This site is intended for healthcare professionals
patient receiving dialysis, hospital image
Chronic Kidney Disease Learning Zone

Treating CKD

Read time: 130 mins
Last updated:25th Apr 2022
Published:21st Oct 2021

Find out about the treatments for chronic kidney disease (CKD) and its comorbidities and explore:

  • Goals for the treatment of CKD in our expert videos with Professor Hiddo Heerspink, Professor Vlado Perkovic and Dr George Bakris
  • Strategies for managing comorbid conditions and end-stage kidney disease (ESKD)
  • CKD treatments just over the horizon and links to CKD guideline resources
  • CKD patient case study quizzes to test your understanding

Treatment goals for chronic kidney disease

What are the priorities for treatment optimisation in chronic kidney disease?

Professor Vlado Perkovic and Dr George Bakris discuss the treatment goals and priorities for optimising treatment of chronic kidney disease (CKD).

The management of CKD relies on a combination of different strategies, including: controlling nephron injury, normalising hyperfiltration, controlling CKD-­related complications, and preparations for kidney replacement therapy.

The core principle of this management is ‘the earlier, the better’, which aims to prevent nephron loss as early as possible and reduce the progression to end-stage renal disease (ESRD)1.

What are the overall goals when managing CKD?

The management of CKD is based on the clinical diagnosis and disease stage, according to glomerular filtration rate (GFR) and albuminuria. Individualised therapies, that are guided by the cause and pathological processes, are determined by the specific clinical diagnosis2. The treatments for CKD aim to2,3:

  • prevent disease development
  • slow disease progression
  • reduce the complications of decreased GFR
  • reduce the risk of cardiovascular disease
  • improve survival and quality of life

The disease stage can also be used to guide non-specific therapies that slow the progression of the disease and reduce the risk of complications2,3. Recommendations based on disease stage are cumulative: early-stage recommendations are included within late-stage recommendations2,3.

How do you control ongoing nephron injury?

As a variety of triggers can drive nephron injury, disrupting these triggers with treatment can slow the progression of CKD to end-stage renal disease (ESRD)1.

For genetic causes of kidney disease, treatments are limited to enzyme replacement therapy or substrate supplementation. In cases of immune mediated nephron injury that have a genetic component, CKD progression associated with C3 glomerulonephritis or atypical haemolytic uraemic syndrome can be treated with complement inhibitors4.

Immunomodulatory drugs can be used to target immune mediated CKD disorders with the aim of limiting nephron injury5

In acute forms of immune mediated nephron injury, presentation is seen as either vasculitis, immune complex glomerulonephritis, or interstitial nephritis (such as allograft rejection).

However, in smouldering immune injuries (such as chronic IgA nephropathy) it is difficult to differentiate immune versus non­immune mechanisms in the progression of CKD and, as a consequence, the efficacy of management through immunosuppression or RAS blockade and blood pressure control is less apparent6.

How do you minimise the stress on remnant nephrons in CKD?

It is essential to minimise the stress on remnant nephrons by preventing further acute kidney injury (AKI). It is therefore important to avoid1:

  • nephrotoxins (such as high volumes of radio contrast media, non-steroidal anti-inflammatory drugs, proton pump inhibitors or occupational toxins)
  • hypovolaemic states
  • urinary outflow obstruction
  • smoking-related cardiovascular disease
  • specific antibiotic treatment (limited to cases of dysuria, bacteriuria, and leukocyturia)

Specific medications do exist to prevent some of these triggers, such as urinary tract obstruction, infections, and some forms of toxic injury; however, the recovery of the lost nephrons is not possible1.

How do you normalise single-nephron hyperfiltration?

Angiotensin-converting enzyme inhibitors (ACEi) or angiotensin receptor blockers (ARBs) can inhibit the renin-angiotensin system (RAS) and consequently significantly reduce single-nephron GFR and glomerular filtration pressure. This leads to decline in proteinuria and total GFR and moderately increased serum creatinine levels7.

A moderate increase in serum creatinine levels indicating a decline in single nephron GFR, can be a powerful predictor of a nephroprotective effect8

While increased serum creatinine may be disconcerting to patients and some physicians, reducing hyperfiltration in remnant nephrons is essential for reducing the progression of CKD in patients with proteinuria1. Conversely, ACEi or ARBs do not reduce non­proteinuric forms of CKD progression, such as autosomal dominant polycystic kidney disease9.

Various randomised clinical trials have shown that RAS inhibitors can reduce or even prevent CKD progression10. A further reduction in proteinuria and CKD progression can be achieved with a reduction in dietary salt as well as medications that support the control of blood pressure and hyperlipidaemia11,12. These interventions can be particularly necessary where kidney replacement therapy is not available or affordable1.

In diabetic kidney disease (DKD), sodium-glucose cotransporter 2 (SGLT2) inhibition can cause an immediate functional reduction in GFR. The GFR reduction is thought to reduce the tubular transport work and metabolic demand, improving renal cortical oxygenation, which is associated with protection from CKD13,14. The effect of SGLT2 inhibitors to reduce glomerular hyperfiltration is thought to be a shared mechanism between DKD and CKD15. In the DAPA-CKD trial, dapagliflozin was evaluated for proteinuric CKD attributed to hypertension or glomerular diseases that did not require immunosuppression. On a background of RAS inhibition, similar risk reductions for the primary and secondary outcomes were observed irrespective of diabetes status or CKD aetiology, with no evidence of statistical interaction by subgroup across primary and secondary outcomes16.

Why is it important to control the factors associated with CKD progression?

An important part of the management of CKD is addressing the multiple factors that are associated with disease progression. Various general measures have been described in the Kidney Disease: Improving Global Outcomes (KDIGO) guidelines that address cardiovascular health and CKD together, or separately17.

Cardiovascular disease (CVD) risk factors can indirectly and directly impact CKD progression and so general lifestyle measures are recommended to improve17:

  • cardiovascular health
  • blood pressure (BP) control
  • Interruption of the renin-angiotensin-aldosterone system (RAAS)

Metabolic parameters, such as blood sugar, uric acid, acidosis, and dyslipidaemia, are also recommended to monitor and control17.

Help us create relevant CKD content for you

Managing chronic kidney disease comorbid conditions

Professor Hiddo Heerspink details the key findings from the DAPA-CKD and FIDELIO-DKD clinical trials for non-diabetic chronic kidney disease and diabetic kidney disease.

CKD patients can have multiple comorbid conditions, including diabetes, hypertension, and cardiovascular disease (CVD); high risks of developing hypoglycaemia and adverse drug reactions, multiple life demands, and psychosocial factors that influence behaviours and clinical outcomes. As a result, appropriate treatments are needed to reduce symptoms and minimise the impact on quality of life18,19.

How do you manage diabetes and blood pressure in chronic kidney disease patients?

The Kidney Disease: Improving Global Outcomes (KDIGO) guideline for the management of blood pressure in CKD recommends a target systolic blood pressure for adults with high blood pressure and CKD of <120 mm Hg18. This can be achieved through the use of renin-angiotensin system inhibitors (RASi) for people with high blood pressure, CKD, and moderately-to-severely increased albuminuria with diabetes18.

The management of patients with type 2 diabetes (T2D) and CKD (Figure 1) should involve19:

  • lifestyle therapy
  • first-line treatment with metformin and a sodium-glucose cotransporter-2 (SGLT2) inhibitor
  • additional drug therapy as needed for glycaemic control

T3 CKD_Fig1_T3 CKD_Fig1.png

Figure 1. Treatment algorithm for patients with type 2 diabetes and chronic kidney disease (Adapted19). Kidney icon indicates eGFR (mL/min/1.73 m2); dialysis machine icon indicates dialysis. CKD, chronic kidney disease; DPP-4, dipeptidyl peptidase-4; eGFR, estimated glomerular filtration rate; GLP-1, glucagon-like peptide-1; SGLT2, sodium-glucose cotransporter-2; TZD, thiazolidinedione.

The higher cardiovascular burden present in these patients also requires comprehensive management through lifestyle interventions that address the underlying comorbidities as well as appropriate pharmacotherapy depending on the severity and stage of the CKD20.

What treatments are available for chronic kidney disease with comorbid blood pressure control, cardiovascular disease, or diabetes?

Professor Hiddo Heerspink points out that the ultimate treatment goal for chronic kidney disease is to slow down the loss of kidney function, a goal that is becoming more achievable with recent approvals for SGLT2 inhibitors and mineralocorticoid receptor antagonists.

