL/min/1.73 m2 or less to 2.5 mg daily.

Alogliptin also requires a dose reduction from 25 mg daily to 12.5 mg daily in patients with an eGFR of less than 60 mL/min/1.73 m2 and to 6.25 mg daily if the eGFR is less than 30 mL/min/1.73 m2.

In contrast, only the kidney clears a small amount of linagliptin; thus, no dose adjustment is necessary in patients with a reduced GFR. [31]

Alpha-glucosidase inhibitors

Alpha-glucosidase inhibitors (acarbose, miglitol) decrease the breakdown of oligosaccharides and disaccharides in the small intestine, slowing the absorption of glucose after a meal. The major adverse effects are bloating, flatulence, and abdominal cramping.

Acarbose is minimally absorbed, with less than 2% of the drug and active metabolites present in the urine. However, in patients with reduced renal function, serum levels of acarbose and metabolites are significantly higher. Miglitol has greater systemic absorption with greater than 95% renal excretion. It is recommended that miglitol be avoided if the GFR is less than 25 mL/min/1.73 m2. [32] These drugs have not been studied in patients with advanced kidney disease, and their use should be avoided in this population.

Sodium-glucose cotransporter 2 (SGLT2) inhibitors

There are several SGLT2 inhibitors under development or consideration by the US Food and Drug Administration (FDA), and canagliflozin (Invokana) is the first SGLT2 inhibitor approved for use in the United States. Canagliflozin inhibits renal glucose absorption in the proximal tubule, the site in the kidney where approximately 90% of glucose reabsorption occurs. This leads to increased excretion of glucose in the urine, which may help subjects lose up to 5 kg of weight over a year. The decreased glucose reabsorption is also accompanied by increased urinary excretion of sodium, which, in turn, may help with further blood pressure lowering, which could be an advantage in patients with diabetic kidney disease and hypertension.

At this time, the maximum dose in patients with an eGFR of 45 to less than 60 mL/min/1.73 m2 is 100 mg once daily and it is not recommended in patients with an eGFR of less than 45 mL/min/1.73 m2. Increased glucosuria is believed to increase the risk of urinary tract infections, especially candidal infections, and more frequently in women. [33]

In April 2019, it was reported that the CREDENCE trial, a multicenter, randomized, double-blind, placebo-controlled study of 4401 patients, had shown that canagliflozin leads to a 30% reduction in the likelihood that patients with type 2 diabetes and chronic kidney disease will progress to end-stage kidney disease, and also significantly decreases the chances for major cardiovascular events. [34]

Glucagonlike peptide-1 (GLP-1) receptor agonistsor incretin mimetics

The GLP-1 agonists exenatide (Byetta) and liraglutide (Victoza) are also known as injectable incretin mimetics. These drugs enhance central satiety and reduce appetite, thus helping with weight loss. These drugs promote insulin release, delay glucagon release, and slow gastric emptying and are less likely to cause hypoglycemia. There is some concern, however, about safety, as they may cause pancreatitis. Exenatide clearance is GFR dependent and is reduced at low GFRs. [35]

Although disputed, there are cases of acute renal failure associated with exenatide, and it is recommended to be used with caution in patients with a GFR of 30-50 mL/min and not be used at all if the eGFR is less than 30 mL/min. Liraglutide is not metabolized by the kidney, and no dose adjustment is necessary in patients with a decreased GFR, including ESRD, although data in this population are limited. [36] Cases of acute renal failure and worsening of chronic renal impairment have been reported with liraglutide, and the manufacturer advises caution in initiating or increasing the dose in patients with kidney disease.

Amylin analogs

Amylin is a 37-amino acid peptide co-secreted by β cells with insulin and is deficient in diabetes. Its levels parallel insulin levels, and its actions are complementary to insulin in regulating plasma glucose concentration. Amylin slows gastric emptying, reduces postprandial glucagon, and can suppress appetite.

Pramlintide (Symlin) is the only available amylin analog; it is given as an injection along with insulin therapy at meals. Dose adjustments for pramlintide are not required in the presence of mild-to-moderate renal disease, but there are no data on its use in end-stage kidney disease. [37]

Management of Hypertension

In general, antihypertensive therapy, irrespective of the agent used, slows the development of diabetic glomerulopathy. Mogensen showed that antihypertensive treatment attenuates the rate of decline in renal function in patients who have type 1 DM, hypertension, and proteinuria. [38] This is particularly significant when lowering of systemic blood pressure is accompanied with concomitant lessening of glomerular capillary pressure.

Careful blood pressure control is needed to prevent the progression of diabetic nephropathy and other complications; however the optimal lower limit for systolic blood pressure is unclear. In the UKPDS, a 12% risk reduction in diabetic complications was found with each 10 mm Hg drop in systolic pressure, the lowest risk being associated with a systolic pressure below 120 mm Hg. [29]

Angiotensin-converting enzyme inhibitors

From a therapeutic standpoint, preventing the progression of kidney disease is better achieved with a nonglycemic intervention, such as treatment with angiotensin-converting enzyme (ACE) inhibitors, which confer superior long-term protection even in comparison with triple therapy with reserpine, hydralazine, and hydrochlorothiazide or a calcium channel blocker (nifedipine).

Long-term treatment with ACE inhibitors, usually combined with diuretics, reduces blood pressure and albuminuria and protects kidney function in patients with hypertension, type 1 DM, and nephropathy. Beneficial effects on kidney function have also been reported in patients with normotension, type 1 DM, and nephropathy.