Novel treatments for patients with CKD or comorbid CKD have been developed, or approved for clinical use, including21:

  • Sodium-glucose cotransporter-2 (SGLT2) inhibitors
  • Mineralocorticoid receptor antagonists (MRA)
  • Glucagon-like peptide-1 receptor agonists (GLP-1 RA)
  • Dipeptidyl peptidase-4 inhibitors (DPP4i)

Drugs within these classes have been examined through large clinical trials to evaluate the cardiovascular and kidney outcomes in patients with T2D, CKD, and comorbid CKD19. The results from these trials were used as the basis for recommendations made within the KDIGO guidelines for patients with T2D and CKD19.

People with CKD have high cardiovascular risk, with cardiovascular death the leading cause of death22.

Several new treatments to decrease the risk of cardiovascular diseases in CKD are in clinical development or have been already approved, such as SGLT2 inhibitors or mineralocorticoid receptor antagonists, giving hope that cardiovascular risk in patients with CKD may be modifiable22

Currently, there is limited evidence on the use of specific antihypertensive agents to treat hypertension in CKD. In patients with CKD and high blood pressure, combination therapy with two or more antihypertensive drugs is recommended23. Antihypertensive treatment algorithms in CKD are based on expert opinion, pathophysiologic or pharmacodynamic considerations, or extrapolated from research findings23.

Renin-angiotensin system inhibitors (RASi) for diabetic kidney disease

RASi are recommended for patients with diabetes, hypertension, and albuminuria (albumin-creatinine ratio >30 mg/g)19. RASi slow the progression of CKD in patients with albuminuria and hypertension independent of their blood pressure effects24. KDIGO consider RASi as foundational therapy for diabetic kidney disease (DKD)25.

A network meta-analysis of 119 randomised trials (N = 64,768) of CKD patients, with or without diabetes and albuminuria, examined RASi for kidney and cardiovascular outcomes compared to other treatments and placebo26.

The results showed that angiotensin-converting enzyme inhibitors (ACEi) and angiotensin II receptor blockers (ARBs) both reduce the risk of kidney failure and major cardiovascular events. However, only ACEi reduced the risk of all-cause death compared to active control26.

In CKD patients without diabetes and severely increased albuminuria, three phase 3 trials in the nineties also highlighted the potential cardiovascular benefits of RASi compared to placebo in addition to renal benefits27–29:

  • REIN 1 Phase 3 trial: 2-arm Ramipril Efficacy in Nephropathy Stratum-1
  • REIN 2 Phase 3 trial: 3-arm Ramipril Efficacy in Nephropathy Stratum-2
  • AIPRI Phase 3 trial: 2-arm Angiotensin-converting enzyme inhibition in progressive renal insufficiency

The early 2000’s also saw two phase 3 studies that demonstrated a beneficial effect of RASi independent of blood pressure control, in CKD compared to placebo30,31:

  • IDNT Phase 3 trial: 3-arm Irbesartan Diabetic Nephropathy Trial
  • RENAAL Phase 3 trial: 2-arm Reduction of Endpoints in NIDDM with the Angiotensin II Antagonist Losartan Study

Metformin for diabetes

Since its approval in the nineties, metformin has been prescribed for the management of T2D for over 20 years32. While prescribing metformin, the eGFR should be monitored and the dose reduced when the rate is less than 45 mL/min/1.73 m2 or withdrawn when eGFR drops to <30 mL/min/1.73 m2 or kidney failure develops19.

Currently, treatment with metformin and a SGLT2 inhibitor is recommended in most patients with an eGFR of ≥30 mL/min/1.73 m2, diabetes, and CKD19

Metformin may increase risk for lactic acidosis and vitamin B12 deficiency; therefore, monitoring of levels is recommended with long-term use33. When these drugs are not well tolerated or are insufficient to attain glycaemic goals, additional drugs can be considered based on various factors, such as patient preferences, comorbidities, eGFR, and costs (Figure 2)19.

T3 CKD_Fig2.png

Figure 2. Patient factors influencing selection of glucose-lowering drugs other than SGLT2 inhibitors and metformin in type 2 diabetes and CKD (Adapted19). AGI, alpha-glucosidase inhibitor; ASCVD, atherosclerotic cardiovascular disease; DPP4i, dipeptidyl peptidase-4 inhibitor; eGFR, estimated glomerular filtration rate; GLP1RA, glucagon-like peptide-1 receptor agonist; SU, sulfonylurea; TZD, thiazolidinedione.

GLP-1 RAs are notably recommended as additional agents due to their beneficial effects in reducing cardiovascular events as well as their potential to prevent macroalbuminuria or a reduction in eGFR decline19.

Mineralocorticoid receptor antagonists for diabetic kidney disease

The mineralocorticoid receptor antagonist finerenone is indicated by the US Food and Drugs Administration (USFDA) to reduce the risk of sustained eGFR decline, end-stage kidney disease (ESKD), cardiovascular death, non-fatal myocardial infarction, and hospitalisation for heart failure (HF), in adult patients with CKD associated with type 2 diabetes34,35.

In the phase II ARTS trial (Arterial Revascularization Therapies Study) with >450 patients with CKD and congestive heart failure (HF), the mineralocorticoid receptor antagonist (MRA) finerenone reduced the urinary albumin-creatinine ratio and NT-proBNP (N-terminal pro-BNP) as much as spironolactone, with significantly lower rates of deteriorating kidney function and hyperkalemia36.

In the phase III trials FIDELIO-DKD (Efficacy and Safety of Finerenone in Subjects With Type 2 Diabetes Mellitus and Diabetic Kidney Disease) and FIGARO-DKD (Finerenone in Reducing CV Mortality and Morbidity in Diabetic Kidney Disease), >13 000 T2D patients with CKD were evaluated to determine whether finerenone can reduce cardiovascular morbidity and mortality or prevent progression of kidney disease37,38.

In FIDELIO-DKD, during a median follow-up of 2.6 years, a primary outcome event (kidney failure, a sustained decrease of at least 40% in the eGFR from baseline, or death from renal causes) occurred in 504 of 2,833 patients (17.8%) in the finerenone group and 600 of 2,841 patients (21.1%) in the placebo group (HR 0.82; 95% CI, 0.73–0.93; P=0.001). A key secondary outcome event (death from cardiovascular causes, nonfatal myocardial infarction, nonfatal stroke, or hospitalisation for HF) occurred in 367 patients (13.0%) in the finerenone group, and 420 patients (14.8%) in the placebo group (HR 0.86; 95% CI, 0.75–0.99; P=0.03)37.

In FIGARO-DKD, among the patients included in the analysis, during a median follow-up of 3.4 years, a primary outcome event occurred in 458 of 3,686 patients (12.4%) in the finerenone group, and in 519 of 3,666 (14.2%) in the placebo group (HR 0.87; 95% CI, 0.76–0.98; P=0.03), with the benefit driven by a lower incidence of hospitalisation for HF (HR 0.71; 95% CI, 0.56–0.90). The secondary composite outcome (kidney failure, a sustained decrease of at least 40% in the eGFR from baseline, or death from renal causes) occurred in 350 patients (9.5%) in the finerenone group, and in 395 (10.8%) in the placebo group (HR 0.87; 95% CI, 0.76–1.01)38.

As FIDELIO-DKD and FIGARO-DKD show, in patients with CKD and T2D, finerenone therapy can provide cardiorenal protection, compared with placebo37,38

SGLT2 inhibitors for chronic kidney disease and diabetic kidney disease

In patients with diabetes and CKD, SGLT2 inhibitors have been investigated in various trials for the treatment of diabetes in CKD (Table 1)39-42. Three SGLT2 inhibitors canagliflozin, dapagliflozin, and empagliflozin, have been approved by the USFDA and EMA, are recommended by the KDIGO guidelines for the treatment of hyperglycaemia in T2D, and as foundational therapy in diabetic kidney disease (DKD)19,25,43. In an important regulatory shift, some SGLT2 inhibitors are no longer indicated as therapy only for comorbid CKD; they have been approved for treating CKD itself44–46.

Canagliflozin

Canagliflozin is USFDA and EMA indicated to reduce the risk of ESKD, doubling of serum creatinine, cardiovascular death, and hospitalisation for heart failure in adults with T2D, and diabetic nephropathy with albuminuria47,48.

The CANVAS programme (Table 1) comprised two double-blind, randomised trials (CANVAS and CANVAS-R) that aimed to evaluate the long-term renal effects of canagliflozin40. In the CANVAS study (N = 4,330), patients with T2D were randomised 1:1:1 to receive 300 mg canagliflozin, 100 mg canagliflozin, or placebo49. Similarly, CANVAS-R (N = 5,812) patients were randomised 1:1 to receive canagliflozin 100 mg or a matching placebo49.

Sustained doubling of serum creatinine, ESKD, and death from renal causes occurred less frequently in the canagliflozin group, compared with the placebo group (HR 0.53; 95% CI, 0.33–0.84; P heterogeneity = 0.21 and >0.50, respectively) in the overall CANVAS programme population40,50.

In the more recent CREDENCE study (N = 4,401), the first study of an SGLT2 inhibitor with a combined renal end-point as a primary outcome, patients with T2D, CKD, and albuminuria were randomised to receive canagliflozin (100 mg/day) or placebo41.

The CREDENCE study was prematurely ended after a planned interim analysis due to the clear benefit of canagliflozin compared to placebo41

The results of the CREDENCE trial showed that the relative risk of the renal-specific composite of ESKD, a doubling of the creatinine level, or death from renal causes was lower by 34% (HR 0.66; 95% CI, 0.53–0.81; P<0.001), and the relative risk of ERSD was lower by 32% (HR 0.68, 95%, CI 0.54–0.86; P = 0.002)41.

Table 1. Overview of selected large, placebo-controlled clinical outcome trials assessing the benefits and adverse effects of SGLT2i in chronic kidney disease (Adapted51,25). ACR, albumin-creatinine ratio; DKA, diabetic ketoacidosis; eGFR, estimated glomerular filtration rate; GFR, glomerular filtration rate; GMI, genital mycotic infections; MACE, major adverse cardiovascular events; NA, data not published. ↔, no significant difference; ↓, significant reduction in risk [hazard ratio (HR) estimate >0.7 and 95% confidence interval (CI) not overlapping 1]; ↓↓, significant reduction in risk [HR estimate ≤0.7 and 95% CI not overlapping 1].

T3_CKD_Table1a.png

Dapagliflozin

Dapagliflozin is USFDA indicated to reduce the risk of sustained eGFR decline, ESKD, cardiovascular death, and hospitalisation for HF in adults with CKD at risk of progression45. It became the first SGLT2 inhibitor to receive approval by the EMA for the treatment of adults with CKD, with or without diabetes44. 

DECLARE-TIMI 58 (Table 1) was a randomised, double-blind, multi-national phase III trial that studied the effects of dapagliflozin on hard renal outcomes42.

In T2D patients at risk for atherosclerotic cardiovascular disease, dapagliflozin resulted in a lower rate of cardiovascular death or hospitalization for heart failure42

The results from the trial showed that dapagliflozin met the prespecified criterion for non-inferiority to placebo in terms of major adverse cardiovascular events (P<0.001)42.

The two primary efficacy analyses showed that, while dapagliflozin did not produce a lower rate of major adverse cardiovascular events (defined as cardiovascular death, myocardial infarction, or ischemic stroke) compared to placebo (8.8% vs 9.4%; HR 0.93; 95% CI, 0.84–1.03; P=0.17), dapagliflozin resulted in a lower rate of cardiovascular death or hospitalisation for heart failure (4.9% vs 5.8%; HR 0.83; 95% CI, 0.73–0.95; P=0.005)42.

In the more recent Dapagliflozin and Prevention of Adverse Outcomes in Chronic Kidney Disease (DAPA-CKD) trial, the long-term efficacy and safety of dapagliflozin in patients with chronic kidney disease, with or without T2D, was assessed51.

The primary outcome was a composite of a sustained decline in the estimated GFR of at least 50%, end-stage kidney disease, or death from renal or cardiovascular causes51.

It was found that, over a median of 2.4 years, CKD patients receiving dapagliflozin had a significantly lower risk of experiencing a primary outcome event than patients receiving placebo (9.2% vs 14.5%; HR 0.61; 95% CI, 0.51–0.72; P<0.001)51. Moreover, death occurred in fewer patients receiving dapagliflozin than patients receiving placebo (4.7% vs 6.8%; HR 0.69; 95% CI, 0.53 to 0.88; P=0.004)51.

 Empagliflozin

Empagliflozin is USFDA and EMA indicated to reduce the risk of cardiovascular death in adults with T2D, and to reduce the risk of cardiovascular death and hospitalisation for HF and reduced ejection fraction52,53.

The EMPA-REG OUTCOME study (N = 7,020) randomised patients with T2D to 10 or 25 mg of empagliflozin compared to placebo (Table 1). Of these patients, 5,665 were also receiving treatment with an ACEi or an ARB39

In T2D patients at high cardiovascular risk, empagliflozin is associated with slower kidney disease progression and lower rates of renal events than placebo when added to standard care39

The results showed that patients treated with empagliflozin had a reduced incidence in the combined endpoint of progression to macroalbuminuria, renal replacement therapy initiation or death from renal disease (HR [Hazard Ratio] 0.61; 95% CI, 0.53–0.70; P<0.001) compared to placebo39.

Patients in the intervention treatment arm also had lower rates of a post-hoc defined renal composite outcomes: initiation of renal replacement therapy, doubling of serum creatinine, or death from renal disease (HR 0.54; 95% CI, 0.40–0.75; P<0.001)39.

The EMPA-KIDNEY study, a double-blind, randomised, placebo-controlled trial, evaluated the efficacy and safety of empagliflozin in patients with CKD with or without diabetes. The trial, in which more than 6,600 patients were recruited, was stopped early, in March 2022, after an interim analysis of the results showed a clear benefit for patients with CKD54. Publication of the full results is pending.

Glucagon-like peptide 1 receptor agonists (GLP-1 RA) for diabetic kidney disease

A number of trials have investigated glucagon-like peptide 1 receptor agonists (GLP-1 RA) for the treatment of diabetes in chronic kidney disease (Table 2)25.

GLP-1 receptor agonists and DPP-4 inhibitors are beneficial for managing patients with T2D across a range of haemoglobin A1C (HbA1c) levels; separately and in addition to glucose lowering therapies55. Overall, GLP-1 receptor agonists are preferred over DPP-4 inhibitors due to a greater reduction in HbA1c and clinically significant weight loss55.

Liraglutide and semaglutide

In the randomised, placebo-controlled Liraglutide Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results (LEADER) trial (N = 9,350) and Evaluate Cardiovascular and Other Long-term Outcomes with Semaglutide in Subjects with Type 2 Diabetes (SUSTAIN-6) preapproval trial (N = 3,297), the effect of liraglutide and semaglutide on composite renal outcomes was assessed (Table 2)56,57:

  • Overt albuminuria development
  • Serum creatinine doubling and eGFR <45 mL/min/1.73 m2
  • Renal replacement therapy
  • Kidney disease-related death

Composite renal outcomes were decreased in the GLP-1 receptor agonist treatment group compared to placebo (LEADER: HR 0.88; 95% CI, 0.81–0.96; P=0.005 and SUSTAIN-6: HR 0.74; 95% CI, 0.62–0.89; P=0.002)56,57.

Table 2. Overview of selected large, placebo-controlled clinical outcome trials assessing the benefits and adverse effects of GLP-1 RA in chronic kidney disease (Adapted25). eGFR, estimated glomerular filtration rate; GFR, glomerular filtration rate; GI, gastrointestinal symptoms; MACE, major adverse cardiovascular events; NA, data not published. ↔, no significant difference; ↓, significant reduction in risk [hazard ratio (HR) estimate >0.7 and 95% confidence interval (CI) not overlapping 1]; ↓↓, significant reduction in risk [HR estimate ≤0.7 and 95% CI not overlapping 1].

T3_CKD_Table1b.png

Exenatide and lixisenatide

In the Exenatide Study of Cardiovascular Event Lowering (EXSCEL) study (N = 14,752), there was no significant difference (P=0.39) in eGFR levels between exenatide and placebo (Table 2). New macroalbuminuria was observed in 2.2% and 2.5% of the exenatide and placebo groups, respectively (P=0.19). The renal composite outcome (composite of 40% eGFR decline, renal replacement and renal death) was also not significantly different between the groups (HR 0.88, 95% CI, 0.74­–1.05; P=0.164)58.