ACE inhibition has been shown to delay the development of diabetic nephropathy. In the ACE inhibition arm of a large trial, only 7% of patients with microalbuminuria experienced progression to overt nephropathy; however, in the placebo-treated group, 21% of patients experienced progression to overt nephropathy. The beneficial effect of ACE inhibition on preventing progression from microalbuminuria to overt diabetic nephropathy is long-lasting (8 y) and is associated with the preservation of a normal glomerular filtration rate (GFR).

The impact of ACE inhibition in patients with microalbuminuric type 2 DM has also been evaluated. Treatment with an ACE inhibitor for 12 months has significantly reduced mean arterial blood pressure and the urinary albumin excretion rate in type 2 DM patients who have microalbuminuria.

In a study of normotensive patients with microalbuminuric type 2 DM who received enalapril or placebo for 5 years, 12% of those in the actively treated group experienced diabetic nephropathy, with a rate of decline in kidney function of 13%, and 42% of those in the placebo group experienced nephropathy.

Meta-analysis has shown that ACE inhibitors are superior to beta-blockers, diuretics, and calcium channel blockers in reducing urinary albumin excretion in normotensive and hypertensive type 1 and type 2 DM patients. This superiority is pronounced in the normotensive state, whereas it is diminished progressively with progressive blood pressure reduction. Reduced glomerular capillary hydraulic pressure in combination with diminished size- and charge-selective properties of the glomerular capillary membrane are the most likely mechanisms involved in the antiproteinuric effect of ACE inhibitors.

The antiproteinuric effect of ACE inhibition in patients with diabetic nephropathy varies considerably. Individual differences in the renin-angiotensin system (RAS) may influence this variation. A potential role may exist for an insertion/deletion polymorphism of the ACE gene on this early antiproteinuric responsiveness in young patients with hypertension and type 1 DM who have developed diabetic nephropathy.

In addition to beneficial cardiovascular effects, ACE inhibition has also been demonstrated to have a significant beneficial effect on the progression of diabetic retinopathy and on the development of proliferative retinopathy.

Angiotensin receptor blockers for renin-angiotensin system inhibition

RAS inhibition is effective in treating type 1 and type 2 diabetic nephropathy. [39] It is important to consider type 2 diabetic nephropathy separately from type 1, as there are significant differences between them. Both are characterized by the appearance of microalbuminuria, which leads to overt proteinuria and progressive loss of GFR. A series of renal biopsy samples from patients with type 2 DM and proteinuria revealed that a significant proportion of these patients had glomerular lesions other than the classic lesions associated with type 1 diabetic nephropathy.

ACE inhibitors reduce the risk of progression of overt type 1 diabetic nephropathy to end-stage renal disease (ESRD) and in type 1 patients with microalbuminuria to overt nephropathy. Although ACE inhibition improves glomerular permeability in patients with type 1 DM as assessed by dextran clearances, it does not do so in patients with type 2 DM. Furthermore, the superior effect of blockade of the RAS has been difficult to prove.

Two studies (the Reduction of Endpoints in NIDDM with the Angiotensin II Antagonist Losartan [RENAAL] Study and the Irbesartan Diabetic Nephropathy Trial [IDNT]) demonstrated that angiotensin II receptor blockers (ARBs) are superior to conventional therapy and amlodipine in slowing the progression of overt nephropathy.

These trials were performed with ARBs and not ACE inhibitors. This raised the question as to whether such beneficial results in patients with type 2 DM would be seen with ACE inhibitors as well. Unfortunately, a large head-to-head comparison of ACE inhibitors and ARBs is unlikely to be made.

The choice between an ARB and an ACE inhibitor is made more difficult by the results of the Microalbuminuria-Heart Outcomes Prevention Evaluation (MICRO-HOPE) Trial, in which ramipril reduced the risk for myocardial infarction, stroke, or cardiovascular death by 26% after 2 years. Perhaps the more interesting question is whether the combination of an ACE inhibitor and an ARB is more effective than either drug alone. One meta-analysis showed that ACEI + ARB reduced 24-hour proteinuria to a greater extent than ACEI alone. However, this benefit was associated with small effects on GFR, serum creatinine, potassium, and blood pressure. [40]

A study by Imai et al determined that combined treatment with ACE inhibitors and ARBs significantly decreased blood pressure, proteinuria, and rate of change of reciprocal serum creatinine; however, higher cardiovascular death was reported among the olmesartan-treated patients compared with placebo. Major adverse cardiovascular events and all–cause data were similar between the 2 groups. Hyperkalemia was more frequent in the olmesartan–treated group than in the placebo group. These findings confirm previous studies that combined therapy for patients with diabetic nephropathy may improve short-term biomarkers but is not associated with improvement in long-term hard endpoints. [41]

However, the recent Nephron-D trial, which evaluated the effect of adding losartan, an ARB, to the ACE inhibitor lisinopril on albumin-to-creatinine ratio in 1448 patients with type 2 diabetes was stopped early because of safety concerns. Combination therapy significantly increased the risk of hyperkalemia and acute kidney injury. [42] Thus, the combination should be avoided as a strategy to reduce proteinuria with the hope of slowing progression of diabetic nephropathy, and should be reserved for individual situations in which optimal control of blood pressure may require it. Until further studies shed additional light, for antiproteinuric effect, the addition of an aldosterone antagonist (ie, spironolactone) may be more effective.