In the Evaluation of Lixisenatide in Acute Coronary Syndrome (ELIXA) trial (N = 6,068), patients on lixisenatide were approximately 19% less likely to develop new-onset macroalbuminuria (HR 0.808; 95% CI, 0.660­–0.991; P=0.0404). No differences were observed in normoalbuminuria progression to macroalbuminuria or in the proportion of macroalbuminuria showing regression to micro-/normoalbuminuria (Table 2)59.

Dipeptidyl peptidase-4 inhibitors (DPP-4i) for diabetes

DPP-4 inhibitors, also known as gliptins, have been shown to have a low hypoglycaemic risk and weight-neutral effects. Moreover, saxagliptin, sitagliptin, alogliptin, and linagliptin have had their cardiovascular safety assessed in a number of randomised clinical trials25.

However, while DPP-4 inhibitors have displayed satisfactory cardiovascular safety, the results of their renal outcomes have been conflicting (Table 3)25,60.

Saxagliptin

In the Saxagliptin Assessment of Vascular Outcomes Recorded in Patients with Diabetes Mellitus-Thrombolysis in Myocardial Infarction 53 (SAVOR-TIMI 53) trial (N = 16,492), patients were randomised to saxagliptin or placebo (Table 3)61.

Compared to placebo, saxagliptin was more likely to improve the urine albumin-to-creatinine ratio (UACR) (10.7% vs 8.7%, P<0.001), and less likely to have a worsening UACR (13.3% vs 15.9%, P<0.001) at the end of treatment. Moreover, the difference in mean UAC between groups was –3.88 mg/mmol (P<0.004) at 2 years. However, the eGFR change from baseline was similar between groups, and there was no benefit or harm seen in hard renal outcomes61.

Alogliptin

In the Examination of Cardiovascular Outcomes with Alogliptin versus Standard of Care (EXAMINE) trial (N = 5,380) the primary end-point of a death from cardiovascular causes, non-fatal myocardial infarction, or non-fatal stroke composite was met in 11.3% patients receiving alogliptin (HR 0.96; P<0.001 for non-inferiority). Glycated haemoglobin levels were significantly lower with alogliptin than placebo (−0.36; P<0.001). Incidences of hypoglycaemia, cancer, pancreatitis, and initiation of dialysis were similar between groups (Table 3)62.

Table 3. Overview of selected large, placebo-controlled clinical outcome trials assessing the benefits and adverse effects of DPP-4i in chronic kidney disease (Adapted25). eGFR, estimated glomerular filtration rate; GFR, glomerular filtration rate; HF, heart failure hospitalisation; MACE, major adverse cardiovascular events; NA, data not published. ↔, no significant difference; ↓, significant reduction in risk [hazard ratio (HR) estimate >0.7 and 95% confidence interval (CI) not overlapping 1]; ↓↓, significant reduction in risk [HR estimate ≤0.7 and 95% CI not overlapping 1].

T3_CKD_Table1c.png

Sitagliptin

In the Trial Evaluating Cardiovascular Outcomes with Sitagliptin (TECOS) study (N = 14,671) median UACR levels at 4 years were lower with sitagliptin than placebo, with a mean difference of –0.02 mg/mmol (P=0.031). Mean eGFR was lower with sitagliptin compared to placebo with a difference of –1.34 ml/min/1.73 m2 (P<0.001). Incidence rates of microalbuminuria and renal failure were similar between sitagliptin and placebo (Table 3)63.

Linagliptin

The Cardiovascular and Renal Microvascular Outcome (CARMELINA) study (N = 6,991) had a higher proportion of participants (74%) with chronic kidney disease (Table 3)64.

The progression of albuminuria was 14% less likely with linagliptin compared to placebo (HR 0.86; 95% CI, 0.78–0.95; P=0.003). The hard renal outcomes were similar in the linagliptin and the placebo arms (9.4% vs 8.8%; HR 1.04; 95% CI, 0.89–1.22; P=0.62)64.

Can you pass our case study challenge? Test your knowledge and see if you can treat our virtual patient Thomas – a 61-year-old man with CKD with a long history of high blood pressure and smoking.

How well do you know CKD?

Managing end-stage kidney disease

Dr George Bakris discusses the main clinical challenges for preventing progression of chronic kidney disease.

End-stage kidney disease (ESKD), also known as end-stage renal disease (ESRD), occurs when chronic kidney disease reaches an advanced state and the kidneys are no longer able to meet the body's needs. As a result, various management strategies exist to manage comorbid conditions and minimise the burden on the patient.

When does kidney replacement therapy become necessary?

When ESRD is reached, either renal replacement therapy or conservative treatment will be needed. It is therefore important that early counselling is provided, by a nephrologist and a multidisciplinary team, on the available options:

  • Kidney transplant
  • Haemodialysis
  • Peritoneal dialysis
  • No dialysis

Early counselling is essential to prepare patients for kidney failure, while late referral is associated with a65:

  • worse health status at the initiation of kidney replacement therapy
  • higher mortality after starting dialysis
  • reduced access to transplant

Timely decision making can however be hampered by the unpredictability of CKD progression65,66. As CKD may not follow a steady linear decline, early pre­dialysis nephrology care for adults with late-stage CKD may be impacted and lead to compromised outcomes67.

The KDIGO guidelines recommend the initiation of dialysis when symptoms or signs of kidney failure are evident2. Prior to the initiation of dialysis, pre­emptive renal transplantation should be considered in patients with a GFR of <20 ml/min/1.73 m2 as well as evidence of CKD progression in the preceding 6–12 months2.

How do you control CKD-related complications?

A number of secondary complications are associated with CKD; in terms of overall mortality, the most notable of these complications is cardiovascular disease (CVD)68.

Guidelines, on the basis of diabetic status, aim to achieve target blood pressure through the use of RAS blockers, restricting salt intake, and preventing anaemia18,69

There have been a range of large randomised controlled trials of patients on haemodialysis that have assessed a range of interventions that specifically aim to reduce cardiovascular events; however, these were mostly unsuccessful70–72:

  • Frequency and length of dialysis sessions and flux
  • Erythropoietin stimulating agents
  • Statins
  • RAS blockade
  • Folic acid
  • Cinacalcet
  • Vitamin D derivatives

How do you prepare patients for haemodialysis for ESRD?

An important step in preparing patients for haemodialysis is the referral for vascular access placement due to the need for pumps, membranes, and dialysates to clear uraemic toxins from the blood1. Access can be gained through a variety of methods including1:

  • arteriovenous fistulae
  • arteriovenous grafts
  • central venous catheters (short-term use)

Of these methods, arteriovenous access is the preferred option for haemodialysis as it is, in addition to conversion from central venous catheter to arteriovenous access, associated with better outcomes than central venous catheters73–75. As a result, it is necessary to avoid venous puncture or intravenous catheter placement proximal to the wrist so that permanent vascular access is not inhibited1.

What is peritoneal dialysis for ESRD?

Clearing uraemic toxins from the blood can also be carried out by using peritoneal dialysis, a procedure that utilises the peritoneal membrane as an exchange interface. The procedure requires a transcutaneous catheter to be implanted into the peritoneal cavity to allow daily draining and refilling with dialysate fluid. Once equilibrium between uraemic blood and fresh dialysate is established, each dwell can drain excess fluid and metabolic waste products1.

In the first two years of dialysis, peritoneal dialysis shows improved outcomes and preservation of residual renal function compared to haemodialysis; however, differences in outcomes and preservation normalise after two years76.

What are the outcomes of kidney transplantation?

As kidney transplantation can produce superior outcomes to dialysis as a treatment for ESRD, it can be considered an essential component of an integrated treatment and management plan for CKD77. Pre-emptive transplantation may also be beneficial in patients with ESRD78.

In addition to age, the decision to perform a kidney transplantation is informed by the presence of comorbidities79, such as cancer, chronic infections, cardiac or peripheral vascular disease, and the risk of medical noncompliance1.

The half-life of a transplanted kidney is <20 years; patients will therefore likely continue needing CKD treatments during their life80

A number of complications can also occur following transplantation, such as recurrent glomerulonephritis; an unpredictable complication that can impact graft outcome81.

Rigorous testing is therefore required to improve the short-term and long-term health of the donor1. Eligibility of potential donors is assessed through a comprehensive health assessment that includes1:

  • blood group and human leukocyte antigen compatibility tests
  • GFR measures
  • imaging of the kidneys and the urinary tract
  • cardiac testing
  • specific tests informed by the medical history

When kidney replacement therapy is not an option, such as in cases of older patients with ESRD and comorbidities, dialysis may not increase lifespan or improve quality of life82–84. In these cases, palliative care aims to control the symptoms of uraemia that may impact quality of life85. Moreover, education should be provided to explain comorbidity management that may be needed or to discuss the decision to withdraw from dialysis86.

Help us create relevant CKD content for you

Chronic kidney disease treatment guidelines


Professor Vlado Perkovic outlines the recommendations for treating chronic kidney disease and drugs most recently approved that can preserve kidney function.

Explore the recommendations and available data for the diagnosis, management, and treatment of chronic kidney disease (CKD). Read the Kidney Disease: Improving Global Outcomes (KDIGO) guidelines on CKD evaluation and management as well as the management of diabetes and blood pressure in patients with CKD. Also find published guidelines from the National Institute for Health care and Excellence (NICE) and the American College of Physicians (ACP).

Kidney Disease: Improving Global Outcomes (KDIGO)

Originally established by the National Kidney Foundation, KDIGO is a global organisation for developing and implementing evidence based clinical practice guidelines in kidney disease.

KDIGO guidelines focus on topics related to kidney disease prevention and management of individuals with kidney diseases

Kidney Disease: Improving Global Outcomes guidelines

The criteria used in the guidelines to prioritise topics include:

  • burden of illness based on prevalence and scope of the condition or clinical problem
  • amenability of a particular condition to prevention or treatment and expected impact
  • existence of a body of evidence of sufficient breadth and depth to enable the development of evidence-based guidelines
  • potential of guidelines to reduce variations in practices, improve health outcomes, or lower treatment costs

KDIGO 2012 Clinical Practice Guideline for the Evaluation and Management of CKD

After a decade of innovative research, the KDIGO 2012 Clinical Practice Guideline for the Evaluation and Management of CKD updated the 2002 Kidney Disease Outcomes Quality Initiative (KDOQI) clinical practice guidelines for CKD87.

The guideline aims to give up to date guidance on the evaluation, management, and treatment of CKD patients. Specifically providing:

  • an enhanced classification framework for CKD
  • elaboration on the identification and prognosis of CKD
  • guidance on the management of progression and complications of CKD

The updated guideline also further develops the range of CKD care from the timings of specialist referral and dialysis initiation to ongoing management of progressive CKD and implementing a comprehensive treatment programme.

KDIGO guidelines for the Evaluation and Management of CKD

The guideline explores treatment approaches and recommendations based on systematic reviews of relevant clinical trials. It also provides practical comments and statements with important information for healthcare professionals; however, these are ungraded.

The quality of the evidence used in these guidelines, and the strength of recommendations, were appraised though the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach. Controversies, limitations, and international relevance are provided for discussion as well as suggestions for future research.

KDIGO 2020 Clinical Practice Guideline for Diabetes Management in CKD

The KDIGO 2020 Clinical Practice Guideline for Diabetes Management in CKD is the first KDIGO guideline specifically for diabetes management in CKD and includes topics such as:

  • comprehensive care
  • glycaemic monitoring and targets
  • lifestyle and antihyperglycemic interventions
  • approaches to self-management
  • optimal models of care

The aim of this guide is to be a useful resource of actionable recommendations for a broad audience of clinicians treating diabetes and CKD. It also aims to propose future research areas and discusses the limitations of the evidence.

KDIGO guidelines for Diabetes Management in CKD

The treatment approaches recommended in these guidelines followed an explicit process of evidence review and appraisal. They are also based on systematic reviews of relevant studies, appraisal of the quality of the evidence, and the strength of recommendations following the GRADE approach.

KDIGO 2021 Clinical Practice Guideline on the Management of BP in CKD

The KDIGO 2021 Clinical Practice Guideline on the Management of Blood Pressure (BP) in Chronic Kidney Disease (CKD) updates the 2012 KDIGO blood pressure (BP) guideline. It provides recommendations on the management of blood pressure for patients with non-dialysis CKD as well as recommendations for improving BP control, reducing cardiovascular disease risk in adults with CKD, and other important related topics.

KDIGO guidelines on the Management of BP in CKD

National Institute for Health care and Excellence (NICE) guideline

Last updated in 2015, the NICE guideline provided recommendations for the care and treatment of patients with, or at risk of developing, CKD. The guideline aims to help prevent or delay the progression of CKD as well as reduce or prevent the development of complications and the risk of cardiovascular disease.

NICE guidelines on chronic kidney disease in adults: assessment and management

American College of Physicians

The American College of Physicians (ACP) guideline provides evidence and clinical recommendations for the screening, monitoring, and treatment of adults with stage one to three CKD.

ACP guidelines on screening, monitoring, and treatment of Stage 1–3 chronic kidney disease

Future chronic kidney disease treatments

What is over the horizon? Dr George Bakris describes the range of current and future treatments and management strategies that may change the future of CKD care.

Key areas in the development of novel CKD treatments are arising due to a growing understanding of causes and consequences of CKD and the development and testing of new therapeutic strategies88. Currently, multiple investigational strategies are under investigation for the management of CKD.

Incretin-based treatments

While there is controversy with regards to the reduction of the GFR decrease, previous randomised control trials have established that incretin-based drugs can reduce albuminuria of diabetic kidney disease patients89.

Notably, albuminuria is associated with the GFR decrease in diabetic kidney disease and as a consequence, incretin-based drugs are likely to be effective. Moreover, incretin-based treatments are used currently as hypoglycaemic agents in diabetic patients89.

NF-E2–related factor 2 activators (Nrf2 activators)

Currently recommended treatments, such as RAS inhibitors and SGLT2 inhibitors, only slow GFR decline89. This has led to a heightened interest in NF-E2-related factor 2 (Nrf2) activators; a novel drug class that has the potential to improve the GFR of diabetic kidney disease patients90.

Notably, the phase III multicentre, placebo-controlled Bardoxolone Methyl in Patients with Diabetic Kidney Disease (AYAME) trial is currently ongoing in Japan (Figure 3). Similar to the previous TSUBAKI study, this study will assess the efficacy of bardoxolone methyl for diabetic kidney disease patients with the aim of elucidating any renoprotective effects89.

T3 CKD_Fig3.png

Figure 3. Clinical trials for incretin-based drugs, Nrf2 activators, AGE inhibitors, HIF-PH inhibitors, and epigenetic regulators (Adapted89). AGE, advanced glycation end product; HIF-PH, hypoxia-inducible factor prolyl hydroxylase inhibitor; NrF2, NF-E2–related factor 2; PYR, pyridoxamine.

Hypoxia-inducible factor prolyl hydroxylase inhibitors (HIF-PH)

Irreversible kidney damage can occur following a continued progression of CKD past a certain point. It is believed that a vicious cycle of tubular interstitial hypoxia is a common pathway for this progression91.

A novel treatment class that is currently being investigated are HIF prolyl hydroxylase (HIF-PH) inhibitors that inhibit HIF-α degradation by suppressing HIF-PH (Figure 3). HIF-PH is responsible for the oxygen-dependent degradation, stabilising of expression, and activation of HIF; a key transcription factor in the response to hypoxia92.

HIF-PH inhibitors have already been approved for renal anaemia in CKD patients not yet on dialysis. Future clinical trials will be needed to assess the efficacy of HIF-PH inhibitors as a CKD treatment89.

Advanced glycation end product inhibitors (AGE inhibitors)

Advanced glycation end product (AGE) is produced through non-enzymatic protein and nucleic acid glycation as a result of high blood sugar and oxidative stress93. As it accumulates it can cause damage to various organs and is associated with diabetic kidney disease progression94,95.

Clinical trials have previously been carried out to assess AGE inhibitors; however, their efficacy is controversial (Figure 3)89. A number of trials failed to show that AGE inhibitors are effective in treating diabetic kidney disease. Despite this, AGE inhibitors are still drawing attention due to the involvement of AGE accumulation in metabolic memory89.

Epigenetic regulators

Epigenetics is a DNA sequence-independent regulatory mechanism of gene expression, involving89:

  • DNA methylation
  • histone modification
  • non-coding RNA

Epigenetic changes caused by specific factors, such as hyperglycaemia or hypoxia, can be stored as cellular memory and may lead to irreversible renal damage96. For this reason, histone modification inhibitors have garnered attention as a novel treatment for these epigenetic changes (Figure 3).