Following the advent of ACE inhibitors and ARBs, progress in diabetes and kidney disease outcomes appeared to be stalled for nearly three decades. However, trials with SGLT2 inhibitors in diabetes appear to be ushering in a new era in the prevention or slowing of diabetic kidney disease as well as in the reduction of cardiovascular morbidity. [43, 44, 45, 46] Another bright spot has been the favorable effects on renal and cardiovascular outcomes by the non-steroidal aldosterone antagonist finerenone. [43, 47, 48, 49, 50, 51]

Direct renin inhibitors

In a small double-blind, randomized, crossover trial, Persson et al observed the combination of aliskiren and irbesartan to be more antiproteinuric in type 2 diabetes mellitus than was monotherapy with either drug. [52] This study assessed the effect of aliskiren, a direct renin inhibitor, on proteinuria in patients with type 2 DM (n = 26) and compared the effect with that of placebo, irbesartan (an ARB), and the combination of aliskiren and irbesartan.

Patients were assigned to four 2-month treatments in random order. Monotherapy with either aliskiren or irbesartan significantly improved albuminuria when compared with placebo. Combination therapy with aliskiren and irbesartan reduced albuminuria by 71%, more than did either monotherapy (aliskiren monotherapy 48%; irbesartan monotherapy 58%). Use of direct renin inhibitors with ARBs or ACEIs is no longer recommended. This after the FDA issued an advisory in April 2012 based on deliterios serious side effects when such combinations were used.

Research suggests that vitamin D may have a role in renin inhibition and that vitamin D supplementation may be useful in reducing proteinuria in patients with diabetic nephropathy. Patients with diabetic nephropathy with stage 3 chronic kidney disease (eGFR 59 – 30 mL/min/1.73 m2) or more advanced stages should be evaluated for their vitamin D and parathyroid hormone status as recommended by the National Kidney Foundation- Kidney Disease Dialysis Outcomes Quality Initiative (NKF-KDOQI). [53] If vitamin D levels are low, patients should be given vitamin D supplementation. One randomized controlled trial suggested that vitamin D supplementation may reduce proteinuria in patients with diabetic nephropathy. [54, 55]

Mineralocorticoid Receptor Antagonist Therapy

In July 2021, the FDA approved finerenone (Kerendia) to lower the chances of sustained eGFR decline, end-stage kidney disease, cardiovascular death, nonfatal myocardial infarction, and hospitalization for heart failure in adults with chronic kidney disease (CKD) associated with type 2 DM. It is the first nonsteroidal mineralocorticoid receptor (MR) antagonist to be approved for this purpose. Approval was based on the FIDELIO-DKD trial, a placebo-controlled study that involved over 5700 patients with type 2 DM to whom the maximum-tolerated dose of renin-angiotensin system inhibitor (RASI) was already being administered. However, until more data on finerenone is gathered, RASIs and SGLT2 inhibitors will be the preferred agents for slowing chronic kidney disease in type 2 diabetes. [47, 56, 57]

Endothelin Antagonist Therapy

Endothelin antagonists have demonstrated antifibrotic, anti-inflammatory, and antiproteinuric effects in experimental studies.

A randomized, placebo-controlled, double-blind, parallel-design, dosage-range study on the effect of the endothelin-A antagonist avosentan on urinary albumin excretion rate in 286 patients with diabetic nephropathy, macroalbuminuria, and a blood pressure of < 180/110 mm Hg found that all dosages of avosentan, administered in addition to standard treatment with an ACE inhibitor or an ARB, reduced the mean relative urinary albumin excretion rate (-16.3% to -29.9%, relative to baseline). [58]

Renal Replacement Therapy

As for any other patient with ESRD, diabetic patients with ESRD can be offered renal replacement therapy. Carefully explain the therapeutic options and modalities of renal replacement therapy to patients, their partners, and their families in an early stage of renal failure. In chronically ill patients with diabetes, this tends to be much more important than in those renal patients who do not have diabetes.

In patients with diabetic nephropathy, starting at a creatinine clearance or estimated GFR of 10-15 mL/min is wise. In diabetic patients, starting earlier is useful when hypervolemia renders blood pressure uncontrollable, when the patient experiences anorexia and cachexia or other uremic symptoms, and when severe vomiting is the combined result of uremia and gastroparesis.

In principle, diabetic patients who require renal replacement therapy have the following 4 options:

  • Refusal of further treatment for uremia, leading to a progressive decline in general health and ultimately leading to death

  • Peritoneal dialysis (eg, machine-assisted intermittent peritoneal dialysis, continuous ambulatory peritoneal dialysis, continuous cyclic peritoneal dialysis)

  • Hemodialysis (eg, facility hemodialysis, home hemodialysis)

  • Renal transplantation (eg, cadaver donor kidney, living related-donor kidney, living unrelated-donor kidney [emotionally related donor], living unrelated-donor kidney [unrelated by family or emotionally; the so-called altruistic donor], pancreas plus kidney transplantation)

Peritoneal dialysis and hemodialysis

Dialysis treatment partially reverses insulin resistance so that insulin requirements are often reduced. Adequate control of glycemia is important to prevent hyperglycemia-induced thirst, which can lead to volume overload and hyperkalemia. Proper attention must be given to optimizing nutrition, correcting anemia, controlling hypertension and hyperlipidemia, and modifying associated cardiovascular risk factors.