The histone deacetylase inhibitors, such as vorinostat, valproate, sodium butyrate, and trichostatin, have been shown to reduce proteinuria and improve oxidative stress, fibrosis, glomerular damage, and inflammation in murine models97–101.

If the epigenetic changes in CKD are elucidated, this may increase the therapeutic options that can reverse CKD progression for CKD patients whose disease has progressed irreversibly past a certain point102.

Can you pass our case study challenge? Test your knowledge and see if you can treat our virtual patient Thomas – a 61-year-old man with CKD with a long history of high blood pressure and smoking.

How well do you know CKD?

References

  1. Romagnani P, Remuzzi G, Glassock R, Levin A, Jager KJ, Tonelli M, et al. Chronic kidney disease. Nat Rev Dis Prim. 2017;3.
  2. Kidney Disease: Improving Global Outcomes (KDIGO) CKD Work Group. KDIGO 2012 clinical practice guideline for the evaluation and management of chronic kidney disease. Kidney Int Suppl. 2013;3(1):1–150.
  3. Levey AS, Coresh J. Chronic kidney disease. Lancet. 2012;379(9811):165–180.
  4. Noris M, Remuzzi G. Glomerular Diseases Dependent on Complement Activation, Including Atypical Hemolytic Uremic Syndrome, Membranoproliferative Glomerulonephritis, and C3 Glomerulopathy: Core Curriculum 2015. Am J Kidney Dis. 2015;66(2):359–375.
  5. Hildebrand AM, Huang SHS, Clark WF. Plasma exchange for kidney disease: What is the best evidence? Adv Chronic Kidney Dis. 2014;21(2):217–227.
  6. Rauen T, Eitner F, Fitzner C, Sommerer C, Zeier M, Otte B, et al. Intensive Supportive Care plus Immunosuppression in IgA Nephropathy. N Engl J Med. 2015;373(23):2225–2236.
  7. Cravedi P, Ruggenenti P, Remuzzi G. Intensified inhibition of renin-angiotensin system: A way to improve renal protection? Curr Hypertens Rep. 2007;9(5):430–436.
  8. Holtkamp FA, De Zeeuw D, Thomas MC, Cooper ME, De Graeff PA, Hillege HJL, et al. An acute fall in estimated glomerular filtration rate during treatment with losartan predicts a slower decrease in long-term renal function. Kidney Int. 2011;80(3):282–287.
  9. Schrier RW, Abebe KZ, Perrone RD, Torres VE, Braun WE, Steinman TI, et al. Blood Pressure in Early Autosomal Dominant Polycystic Kidney Disease. N Engl J Med. 2014;371(24):2255–2266.
  10. Ruggenenti P, Cravedi P, Remuzzi G. Mechanisms and treatment of CKD. J Am Soc Nephrol. 2012;23(12):1917–1928.
  11. Lu JL, Molnar MZ, Naseer A, Mikkelsen MK, Kalantar-Zadeh K, Kovesdy CP. Association of age and BMI with kidney function and mortality: A cohort study. Lancet Diabetes Endocrinol. 2015;3(9):704–714.
  12. Daina E, Cravedi P, Alpa M, Roccatello D, Gamba S, Perna A, et al. A Multidrug, Antiproteinuric Approach to Alport Syndrome: A Ten-Year Cohort Study. Nephron. 2015;130(1):13–20.
  13. Nespoux J, Vallon V. Renal effects of SGLT2 inhibitors: An update. Curr Opin Nephrol Hypertens. 2020;29(2):190–198.
  14. Fioretto P, Zambon A, Rossato M, Busetto L, Vettor R. SGLT2 inhibitors and the diabetic kidney. Diabetes Care. 2016;39(Supplement 2):S165–S171.
  15. Alicic RZ, Rooney MT, Tuttle KR. Diabetic kidney disease: Challenges, progress, and possibilities. Clin J Am Soc Nephrol. 2017;12(12):2032–2045.
  16. Wheeler DC, Stefánsson B V., Jongs N, Chertow GM, Greene T, Hou FF, et al. Effects of dapagliflozin on major adverse kidney and cardiovascular events in patients with diabetic and non-diabetic chronic kidney disease: a prespecified analysis from the DAPA-CKD trial. Lancet Diabetes Endocrinol. 2021;9(1):22–31.
  17. Kidney Disease: Improving Global Outcomes (KDIGO) Diabetes Work Group. KDIGO 2012 Clinical Practice Guideline for the Evaluation and Management of Chronic Kidney Disease. Kidney Int Suppl. 2020;98(4S):S1–S115.
  18. Cheung AK, Chang TI, Cushman WC, Furth SL, Hou FF, Ix JH, et al. KDIGO 2021 Clinical Practice Guideline for the Management of Blood Pressure in Chronic Kidney Disease. Kidney Int. 2021;99(3):S1–S87.
  19. Kidney Disease: Improving Global Outcomes (KDIGO) Diabetes Work Group. KDIGO 2020 Clinical Practice Guideline for Diabetes Management in Chronic Kidney Disease. Kidney Int. 2020;98(4):S1–S115.
  20. Gæde P, Vedel P, Larsen N, Jensen GVH, Parving H-H, Pedersen O. Multifactorial Intervention and Cardiovascular Disease in Patients with Type 2 Diabetes. N Engl J Med. 2003;348(5):383–393.
  21. Mahmoodi BK, Matsushita K, Woodward M, Blankestijn PJ, Cirillo M, Ohkubo T, et al. Associations of kidney disease measures with mortality and end-stage renal disease in individuals with and without hypertension: A meta-analysis. Lancet. 2012;380(9854):1649–1661.
  22. Jankowski J, Floege J, Fliser D, Böhm M, Marx N. Cardiovascular Disease in Chronic Kidney Disease: Pathophysiological Insights and Therapeutic Options. Circulation. 2021;143:1157–1172.
  23. Cheung AK, Chang TI, Cushman WC, Furth SL, Hou FF, Ix JH, et al. Executive summary of the KDIGO 2021 Clinical Practice Guideline for the Management of Blood Pressure in Chronic Kidney Disease. Kidney Int. 2021;99(3):559–569.
  24. Strippoli GFM, Bonifati C, Craig M, Navaneethan SD, Craig JC. Angiotensin converting enzyme inhibitors and angiotensin II receptor antagonists for preventing the progression of diabetic kidney disease. Cochrane Database Syst Rev. 2006;18(4). doi:10.1002/14651858.CD006257.
  25. de Boer IH, Caramori ML, Chan JCN, Heerspink HJL, Hurst C, Khunti K, et al. Executive summary of the 2020 KDIGO Diabetes Management in CKD Guideline: evidence-based advances in monitoring and treatment. Kidney Int. 2020;98(4):839–848.
  26. Xie X, Liu Y, Perkovic V, Li X, Ninomiya T, Hou W, et al. Renin-Angiotensin System Inhibitors and Kidney and Cardiovascular Outcomes in Patients with CKD: A Bayesian Network Meta-analysis of Randomized Clinical Trials. Am J Kidney Dis. 2016;67(5):728–741.
  27. Ruggenenti P, Perna A, Gherardi G, Garini G, Zoccali C, Salvadori M, et al. Renoprotective properties of ACE-inhibition in non-diabetic nephropathies with non-nephrotic proteinuria. Lancet. 1999;354(9176):359–364.
  28. Remuzzi G, Ruggenenti P, Perna A, Mosconi L, Matalone M, Garini G, et al. Randomised placebo-controlled trial of effect of ramipril on decline in glomerular filtration rate and risk of terminal renal failure in proteinuric, non-diabetic nephropathy. Lancet. 1996;349(9069):1857–1863.
  29. Maschio G, Alberti D, Janin G, Locatelli F, Mann JFE, Motolese M, et al. Effect of the Angiotensin-Converting–Enzyme Inhibitor Benazepril on the Progression of Chronic Renal Insufficiency. N Engl J Med. 1996;334(15):939–945.
  30. Lewis EJ, Hunsicker LG, Clarke WR, Berl T, Pohl MA, Lewis JB, et al. Renoprotective Effect of the Angiotensin-Receptor Antagonist Irbesartan in Patients with Nephropathy Due to Type 2 Diabetes. N Engl J Med. 2001;345(12):851–860.
  31. Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving H-H, et al. Effects of Losartan on Renal and Cardiovascular Outcomes in Patients with Type 2 Diabetes and Nephropathy. N Engl J Med. 2001;345(12):861–869.
  32. Inzucchi SE, Lipska KJ, Mayo H, Bailey CJ, McGuire DK. Metformin in patientswith type 2 diabetes and kidney disease a systematic review. JAMA - J Am Med Assoc. 2014;312(24):2668–2675.
  33. De Jager J, Kooy A, Lehert P, Wulffelé MG, Van Der Kolk J, Bets D, et al. Long term treatment with metformin in patients with type 2 diabetes and risk of vitamin B-12 deficiency: Randomised placebo controlled trial. BMJ. 2010;340(7757):1177.
  34. US Food & Drugs Administration. Highlight of prescribing information. Kerendia®. 2021 www.fda.gov/medwatch. Accessed 12 October 2021.
  35. European Medicines Agency (EMA). Finerenone. 2019. https://www.ema.europa.eu/en/medicines/human/paediatric-investigation-plans/emea-001623-pip01-14-m03. Accessed 12 October 2021.
  36. Pitt B, Kober L, Ponikowski P, Gheorghiade M, Filippatos G, Krum H, et al. Safety and tolerability of the novel non-steroidal mineralocorticoid receptor antagonist BAY 94-8862 in patients with chronic heart failure and mild or moderate chronic kidney disease: A randomized, double-blind trial. Eur Heart J. 2013;34(31):2453–2463.
  37. Bakris GL, Agarwal R, Anker SD, Pitt B, Ruilope LM, Rossing P, et al. Effect of Finerenone on Chronic Kidney Disease Outcomes in Type 2 Diabetes. N Engl J Med. 2020;383(23):2219–2229.
  38. Pitt B, Filippatos G, Agarwal R, Anker SD, Bakris GL, Rossing P, et al. Cardiovascular Events with Finerenone in Kidney Disease and Type 2 Diabetes. N Engl J Med. 2021. doi:10.1056/nejmoa2110956.
  39. Wanner C, Inzucchi SE, Lachin JM, Fitchett D, von Eynatten M, Mattheus M, et al. Empagliflozin and Progression of Kidney Disease in Type 2 Diabetes. N Engl J Med. 2016;375(4):323–334.
  40. Perkovic V, de Zeeuw D, Mahaffey KW, Fulcher G, Erondu N, Shaw W, et al. Canagliflozin and renal outcomes in type 2 diabetes: results from the CANVAS Program randomised clinical trials. Lancet Diabetes Endocrinol. 2018;6(9):691–704.
  41. Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, et al. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. N Engl J Med. 2019;380(24):2295–2306.
  42. Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn A, et al. Dapagliflozin and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med. 2019;380(4):347–357.
  43. Tuttle KR, Brosius FC, Cavender MA, Fioretto P, Fowler KJ, Heerspink HJL, et al. SGLT2 Inhibition for CKD and Cardiovascular Disease in Type 2 Diabetes: Report of a Scientific Workshop Sponsored by the National Kidney Foundation. Am J Kidney Dis. 2021;77(1):94–109.
  44. European Medicines Agency. Forxiga®. 2021. https://www.ema.europa.eu/en/medicines/human/EPAR/forxiga. Accessed 12 October 2021.
  45. USFDA. Highlights of prescribing information. Farxiga®. 2021 www.fda.gov/medwatch. Accessed 8 October 2021.
  46. AstraZeneca. FARXIGA® (dapagliflozin) Prescribing Information . 2021. https://medicalinformation.astrazeneca-us.com/home/prescribing-information/farxiga-pi.html. Accessed 12 October 2021.
  47. USFDA. Highlights of prescribing Information. Invokana®. 2020 https://www.janssenlabels.com/package-insert/product-monograph/prescribing-information/INVOKANA-pi.pdf. Accessed 7 October 2021.
  48. European Medicines Agency. Summary of product characteristics. Invokana®. 2021 https://www.ema.europa.eu/en/documents/product-information/invokana-epar-product-information_en.pdf. Accessed 12 October 2021.
  49. Neal B, Perkovic V, Matthews DR, Mahaffey KW, Fulcher G, Meininger G, et al. Rationale, design and baseline characteristics of the CANagliflozin cardioVascular Assessment Study–Renal (CANVAS-R): A randomized, placebo-controlled trial. Diabetes, Obes Metab. 2017;19(3):387–393.
  50. Neuen BL, Ohkuma T, Neal B, Matthews DR, De Zeeuw D, Mahaffey KW, et al. Cardiovascular and renal outcomes with canagliflozin according to baseline kidney function data from the CANVAS program. Circulation. 2018;138(15):1537–1550.
  51. Heerspink HJL, Stefánsson B V., Correa-Rotter R, Chertow GM, Greene T, Hou F-F, et al. Dapagliflozin in Patients with Chronic Kidney Disease. N Engl J Med. 2020;383(15):1436–1446.
  52. US Food & Drugs Administration. Highlights of prescribing information. Jardiance®. 2021 https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/204629s026lbl.pdf. Accessed 12 October 2021.
  53. European Medicines Agency. Summary of product characteristics. Jardiance®. 2021. https://www.ema.europa.eu/en/documents/product-information/jardiance-epar-product-information_en.pdf. Accessed 12 October 2021.
  54. Empagliflozin trial halted after successful outcome in CKD. https://www.europeanpharmaceuticalreview.com/news/169452/empagliflozin-trial-halted-after-successful-outcome-in-chronic-kidney-disease/. Accessed 20 April 2022.
  55. Gilbert MP, Pratley RE. GLP-1 Analogs and DPP-4 Inhibitors in Type 2 Diabetes Therapy: Review of Head-to-Head Clinical Trials. Front Endocrinol (Lausanne). 2020;11:178.
  56. Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JFE, Nauck MA, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. Drug Ther Bull. 2016;54(9):101.
  57. Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jódar E, Leiter LA, et al. Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med. 2016;375(19):1834–1844.
  58. Holman RR, Bethel MA, Mentz RJ, Thompson VP, Lokhnygina Y, Buse JB, et al. Effects of Once-Weekly Exenatide on Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med. 2017;377(13):1228–1239.
  59. Pfeffer MA, Claggett B, Diaz R, Dickstein K, Gerstein HC, Køber L V., et al. Lixisenatide in Patients with Type 2 Diabetes and Acute Coronary Syndrome. N Engl J Med. 2015;373(23):2247–2257.
  60. Kawanami D, Takashi Y, Takahashi H, Motonaga R, Tanabe M. Renoprotective effects of dpp-4 inhibitors. Antioxidants. 2021;10(2):1–17.
  61. Špinar J, Šmahelová A. SAVOR-TINI53 - Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. Vnitr Lek. 2013;59(11):1003–1007.
  62. White WB, Cannon CP, Heller SR, Nissen SE, Bergenstal RM, Bakris GL, et al. Alogliptin after acute coronary syndrome in patients with type 2 diabetes. Austrian J Clin Endocrinol Metab. 2014;7(2):77.
  63. Green JB, Bethel MA, Armstrong PW, Buse JB, Engel SS, Garg J, et al. Effect of Sitagliptin on Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med. 2015;373(3):232–242.
  64. Rosenstock J, Perkovic V, Johansen OE, Cooper ME, Kahn SE, Marx N, et al. Effect of Linagliptin vs Placebo on Major Cardiovascular Events in Adults with Type 2 Diabetes and High Cardiovascular and Renal Risk: The CARMELINA Randomized Clinical Trial. JAMA - J Am Med Assoc. 2019;321(1):69–79.
  65. Sumida K, Kovesdy CP. Disease Trajectories Before ESRD: Implications for Clinical Management. Semin Nephrol. 2017;37(2):132–143.
  66. Tangri N, Grams ME, Levey AS, Coresh J, Appel LJ, Astor BC, et al. Multinational assessment of accuracy of equations for predicting risk of kidney failure ameta-analysis. JAMA - J Am Med Assoc. 2016;315(2):164–174.
  67. Ricardo AC, Roy JA, Tao K, Alper A, Chen J, Drawz PE, et al. Influence of Nephrologist Care on Management and Outcomes in Adults with Chronic Kidney Disease. J Gen Intern Med. 2016;31(1):22–29.
  68. Thomas B, Matsushita K, Abate KH, Al-Aly Z, Ärnlöv J, Asayama K, et al. Global cardiovascular and renal outcomes of reduced GFR. J Am Soc Nephrol. 2017;28(7):2167–2179.
  69. KIDGO Work Group. KDIGO clinical practice guideline for anemia in chronic kidney disease. Kidney Int Suppl. 2012;2(4):1–335.