Regarding peritoneal dialysis, in a recently completed study, female patients with diabetes mellitus had a better outcome in the first 3 years of requiring renal replacement therapy when they chose peritoneal dialysis over hemodialysis. This positive effect did not continue beyond 3 years.

Kidney transplantation and kidney-pancreas transplantation

Except in patients with severe macroangiopathic complications, renal transplantation should be considered a first-line objective because it offers the best degree of medical rehabilitation in patients with uremia and diabetes. This option must be discussed early on with the patient and his or her family. Transplantation even before dialysis (preemptive transplantation) is becoming increasingly popular in some centers.

Renal transplantation is generally restricted to younger patients with type 1 DM; this may not be completely justified because good results have also been achieved in patients with type 2 DM if high-risk patients with macrovascular disease are excluded. Because of higher cardiovascular mortality, long-term survival of patients with diabetes with renal allografts is definitely inferior to that of those without diabetes.

The major rationale for combined kidney and pancreas transplantation is the increased quality of life and, probably, (controversial) halting or even reversing diabetic complications. Transplantation of the more immunogenic pancreas appears to have a higher risk of biopsy-proven acute kidney graft rejection episodes, but the 1-year graft and patient survival rates are not different from those in patients who had kidney transplantation alone.

In patients with type 1 DM, pancreas transplantation is the only treatment that consistently achieves insulin independence. Recently, successful reports of islet cell transplantation have been presented.

Indications for pancreas transplantation in nonuremic patients have not been established. Generally, it is offered to patients with extremely brittle diabetes and documented episodes of hypoglycemia without preceding symptoms. In patients with type 1 DM and renal insufficiency, the following 2 options exist: (1) simultaneous kidney and pancreas transplantation and (2) first kidney and then pancreas transplantation (the latter is usually performed when patients receive a live donor graft).

Urate-Lowering Therapy

A study by Ueno indicated that in patients with type 2 diabetes with hyperuricemia, kidney function significantly improves when serum urate levels are reduced below 6.0 mg/dL, possibly demonstrating a means of slowing nephropathy progression in these patients. [59]

Dietary Changes

A meta-analysis examining the effects of dietary protein restriction (0.5-0.85 g/kg/d) in diabetic patients suggested a beneficial effect on the GFR, creatinine clearance, and albuminuria. However, a large, long-term prospective study is needed to establish the safety, efficacy, and compliance with protein restriction in diabetic patients with nephropathy. Limitations include ensuring compliance by patients.

The American Diabetic Association suggests diets of various energy intake (caloric values), depending on the patient. With advancing renal disease, protein restriction of as much as 0.8-1 g/kg/d may retard the progression of nephropathy.

When nephropathy is advanced, the diet should reflect the need for phosphorus and potassium restriction, with the use of phosphate binders.

A meta-analysis from the Cochrane Renal Group revealed that dietary salt reduction significantly reduced blood pressure (BP) in individuals with type 1 or type 2 diabetes. [60] These findings, along with other evidence relating salt intake to BP and albuminuria in hypertensive and normotensive patients, make a strong case for a reduction in salt intake among patients with diabetes. The recommendation for the general population in public health guidelines is less than 5-6 g/d. Dietary salt reduction may help slow progression of kidney disease in both type 1 and type 2 diabetes.

Restriction of Activity

No restriction in activity is necessary for persons with diabetic nephropathy, unless warranted by other associated complications of diabetes, such as associated coronary disease or peripheral vascular disease.

Measures for Prevention of Diabetic Nephropathy

Efforts should be made to modify and/or treat associated risk factors such as hyperlipidemia, smoking, and hypertension.

Specific goals for prevention include the following:

  • Optimal blood glucose control (hemoglobin A1c [HbA1c] < 7%)

  • Control of hypertension (BP < 120/70 Hg)

  • Avoidance of potentially nephrotoxic substances such as nonsteroidal anti-inflammatory medications and aminoglycosides

  • Early detection and optimal management of diabetes, especially in the setting of family history of diabetes

Long-Term Monitoring

Regular outpatient follow-up is key in managing diabetic nephropathy successfully. Regular annual urinalysis is recommended for screening for microalbuminuria (see the image below). Ensuring optimal glucose control, optimizing blood pressure, and screening for other associated complications of diabetes (eg, retinopathy, diabetic foot, cardiovascular disease) are also crucial.

What hypertension drug is the first choice for diabetic and renal failure patients?
Screening for and prevention of the progression of microalbuminuria in diabetes mellitus. (ACE-I stands for angiotensin-converting enzyme inhibitor)

To see complete information on the conditions below, please go to the main article by clicking on the title:

Complications

Renal complications of diabetic nephropathy include increased risk of urinary tract infections, which may be further increased with the use of SGLT2 inhibitors. Serum electrolyte, water, and acid-base complications are also more common in patients with diabetic nephropathy. For example, type 4 (hyperkalemic, low–anion gap) renal tubular acidosis is more common in patients with type 2 DM, especially those with moderate renal insufficiency, and is associated with decreased ammoniagenesis.

It has also been noted that kidney stones may be more common in patients with type 2 DM, as well as metabolic syndrome. [61] The increased risk of stone disease is linked to insulin resistance, which, as a result of impaired ammoniagenesis, leads to a reduced urine pH. A low urine pH primarily favors uric acid stone formation; studies have found that female patients with type 2 DM, especially, have strikingly higher rates of uric acid stones. [62]

  1. Tang SC, Chan GC, Lai KN. Recent advances in managing and understanding diabetic nephropathy. F1000Res. 2016. 5:[QxMD MEDLINE Link]. [Full Text].

  2. Bjornstad P, Cherney D, Maahs DM. Early diabetic nephropathy in type 1 diabetes: new insights. Curr Opin Endocrinol Diabetes Obes. 2014 Aug. 21 (4):279-86. [QxMD MEDLINE Link]. [Full Text].

  3. Ekinci EI, Jerums G, Skene A, Crammer P, Power D, Cheong KY. Renal structure in normoalbuminuric and albuminuric patients with type 2 diabetes and impaired renal function. Diabetes Care. 2013 Nov. 36(11):3620-6. [QxMD MEDLINE Link].

  4. Shlipak M. Diabetic nephropathy. Clin Evid (Online). 2009 Jan 14. 2009:[QxMD MEDLINE Link].

  5. Burney BO, Kalaitzidis RG, Bakris GL. Novel therapies of diabetic nephropathy. Curr Opin Nephrol Hypertens. 2009 Mar. 18(2):107-11. [QxMD MEDLINE Link].

  6. Kitagawa N, Kitagawa N, Ushigome E, et al. Impact of Isolated High Home Systolic Blood Pressure and Diabetic Nephropathy in Patients with Type 2 Diabetes Mellitus: A 5-Year Prospective Cohort Study. J Clin Med. 2021 Apr 29. 10 (9):[QxMD MEDLINE Link]. [Full Text].

  7. Hall JE, Henegar JR, Dwyer TM, Liu J, Da Silva AA, Kuo JJ. Is obesity a major cause of chronic kidney disease?. Adv Ren Replace Ther. 2004 Jan. 11(1):41-54. [QxMD MEDLINE Link].

  8. Yip JW, Jones SL, Wiseman MJ, Hill C, Viberti G. Glomerular hyperfiltration in the prediction of nephropathy in IDDM: a 10-year follow-up study. Diabetes. 1996 Dec. 45(12):1729-33. [QxMD MEDLINE Link].

  9. Odegaard JI, Chawla A. Connecting type 1 and type 2 diabetes through innate immunity. Cold Spring Harb Perspect Med. 2012 Mar. 2(3):a007724. [QxMD MEDLINE Link]. [Full Text].

  10. Chiarelli F, Gaspari S, Marcovecchio ML. Role of growth factors in diabetic kidney disease. Horm Metab Res. 2009 Aug. 41(8):585-93. [QxMD MEDLINE Link].

  11. Rask-Madsen C, King GL. Kidney complications: factors that protect the diabetic vasculature. Nat Med. 2010 Jan. 16(1):40-1. [QxMD MEDLINE Link].

  12. Ziyadeh FN. Mediators of diabetic renal disease: the case for tgf-Beta as the major mediator. J Am Soc Nephrol. 2004 Jan. 15 Suppl 1:S55-7. [QxMD MEDLINE Link].

  13. Deshpande SD, Putta S, Wang M, Lai JY, Bitzer M, Nelson RG. Transforming growth factor-ß-induced cross talk between p53 and a microRNA in the pathogenesis of diabetic nephropathy. Diabetes. 2013 Sep. 62(9):3151-62. [QxMD MEDLINE Link].

  14. Bherwani S, Saumya AS, Ahirwar AK, et al. The association of folic acid deficiency and diabetic nephropathy in patients with type 2 diabetes mellitus. Endocr Metab Immune Disord Drug Targets. 2016 Apr 15. [QxMD MEDLINE Link].

  15. de Boer IH, Rue TC, Hall YN, et al. Temporal trends in the prevalence of diabetic kidney disease in the United States. JAMA. 2011 Jun 22. 305(24):2532-9. [QxMD MEDLINE Link].

  16. Klessens CQ, Woutman TD, Veraar KA, et al. An autopsy study suggests that diabetic nephropathy is underdiagnosed. Kidney Int. 2016 Jul. 90 (1):149-56. [QxMD MEDLINE Link].

  17. Fan JZ, Wang R. Non-diabetic renal diseases in patients with type 2 diabetes: a single center study. Intern Med J. 2017 Dec 5. [QxMD MEDLINE Link].

  18. Pavkov ME, Bennett PH, Knowler WC, Krakoff J, Sievers ML, Nelson RG. Effect of youth-onset type 2 diabetes mellitus on incidence of end-stage renal disease and mortality in young and middle-aged Pima Indians. JAMA. 2006 Jul 26. 296(4):421-6. [QxMD MEDLINE Link].

  19. Zhang J, Wang Y, Li L, et al. Diabetic retinopathy may predict the renal outcomes of patients with diabetic nephropathy. Ren Fail. 2018 Nov. 40 (1):243-51. [QxMD MEDLINE Link].

  20. Jiang R, Law E, Zhou Z, Yang H, Wu EQ, Seifeldin R. Clinical Trajectories, Healthcare Resource Use, and Costs of Diabetic Nephropathy Among Patients with Type 2 Diabetes: A Latent Class Analysis. Diabetes Ther. 2018 Mar 29. [QxMD MEDLINE Link].

  21. Rosolowsky ET, Skupien J, Smiles AM, et al. Risk for ESRD in type 1 diabetes remains high despite renoprotection. J Am Soc Nephrol. 2011 Mar. 22(3):545-53. [QxMD MEDLINE Link]. [Full Text].

  22. Kostadaras A. Risk Factors for Diabetic Nephropathy. Astoria Hypertension Clinic. Available at http://www.kidneydoctor.com/dm.htm.

  23. Iliadis F, Didangelos T, Ntemka A, et al. Glomerular filtration rate estimation in patients with type 2 diabetes: creatinine- or cystatin C-based equations?. Diabetologia. 2011 Dec. 54(12):2987-94. [QxMD MEDLINE Link].

  24. Jansson Sigfrids F, Dahlstrom EH, Forsblom C, et al. Remnant cholesterol predicts progression of diabetic nephropathy and retinopathy in type 1 diabetes. J Intern Med. 2021 May 8. [QxMD MEDLINE Link]. [Full Text].

  25. Tervaert TW, Mooyaart AL, Amann K, et al. Pathologic classification of diabetic nephropathy. J Am Soc Nephrol. 2010 Apr. 21 (4):556-63. [QxMD MEDLINE Link]. [Full Text].

  26. Suckling RJ, He FJ, Macgregor GA. Altered dietary salt intake for preventing and treating diabetic kidney disease. Cochrane Database Syst Rev. 2010 Dec 8. 12:CD006763. [QxMD MEDLINE Link].

  27. Heerspink HJ, Holtkamp FA, Parving HH, Navis GJ, Lewis JB, Ritz E, et al. Moderation of dietary sodium potentiates the renal and cardiovascular protective effects of angiotensin receptor blockers. Kidney Int. 2012 Mar 21. [QxMD MEDLINE Link].

  28. Diabetes Control and Complications Research Group. Effect of intensive therapy on the development and progression of diabetic nephropathy in the Diabetes Control and Complications Trial. The Diabetes Control and Complications (DCCT) Research Group. Kidney Int. 1995 Jun. 47(6):1703-20. [QxMD MEDLINE Link].

  29. UK Prospective Diabetes Study Group. 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. 1998 Sep 12. 352(9131):837-53. [QxMD MEDLINE Link].

  30. Bergman AJ, Cote J, Yi B, Marbury T, Swan SK, Smith W. Effect of renal insufficiency on the pharmacokinetics of sitagliptin, a dipeptidyl peptidase-4 inhibitor. Diabetes Care. 2007 Jul. 30(7):1862-4. [QxMD MEDLINE Link].

  31. Scheen AJ. Pharmacokinetic considerations for the treatment of diabetes in patients with chronic kidney disease. Expert Opin Drug Metab Toxicol. 2013 May. 9(5):529-50. [QxMD MEDLINE Link].

  32. Snyder RW, Berns JS. Use of insulin and oral hypoglycemic medications in patients with diabetes mellitus and advanced kidney disease. Semin Dial. 2004 Sep-Oct. 17(5):365-70. [QxMD MEDLINE Link].

  33. Lamos EM, Younk LM, Davis SN. Canagliflozin , an inhibitor of sodium-glucose cotransporter 2, for the treatment of type 2 diabetes mellitus. Expert Opin Drug Metab Toxicol. 2013 Jun. 9(6):763-75. [QxMD MEDLINE Link].

  34. Salamon M. Diabetes Drug 'Home Run' Curtailing Kidney Failure Risk. Medscape Medical News. 2019 Apr 15. [Full Text].

  35. Linnebjerg H, Kothare PA, Park S, Mace K, Reddy S, Mitchell M. Effect of renal impairment on the pharmacokinetics of exenatide. Br J Clin Pharmacol. 2007 Sep. 64(3):317-27. [QxMD MEDLINE Link].

  36. Davidson JA, Brett J, Falahati A, Scott D. Mild renal impairment and the efficacy and safety of liraglutide. Endocr Pract. 2011 May-Jun. 17(3):345-55. [QxMD MEDLINE Link].

  37. Young A. Clinical studies. Adv Pharmacol. 2005. 52:289-320. [QxMD MEDLINE Link].

  38. Mogensen CE. The effect of blood pressure intervention on renal function in insulin-dependent diabetes. Diabete Metab. 1989. 15(5 Pt 2):343-51. [QxMD MEDLINE Link].

  39. Laight DW. Therapeutic inhibition of the renin angiotensin aldosterone system. Expert Opin Ther Pat. 2009 Jun. 19(6):753-9. [QxMD MEDLINE Link].

  40. Jennings DL, Kalus JS, Coleman CI, Manierski C, Yee J. Combination therapy with an ACE inhibitor and an angiotensin receptor blocker for diabetic nephropathy: a meta-analysis. Diabet Med. 2007 May. 24(5):486-93. [QxMD MEDLINE Link].

  41. Imai E, Chan JC, Ito S, et al. Effects of olmesartan on renal and cardiovascular outcomes in type 2 diabetes with overt nephropathy: a multicentre, randomised, placebo-controlled study. Diabetologia. 2011 Dec. 54(12):2978-2986. [QxMD MEDLINE Link].

  42. Fried LF, Emanuele N, Zhang JH, Brophy M, Conner TA, Duckworth W. Combined angiotensin inhibition for the treatment of diabetic nephropathy. N Engl J Med. 2013 Nov 14. 369(20):1892-903. [QxMD MEDLINE Link].

  43. Shenoy SV, Nagaraju SP, Bhojaraja MV, Prabhu RA, Rangaswamy D, Rao IR. Sodium-glucose cotransporter-2 inhibitors and non-steroidal mineralocorticoid receptor antagonists: Ushering in a new era of nephroprotection beyond renin-angiotensin system blockade. Nephrology (Carlton). 2021 Nov. 26 (11):858-71. [QxMD MEDLINE Link]. [Full Text].

  44. Persson F, Rossing P, Vart P, et al. Efficacy and Safety of Dapagliflozin by Baseline Glycemic Status: A Prespecified Analysis From the DAPA-CKD Trial. Diabetes Care. 2021 Aug. 44 (8):1894-7. [QxMD MEDLINE Link]. [Full Text].

  45. Packer M, Anker SD, Butler J, et al. Empagliflozin in Patients With Heart Failure, Reduced Ejection Fraction, and Volume Overload: EMPEROR-Reduced Trial. J Am Coll Cardiol. 2021 Mar 23. 77 (11):1381-92. [QxMD MEDLINE Link]. [Full Text].

  46. Heerspink HJL, Oshima M, Zhang H, et al. Canagliflozin and Kidney-Related Adverse Events in Type 2 Diabetes and CKD: Findings From the Randomized CREDENCE Trial. Am J Kidney Dis. 2021 May 23. [QxMD MEDLINE Link].

  47. Bakris GL, Agarwal R, Anker SD, et al. Effect of Finerenone on Chronic Kidney Disease Outcomes in Type 2 Diabetes. N Engl J Med. 2020 Dec 3. 383 (23):2219-29. [QxMD MEDLINE Link]. [Full Text].

  48. Filippatos G, Anker SD, Agarwal R, et al. Finerenone and Cardiovascular Outcomes in Patients With Chronic Kidney Disease and Type 2 Diabetes. Circulation. 2021 Feb 9. 143 (6):540-52. [QxMD MEDLINE Link]. [Full Text].

  49. Ferreira JP, Zannad F, Pocock SJ, et al. Interplay of Mineralocorticoid Receptor Antagonists and Empagliflozin in Heart Failure: EMPEROR-Reduced. J Am Coll Cardiol. 2021 Mar 23. 77 (11):1397-1407. [QxMD MEDLINE Link]. [Full Text].

  50. Epstein M. Aldosterone and Mineralocorticoid Receptor Signaling as Determinants of Cardiovascular and Renal Injury: From Hans Selye to the Present. Am J Nephrol. 2021. 52 (3):209-16. [QxMD MEDLINE Link]. [Full Text].

  51. Bakris GL, Agarwal R, Filippatos G, FIDELIO-DKD Study Group. Finerenone and Chronic Kidney Disease Outcomes in Type 2 Diabetes. Reply. N Engl J Med. 2021 Mar 18. 384 (11):e42. [QxMD MEDLINE Link].

  52. Persson F, Rossing P, Reinhard H, Juhl T, Stehouwer CD, Schalkwijk C, et al. Renal effects of aliskiren compared with and in combination with irbesartan in patients with type 2 diabetes, hypertension, and albuminuria. Diabetes Care. 2009 Oct. 32(10):1873-9. [QxMD MEDLINE Link]. [Full Text].

  53. [Guideline] National Kidney Foundation. NKF-KDOQI Guidelines. Available at http://www.kidney.org/professionals/kdoqi/guidelines.cfm.

  54. Agarwal R. Vitamin D, proteinuria, diabetic nephropathy, and progression of CKD. Clin J Am Soc Nephrol. 2009 Sep. 4(9):1523-8. [QxMD MEDLINE Link].

  55. de Zeeuw D, Agarwal R, Amdahl M, Audhya P, Coyne D, Garimella T, et al. Selective vitamin D receptor activation with paricalcitol for reduction of albuminuria in patients with type 2 diabetes (VITAL study): a randomised controlled trial. Lancet. 2010 Nov 6. 376(9752):1543-51. [QxMD MEDLINE Link].

  56. US Food and Drug Administration. FDA Approves Drug to Reduce Risk of Serious Kidney and Heart Complications in Adults with Chronic Kidney Disease Associated with Type 2 Diabetes. FDA. Available at https://www.fda.gov/drugs/drug-safety-and-availability/fda-approves-drug-reduce-risk-serious-kidney-and-heart-complications-adults-chronic-kidney-disease. July 9, 2021; Accessed: July 14, 2021.

  57. Zoler ML. FDA Approves Finerenone (Kerendia) for Slowing CKD in Type 2 Diabetes. Medscape Medical News. 2021 Jul 12. [Full Text].

  58. Wenzel RR, Littke T, Kuranoff S, Jürgens C, Bruck H, Ritz E, et al. Avosentan reduces albumin excretion in diabetics with macroalbuminuria. J Am Soc Nephrol. 2009 Mar. 20(3):655-64. [QxMD MEDLINE Link]. [Full Text].

  59. Ueno N. Urate-Lowering Therapy Ameliorates Kidney Function in Type 2 Diabetes Patients With Hyperuricemia. J Clin Med Res. 2017 Dec. 9 (12):1007-12. [QxMD MEDLINE Link]. [Full Text].

  60. Suckling RJ, He FJ, Macgregor GA. Altered dietary salt intake for preventing and treating diabetic kidney disease. Cochrane Database Syst Rev. 2010 Dec 8. 12:CD006763. [QxMD MEDLINE Link].

  61. Daudon M, Jungers P. Diabetes and nephrolithiasis. Curr Diab Rep. 2007 Dec. 7 (6):443-8. [QxMD MEDLINE Link].

  62. Daudon M, Traxer O, Conort P, Lacour B, Jungers P. Type 2 diabetes increases the risk for uric acid stones. J Am Soc Nephrol. 2006 Jul. 17 (7):2026-33. [QxMD MEDLINE Link]. [Full Text].

  63. [Guideline] American Diabetes Association. 10. Microvascular Complications and Foot Care: Standards of Medical Care in Diabetes-2018. Diabetes Care. 2018 Jan. 41 (Suppl 1):S105-18. [QxMD MEDLINE Link]. [Full Text].

  64. Kocasarac C, Yigit Y, Sengul E, Sakalar YB. Choroidal thickness alterations in diabetic nephropathy patients with early or no diabetic retinopathy. Int Ophthalmol. 2017 Apr 11. [QxMD MEDLINE Link].

  65. Park SB, Kim SS, Kim IJ, et al. Variability in glycated albumin levels predicts the progression of diabetic nephropathy. J Diabetes Complications. 2017 Feb 3. [QxMD MEDLINE Link].

  66. Zelnick LR, Weiss NS, Kestenbaum BR, et al. Diabetes and CKD in the United States Population, 2009-2014. Clin J Am Soc Nephrol. 2017 Dec 7. 12 (12):1984-90. [QxMD MEDLINE Link].

  67. Cheung CY, Ma MKM, Chak WL, Tang SCW. Cancer risk in patients with diabetic nephropathy: a retrospective cohort study in Hong Kong. Medicine (Baltimore). 2017 Sep. 96 (38):e8077. [QxMD MEDLINE Link]. [Full Text].

Author

Vecihi Batuman, MD, FASN Huberwald Professor of Medicine, Section of Nephrology-Hypertension, Interim Chair, Deming Department of Medicine, Tulane University School of Medicine

Vecihi Batuman, MD, FASN is a member of the following medical societies: American College of Physicians, American Society of Hypertension, American Society of Nephrology, International Society of Nephrology, Southern Society for Clinical Investigation

Disclosure: Nothing to disclose.

Coauthor(s)

Rebecca J Schmidt, DO, FACP, FASN Assistant Dean for Outreach and Community Engagement, Professor of Medicine and Vice Chair for Rural Outreach and Service to the State, Department of Medicine, Section of Nephrology, West Virginia University School of Medicine

Rebecca J Schmidt, DO, FACP, FASN is a member of the following medical societies: American College of Physicians, American Medical Association, American Society of Nephrology, International Society of Nephrology, National Kidney Foundation, Renal Physicians Association, West Virginia State Medical Association

Disclosure: Nothing to disclose.

Sandeep S Soman, MBBS, MD, DNB Senior Staff Physician, Department of Internal Medicine, Division of Nephrology and Hypertension, Henry Ford Hospital

Sandeep S Soman, MBBS, MD, DNB is a member of the following medical societies: American College of Physicians, American Medical Association, American Society of Nephrology

Disclosure: Nothing to disclose.

Specialty Editor Board

Francisco Talavera, PharmD, PhD Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Chief Editor

Romesh Khardori, MD, PhD, FACP (Retired) Professor, Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Eastern Virginia Medical School

Romesh Khardori, MD, PhD, FACP is a member of the following medical societies: American Association of Clinical Endocrinologists, American College of Physicians, American Diabetes Association, Endocrine Society

Disclosure: Nothing to disclose.

Acknowledgements

George R Aronoff, MD Director, Professor, Departments of Internal Medicine and Pharmacology, Section of Nephrology, Kidney Disease Program, University of Louisville School of Medicine

George R Aronoff, MD is a member of the following medical societies: American Federation for Medical Research, American Society of Nephrology, Kentucky Medical Association, and National Kidney Foundation

Disclosure: Nothing to disclose.

Sreepada TK Rao, MD, FACP Professor, Department of Medicine, State University of New York Downstate Medical Center

Sreepada TK Rao, MD, FACP is a member of the following medical societies: American Society of Hypertension, American Society of Nephrology, and International Society of Nephrology

Disclosure: Nothing to disclose.

Which hypertension drug is the first choice for diabetic and renal failure patients?

Renin–angiotensin aldosterone inhibitors are considered the mainstay treatment for hypertension in diabetic patients, especially in the presence of albuminuria. Whether strict blood pressure reduction entails a favorable renal outcome also in non-albuminuric patients is at present unclear.

What is the drug of choice for a patient with diabetes and hypertension?

ANGIOTENSIN RECEPTOR BLOCKERS Like ACE inhibitors, ARBs reduce the complications of diabetes (e.g., progression of kidney disease), and they are preferred agents for managing hypertension in patients with diabetes.

What is the drug of choice for hypertensive patients who have chronic kidney disease?

Angiotensin-converting enzyme inhibitors (ACEis) and angiotensin II receptor blockers (ARBs) are the mainstays of hypertension treatment in CKD.

Which antihypertensive is used for diabetic nephropathy?

Angiotensin-converting enzyme (ACE) inhibitors are the only antihypertensive agents with proven effectiveness for the primary prevention of diabetic nephropathy (defined as an albumin excretion of less than 30 mg per day on three serial measurements) caused by type 1 or type 2 diabetes.