  70. Baigent C, Landray MJ, Reith C, Emberson J, Wheeler DC, Tomson C, et al. The effects of lowering LDL cholesterol with simvastatin plus ezetimibe in patients with chronic kidney disease (Study of Heart and Renal Protection): A randomised placebo-controlled trial. Lancet. 2011;377(9784):2181–2192.
  71. Xu X, Qin X, Li Y, Sun D, Wang J, Liang M, et al. Efficacy of folic acid therapy on the progression of chronic kidney disease: The renal substudy of the China stroke primary prevention trial. JAMA Intern Med. 2016;176(10):1443–1450.
  72. Rossignol P, Agarwal R, Canaud B, Charney A, Chatellier G, Craig JC, et al. Cardiovascular outcome trials in patients with chronic kidney disease: Challenges associated with selection of patients and endpoints. Eur Heart J. 2019;40(11):880–886.
  73. Alencar de Pinho N, Coscas R, Metzger M, Labeeuw M, Ayav C, Jacquelinet C, et al. Vascular access conversion and patient outcome after hemodialysis initiation with a nonfunctional arteriovenous access: a prospective registry-based study. BMC Nephrol. 2017;18(1):1–11.
  74. Xue H, Ix JH, Wang W, Brunelli SM, Lazarus M, Hakim R, et al. Hemodialysis access usage patterns in the incident dialysis year and associated catheter-related complications. Am J Kidney Dis. 2013;61(1):123–130.
  75. Ravani P, Palmer SC, Oliver MJ, Quinn RR, MacRae JM, Tai DJ, et al. Associations between hemodialysis access type and clinical outcomes: A systematic review. J Am Soc Nephrol. 2013;24(3):465–473.
  76. Leurs P, Machowska A, Lindholm B. Timing of dialysis initiation: When to start? Which treatment? J Ren Nutr. 2015;25(2):238–241.
  77. O’Connell PJ, Brown M, Chan TM, Claure-Del Granado R, Davies SJ, Eiam-Ong S, et al. The role of kidney transplantation as a component of integrated care for chronic kidney disease. Kidney Int Suppl. 2020;10(1):e78–e85.
  78. Sébille V, Hardouin JB, Giral M, Bonnaud-Antignac A, Tessier P, Papuchon E, et al. Prospective, multicenter, controlled study of quality of life, psychological adjustment process and medical outcomes of patients receiving a preemptive kidney transplant compared to a similar population of recipients after a dialysis period of less than t. BMC Nephrol. 2016;17(1). doi:10.1186/s12882-016-0225-7.
  79. Abramowicz D, Cochat P, Claas FHJ, Heemann U, Pascual J, Dudley C, et al. European Renal Best Practice Guideline on kidney donor and recipient evaluation and perioperative care. Nephrology Dialysis Transplantation. 2015;30(11):1790–1797.
  80. Chang P, Gill J, Dong J, Rose C, Yan H, Landsberg D, et al. Living donor age and kidney allograft half-life: Implications for living donor paired exchange programs. Clin J Am Soc Nephrol. 2012;7(5):835–841.
  81. Allen PJ, Chadban SJ, Craig JC, Lim WH, Allen RDM, Clayton PA, et al. Recurrent glomerulonephritis after kidney transplantation: risk factors and allograft outcomes. Kidney Int. 2017;92(2):461–469.
  82. Verberne WR, Tom Geers ABM, Jellema WT, Vincent HH, van Delden JJM, Bos WJW. Comparative survival among older adults with advanced kidney disease managed conservatively versus with dialysis. Clin J Am Soc Nephrol. 2016;11(4):633–640.
  83. Morton RL, Webster AC, McGeechan K, Howard K, Murtagh FEM, Gray NA, et al. Conservative management and End-Of-Life care in an australian cohort with ESRD. Clin J Am Soc Nephrol. 2016;11(12):2195–2203.
  84. Carson RC, Juszczak M, Davenport A, Burns A. Is maximum conservative management an equivalent treatment option to dialysis for elderly patients with significant comorbid disease? Clin J Am Soc Nephrol. 2009;4(10):1611–1619.
  85. Crail S, Walker R, Brown M. Renal supportive and palliative care: Position statement. Nephrology. 2013;18(6):393–400.
  86. Birmelé B, François M, Pengloan J, Français P, Testou D, Brillet G, et al. Death after withdrawal from dialysis: The most common cause of death in a French dialysis population. Nephrol Dial Transplant. 2004;19(3):686–691.
  87. NKF-KDIGO GUIDELINE DEVELOPMENT STAFF. CKD Evaluation and Management – KDIGO. January 2013; Volume 3; Issue 1. 2012;1–150.
  88. Levin A, Tonelli M, Bonventre J, Coresh J, Donner JA, Fogo AB, et al. Global kidney health 2017 and beyond: a roadmap for closing gaps in care, research, and policy. Lancet. 2017;390(10105):1888–1917.
  89. Yamazaki T, Mimura I, Tanaka T, Nangaku M. Treatment of diabetic kidney disease: Current and future. Diabetes Metab J. 2021;45(1):11–26.
  90. Ito M, Tanaka T, Nangaku M. Nuclear factor erythroid 2-related factor 2 as a treatment target of kidney diseases. Curr Opin Nephrol Hypertens. 2020;29(1):128–135.
  91. Nangaku M. Chronic hypoxia and tubulointerstitial injury: A final common pathway to end-stage renal failure. J Am Soc Nephrol. 2006;17(1):17–25.
  92. Kapitsinou PP, Jaffe J, Michael M, Swan CE, Duffy KJ, Erickson-Miller CL, et al. Preischemic targeting of HIF prolyl hydroxylation inhibits fibrosis associated with acute kidney injury. Am J Physiol - Ren Physiol. 2012;302(9):1172–1179.
  93. Brownlee M, Vlassara H, Cerami A. Nonenzymatic glycosylation and the pathogenesis of diabetic complications. Ann Intern Med. 1984;101(4):527–537.
  94. Saulnier PJ, Wheelock KM, Howell S, Weil EJ, Tanamas SK, Knowler WC, et al. Advanced glycation end products predict loss of renal function and correlate with lesions of diabetic kidney disease in American indians with type 2 diabetes. Diabetes. 2016;65(12):3744–3753.
  95. Mallipattu SK, Uribarri J. Advanced glycation end product accumulation: A new enemy to target in chronic kidney disease? Curr Opin Nephrol Hypertens. 2014;23(6):547–554.
  96. Kato M, Natarajan R. Epigenetics and epigenomics in diabetic kidney disease and metabolic memory. Nat Rev Nephrol. 2019;15(6):327–345.
  97. Advani A, Huang Q, Thai K, Advani SL, White KE, Kelly DJ, et al. Long-term administration of the histone deacetylase inhibitor vorinostat attenuates renal injury in experimental diabetes through an endothelial nitric oxide synthase-dependent mechanism. Am J Pathol. 2011;178(5):2205–2214.
  98. Noh H, Eun YO, Ji YS, Mi RY, Young OK, Ha H, et al. Histone deacetylase-2 is a key regulator of diabetes- and transforming growth factor-β1-induced renal injury. Am J Physiol - Ren Physiol. 2009;297(3). doi:10.1152/ajprenal.00086.2009.
  99. Khan S, Jena G. Sodium butyrate, a HDAC inhibitor ameliorates eNOS, iNOS and TGF-β1-induced fibrogenesis, apoptosis and DNA damage in the kidney of juvenile diabetic rats. Food Chem Toxicol. 2014;73:127–139.
  100. Khan S, Jena G, Tikoo K. Sodium valproate ameliorates diabetes-induced fibrosis and renal damage by the inhibition of histone deacetylases in diabetic rat. Exp Mol Pathol. 2015;98(2):230–239.
  101. Sun XY, Qin HJ, Zhang Z, Xu Y, Yang XC, Zhao DM, et al. Valproate attenuates diabetic nephropathy through inhibition of endoplasmic reticulum stress-induced apoptosis. Mol Med Rep. 2016;13(1):661–668.
  102. Heiss EH, Schachner D, Werner ER, Dirsch VM. Active NF-E2-related factor (Nrf2) contributes to keep endothelial NO synthase (eNOS) in the coupled state: Role of reactive oxygen species (ROS), eNOS, and heme oxygenase (HO-1) levels. J Biol Chem. 2009;284(46):31579–31586.

Welcome: