7,581
Views
11
CrossRef citations to date
0
Altmetric
Original Articles

Effect of angiotensin receptor blockers on blood pressure and renal function in patients with concomitant hypertension and chronic kidney disease: a systematic review and meta-analysis

, , , , &
Pages 358-374 | Received 03 Jun 2019, Accepted 12 Jul 2019, Published online: 08 Aug 2019

Abstract

Objective: Angiotensin receptor blockers (ARB) are among the recommended first-line treatment options in patients with hypertension and chronic kidney disease (CKD). This meta-analysis evaluated the effect of ARB on blood pressure (BP) and renal function in patients with concomitant hypertension and CKD with or without diabetes.

Methods: Literature search was performed in PubMed/MEDLINE, EMBASE and BIOSIS to identify parallel-group, randomized controlled trials (≥8 weeks) reporting the effects of ARB on office systolic/diastolic BP (SBP/DBP), estimated glomerular filtration rate (eGFR), serum creatinine (SCr), creatinine clearance (CrCl) or proteinuria in adults with hypertension and CKD. Mean difference (MD, generic inverse variance) with 95% confidence intervals (CIs) was used to report an outcome.

Results: Among the 24 studies identified, 19 evaluated ARB as monotherapy, 4 evaluated ARB as combination therapy and one evaluated ARB both as monotherapy and combination therapy. Median (range) duration of the studies was 12 (1.84–54.0) months. ARB monotherapy significantly (p < 0.01) reduced BP (treatment ≥1 year: SBP [MD: −14.84 mmHg; 95% CI: −17.82 to −11.85]/DBP [−10.27 mmHg; −12.26 to −8.27]) and proteinuria (≥1 year [−0.90 g/L; −1.22 to −0.59]). Results were consistent for combination therapy. In these studies, non-significant changes were observed for eGFR, CrCl and SCr. The impact of SBP changes on eGFR was not significant; however, studies were of a relatively short duration.

Conclusion: ARB had a favorable impact on BP and renal parameters such as proteinuria with monotherapy as well as with combination therapy, highlighting their potential benefits in patients with hypertension and CKD. During the short follow-up of these studies, no significant change in eGFR was observed.

Introduction

Hypertension and chronic kidney disease (CKD) are global health issues [Citation1] with a strong cause and effect relationship [Citation2]. Both hypertension and CKD are associated with a high risk of cardiovascular (CV) morbidity and mortality [Citation3]. Hypertension together with proteinuria contributes to the progression of CKD [Citation4,Citation5], resulting in an increased CV and all-cause mortality [Citation6,Citation7].

Blood pressure (BP) control in patients with CKD reduces the likelihood of progression to end-stage renal disease (ESRD) and the occurrence of CV events [Citation8]. Therefore, guidelines recommend a target BP of ≤140/90 mmHg with careful monitoring of adverse events (AEs) in CKD patients with proteinuria <1 g/24 h and lower targets in those with proteinuria >1 g/24 h [Citation9–11]. Most recently, the 2017 American College of Cardiology/American Heart Association (ACC/AHA) clinical practice guidelines recommended an aggressive BP goal of <130/80 mmHg in all hypertensive patients including those with CKD [Citation12].

The renin–angiotensin–aldosterone system (RAAS) plays a pivotal role in controlling BP and renal function. Angiotensin II receptor blockers (ARB) and angiotensin-converting enzyme inhibitors (ACEIs) have been shown to be effective in lowering BP, slowing the progression of both diabetic and nondiabetic renal disease, reducing proteinuria, and reducing the risk of overt nephropathy [Citation13–21]. In addition, studies such as the Irbesartan in Patients with Type 2 Diabetes and Microalbuminuria study (IRMA-2), Irbesartan in Diabetic Nephropathy Trial (IDNT), Reduction of Endpoints in NIDDM with the Angiotensin II Antagonist Losartan (RENAAL) and Olmesartan Reducing Incidence of End Stage Renal Disease in Diabetic Nephropathy Trial (ORIENT) have demonstrated that the renoprotective effects of ARB in patients with diabetic nephropathy are partially independent of their antihypertensive effect [Citation22–25]. These renoprotective benefits beyond the BP-lowering effects support the recommendations to use ARB or ACEIs in patients with hypertension and CKD, particularly when they have proteinuria. Therefore, guidelines recommend initiating antihypertensive treatment with an ARB or ACEI, either as monotherapy or in combination with existing treatments, as first-line therapy for patients with CKD [Citation9,Citation11,Citation12,Citation26,Citation27]. Even in patients with advanced CKD, ARB and ACEIs are effective in delaying the disease progression [Citation28,Citation29].

Early meta-analysis and network meta-analysis [Citation30,Citation31] have demonstrated that blocking the renin–angiotensin system with ACEIs or ARB has beneficial effects on BP and proteinuria and may be the best approach to prevent ESRD in diabetic and nondiabetic CKD. The benefits of ACEIs or ARB on renal outcomes in placebo-controlled trials appear to result mainly from a BP-lowering effect, while additional renoprotective actions beyond lowering BP remain uncertain in diabetes [Citation30]. However, these meta-analyses included patients with or without hypertension and with or without reduced glomerular filtration rate (GFR) [Citation30,Citation31].

Therefore, we performed a systematic review and meta-analysis evaluating the effects of ARB on office BP, renal function, and proteinuria when prescribed as a monotherapy or in combination with other antihypertensives in patients presenting hypertension and CKD.

Methods

Systematic literature search strategy

We adopted the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines to perform this systematic review [Citation32]. A literature search from the earliest available date to July 2017 was performed in the PubMed/MEDLINE, EMBASE, and BIOSIS databases using MeSH terms (Emtree in case of EMBASE)/keywords: monotherapy, (“losartan” OR “eprosartan” OR “valsartan” OR “irbesartan” OR “tasosartan” OR “candesartan” OR “telmisartan” OR “olmesartan medoxomil” OR “azilsartan medoxomil” OR “fimasartan” OR (“angiotensin receptor blocker” OR “ARB”)) AND “hypertension” AND “chronic kidney disease;” dual therapy, (“losartan” OR “eprosartan” OR “valsartan” OR “irbesartan” OR “tasosartan” OR “candesartan” OR “telmisartan” OR “olmesartan medoxomil” OR “azilsartan medoxomil” OR “fimasartan” OR (“angiotensin receptor blocker” OR “ARB”)) AND (“calcium channel blocker” OR “diuretic”) AND “hypertension” AND “chronic kidney disease”. The literature search was limited to clinical trials (to restrict nonhuman studies such as preclinical, in vitro and in vivo) and English language to retrieve more relevant hits.

Inclusion and exclusion criteria

The studies that met the following criteria were included: (1) adult patients with hypertension and CKD with or without diabetes and who were treated with ARB; (2) parallel-group randomized controlled trials (RCTs) ≥8 weeks in duration; (3) studies that reported at least one of the following outcomes: systolic blood pressure (SBP), diastolic blood pressure (DBP), estimated GFR (eGFR), serum creatinine, creatinine clearance (CrCl) and proteinuria. Exclusion criteria included observational studies, crossover RCTs, studies not reported in English, or manuscripts without any full-text available. However, conference abstracts with relevant data for the aforementioned outcomes were included. We also excluded most studies, which included diabetic and nondiabetic patients with hypertension and CKD, when the results were not presented separately for patients with or without diabetes.

Parameters

The identified studies were analyzed for BP (SBP and DBP) and renal parameters (eGFR, CrCl, serum creatinine, and proteinuria).

Data extraction

The reviewers independently screened for potentially relevant article titles and abstracts based on the inclusion criteria. Whenever necessary, the full-text articles were also retrieved. The authors were independently involved in all stages of study selection and data extraction. Disagreements between reviewers, if any, were resolved by a discussion to obtain a consensus.

Assessment of risk of bias in included studies

The risk of bias of eligible trials, published as full-texts, was assessed using the Cochrane collaborations tool. The risk of bias tool covered selection bias, performance bias, detection bias, attrition bias, and reporting bias. We assigned judgments of low, unclear or high risk of bias under the following domains: random sequence generation (selection bias), allocation concealment (selection bias), blinding of participants and personnel (performance bias), blinding of outcome assessment (detection bias), incomplete outcome data (attrition bias), and selective reporting (reporting bias). Disagreements were resolved by consensus.

Data analysis

Meta-analysis (post-treatment versus pre-treatment) and meta-regression were carried out in R statistical software (version 3.4.1; https://www.r-project.org) using the meta and metafor packages. We used mean difference (MD, generic inverse variance [IV]) with 95% confidence intervals (CIs) to pool all available data for an outcome in a single forest plot. Serum creatinine values, where reported as µmol/L, were converted to mg/dL (1 mg/dL = 88.4 µmol/L) for analysis. The amount of heterogeneity was assessed by I2 test (0–40%: might not be important; 30–60%: may represent moderate heterogeneity; 50%–90%: may represent substantial heterogeneity; 75–100%: considerable heterogeneity) [Citation33]. However, a random-effects model was used in all outcomes as the selected studies differed in the mixes of participants, interventions and treatment duration [Citation34]. For analysis, post-pre standard deviation (SD) was converted to standard error (SE) [Citation35]. For studies that reported results only in graphical format, the numerical values were extracted from the graphs using Adobe® Reader® XI inbuilt measuring tool, version 11.0.06 (Adobe Systems Incorporated, San Jose, California, USA). Analysis was carried out for studies with durations ≥8 weeks to <1 year and ≥1 year. In all the analyses, a p-value <0.05 (two-tailed test) was considered statistically significant.

Results

Study selection and description of included studies

The electronic search retrieved 679 records; 415 unique records were screened after excluding 264 duplicate records. Of the 415 records screened, 165 full-text articles were assessed for eligibility and 24 studies met the inclusion criteria. There was no disagreement about the inclusion of studies among the authors. depicts the process of identifying relevant studies.

Figure 1. Flowchart for the identification of studies.

Figure 1. Flowchart for the identification of studies.

Characteristics of selected studies

The characteristics of the 24 studies [Citation22–25,Citation36–55] included in the meta-analysis are shown in . These studies were published between 1998 and 2017, with sample sizes ranging from 9 to 1715 subjects. The median duration of the studies was 12 months ranging between 1.84 months and 54.0 months. ARB as monotherapy were evaluated in 19 studies and 4 studies evaluated ARB in combination with other antihypertensive drugs. One study evaluated ARB both as monotherapy and combination therapy. Further, out of all the included studies, only five studies [Citation24,Citation25,Citation38,Citation40,Citation45], which were of 3 years duration, evaluated the effect of ARB on CV events, ESRD, and deaths; therefore, these parameters were not included in the present analysis.

Table 1. Characteristics of the studies included in this meta-analysis.

Antihypertensive effects of ARB in patients with hypertension and CKD

Effect on SBP

presents the effects of an ARB treatment on SBP in patients with hypertension and CKD. Overall results suggested that ARB as monotherapy or in combination with other antihypertensive agents significantly (p < 0.01) reduced the SBP. Monotherapy with an ARB for ≥8 weeks to <1 year resulted in a significant reduction of SBP (MD: −12.60 mmHg; 95% CI, −18.53 to −6.67; p < 0.01). Monotherapy with an ARB for ≥1 year also significantly decreased SBP (MD: −14.84 mmHg; 95% CI, −17.82 to −11.85; p < 0.01), and the reduction in SBP with ARB monotherapy for ≥1 year was numerically greater than that with ARB monotherapy for ≥8 weeks to <1 year (≥1 year versus ≥8 weeks to <1 year: MD, −2.24 mmHg; 95% CI, −8.88 to 4.40; p = 0.51). Moreover, combination treatment of ARB with diuretics (HCTZ) for ≥8 weeks to <1 year was also significant (p < 0.01) in reducing the SBP with a greater effect than ARB alone (MD: −18.00 mmHg; 95% CI, −20.86 to −15.14). ARB treatment in combination with calcium channel blockers (CCBs) or diuretics (hydrochlorothiazide [HCTZ]) for ≥1 year resulted in a significant (p < 0.01) reduction in SBP in patients with hypertension and CKD (MD: −13.07 mmHg; 95% CI, −18.93 to −7.22).

Figure 2. Effect of ARB on SBP reduction in patients with hypertension and CKD. *ARB (Olm, 20 mg; Los, 100 mg; Tel, 20 − 40 mg; Cand, 8 mg; Val, 80 − 160 mg). Aml: amlodipine; ARB: angiotensin receptor blocker; Azl: azelnidipine; Cand: candesartan; CCB: calcium channel blocker; CI: confidence interval; CKD: chronic kidney disease; HCTZ: hydrochlorothiazide; IV: inverse variance; Los: losartan; MD: mean difference; Olm: olmesartan; SBP: systolic blood pressure; SE: standard error; Tel: telmisartan; Val: valsartan.

Figure 2. Effect of ARB on SBP reduction in patients with hypertension and CKD. *ARB (Olm, 20 mg; Los, 100 mg; Tel, 20 − 40 mg; Cand, 8 mg; Val, 80 − 160 mg). Aml: amlodipine; ARB: angiotensin receptor blocker; Azl: azelnidipine; Cand: candesartan; CCB: calcium channel blocker; CI: confidence interval; CKD: chronic kidney disease; HCTZ: hydrochlorothiazide; IV: inverse variance; Los: losartan; MD: mean difference; Olm: olmesartan; SBP: systolic blood pressure; SE: standard error; Tel: telmisartan; Val: valsartan.

Effect on DBP

In patients with hypertension and CKD, ARB induced a significant (p < 0.01) reduction in DBP (). ARB monotherapy for ≥8 weeks to <1 year lowered the DBP by −6.52 mmHg (95% CI, −11.27 to −1.77; p < 0.01). The results were consistent for ARB monotherapy for ≥1 year with an MD of −10.27 mmHg (95% CI, −12.26 to −8.27; p < 0.01). However, the reduction in DBP observed with ≥1 year of ARB monotherapy was greater compared with ARB monotherapy for ≥8 weeks to <1 year (≥1 year versus ≥8 weeks to <1 year: MD, −3.75 mmHg; 95% CI, −8.90 to 1.40; p = 0.15). ARB in combination with HCTZ for ≥8 weeks to <1 year also demonstrated a significant reduction in the DBP (MD: −10.0 mmHg; 95% CI, −11.50 to −8.50; p < 0.01). When ARB are prescribed in combination with CCBs/diuretics (HCTZ) for ≥1 year, a significant reduction in DBP was also found (MD: −9.70 mmHg; 95% CI, −13.22 to −6.17; p < 0.01).

Figure 3. Effect of ARB on DBP reduction in patients with hypertension and CKD. *ARB (Olm, 20 mg; Los, 100 mg; Tel, 0 − 40 mg; Cand, 8 mg; Val, 80 − 160 mg). Aml: amlodipine; ARB: angiotensin receptor blocker; Azl: azelnidipine; Cand: candesartan; CCB: calcium channel blocker; CI: confidence interval; CKD: chronic kidney disease; DBP: diastolic blood pressure; HCTZ: hydrochlorothiazide; IV: inverse variance; Los: losartan; MD: mean difference; Olm: olmesartan; SE: standard error; Tel: telmisartan; Val: valsartan.

Figure 3. Effect of ARB on DBP reduction in patients with hypertension and CKD. *ARB (Olm, 20 mg; Los, 100 mg; Tel, 0 − 40 mg; Cand, 8 mg; Val, 80 − 160 mg). Aml: amlodipine; ARB: angiotensin receptor blocker; Azl: azelnidipine; Cand: candesartan; CCB: calcium channel blocker; CI: confidence interval; CKD: chronic kidney disease; DBP: diastolic blood pressure; HCTZ: hydrochlorothiazide; IV: inverse variance; Los: losartan; MD: mean difference; Olm: olmesartan; SE: standard error; Tel: telmisartan; Val: valsartan.

Effects of ARB on renal parameters in patients with hypertension and CKD

Effects on proteinuria

ARB treatment in patients with hypertension and CKD significantly lowered proteinuria (p < 0.01) (). ARB monotherapy for ≥8 weeks to <1 year reduced proteinuria by a mean of −0.60 g/L (95% CI, −0.93 to −0.26; p < 0.01). The results were consistent for ARB monotherapy for ≥1 year (MD: −0.90 g/L; 95% CI, −1.22 to −0.59; p < 0.01). The results were consistent for ≥8 weeks to <1 year of treatment with ARB in combination with HCTZ (MD: −1.40 g/L; 95% CI, −1.71 to −1.09; p < 0.01). Further, ARB in combination with CCBs/diuretics (HCTZ) for ≥1 year induced a significant reduction in proteinuria (MD: −0.33 g/L; 95% CI, −0.46 to −0.20; p < 0.01).

Figure 4. Effect of ARB on proteinuria in patients with hypertension and CKD. *ARB (Olm, 20 mg; Los, 100 mg; Tel, 20–40 mg; Cand, 8 mg; Val, 80–160 mg). Aml: amlodipine; ARB: angiotensin receptor blocker; Azl: azelnidipine; Cand: candesartan; CCB: calcium channel blocker; CI: confidence interval; CKD: chronic kidney disease; HCTZ: hydrochlorothiazide; IV: inverse variance; Los: losartan; MD: mean difference; Olm: olmesartan; SE: standard error; Tel: telmisartan; Val: valsartan.

Figure 4. Effect of ARB on proteinuria in patients with hypertension and CKD. *ARB (Olm, 20 mg; Los, 100 mg; Tel, 20–40 mg; Cand, 8 mg; Val, 80–160 mg). Aml: amlodipine; ARB: angiotensin receptor blocker; Azl: azelnidipine; Cand: candesartan; CCB: calcium channel blocker; CI: confidence interval; CKD: chronic kidney disease; HCTZ: hydrochlorothiazide; IV: inverse variance; Los: losartan; MD: mean difference; Olm: olmesartan; SE: standard error; Tel: telmisartan; Val: valsartan.

Effect on serum creatinine

Treatment with ARB in patients with hypertension and CKD did not induce any significant change in serum creatinine levels in the selected studies (). ARB monotherapy resulted in a nonsignificant increase in serum creatinine levels in patients treated for ≥8 weeks to <1 year (MD: 0.30 mg/dL; 95% CI, −0.69 to 1.29; p = 0.55) as well as in those treated for ≥1 year (MD: 0.02 mg/dL; 95% CI, −0.46 to 0.51; p = 0.92). Similarly, ARB in combination with CCBs/diuretics (HCTZ) for ≥1 year treatment duration also demonstrated non-significant increase in serum creatinine levels (MD: 0.01 mg/dL; 95% CI, −0.02 to 0.04; p = 0.48). However, treatment with ARB/diuretic (HCTZ) combination for ≥8 weeks to <1 year of treatment caused a significant (p < 0.01) increase in serum creatinine (MD: 0.20 mg/dL; 95% CI, 0.06 to 0.34) but the effect was modest and the observation was limited to one study of 50 patients.

Figure 5. Effect of ARB on serum creatinine levels in patients with hypertension and CKD. Aml: amlodipine; ARB: angiotensin receptor blocker; Cand: candesartan; CCB: calcium channel blocker; CI: confidence interval; CKD: chronic kidney disease; HCTZ: hydrochlorothiazide; IV: inverse variance; Los: losartan; MD: mean difference; SE: standard error; Tel: telmisartan; Val: valsartan.

Figure 5. Effect of ARB on serum creatinine levels in patients with hypertension and CKD. Aml: amlodipine; ARB: angiotensin receptor blocker; Cand: candesartan; CCB: calcium channel blocker; CI: confidence interval; CKD: chronic kidney disease; HCTZ: hydrochlorothiazide; IV: inverse variance; Los: losartan; MD: mean difference; SE: standard error; Tel: telmisartan; Val: valsartan.

Effect on eGFR and creatinine clearance

ARB as monotherapy or in combination with other antihypertensive drugs did not induce any significant change in either eGFR () or CrCl (Supplementary Figure S1). ARB monotherapy for ≥8 weeks up to <1 year or for ≥1 year was not associated with any significant improvement or deterioration of eGFR. This was also the case when ARB were combined with CCBs or diuretics (HCTZ).

Figure 6. Effect of ARB on eGFR in patients with hypertension and CKD. *ARB (Olm, 20 mg; Los, 100 mg; Tel, 20–40 mg; Cand, 8 mg; Val, 80–160 mg). Aml: amlodipine; ARB: angiotensin receptor blocker; Azl: azelnidipine; Cand: candesartan; CCB: calcium channel blocker; CI: confidence interval; CKD: chronic kidney disease; eGFR: estimated glomerular filtration rate; IV: inverse variance; HCTZ: hydrochlorothiazide; Los: losartan; MD: mean difference; Olm: olmesartan; SE: standard error; Tel: telmisartan; Val: valsartan.

Figure 6. Effect of ARB on eGFR in patients with hypertension and CKD. *ARB (Olm, 20 mg; Los, 100 mg; Tel, 20–40 mg; Cand, 8 mg; Val, 80–160 mg). Aml: amlodipine; ARB: angiotensin receptor blocker; Azl: azelnidipine; Cand: candesartan; CCB: calcium channel blocker; CI: confidence interval; CKD: chronic kidney disease; eGFR: estimated glomerular filtration rate; IV: inverse variance; HCTZ: hydrochlorothiazide; Los: losartan; MD: mean difference; Olm: olmesartan; SE: standard error; Tel: telmisartan; Val: valsartan.

Impact of treatment duration on SBP changes

Meta-regression analysis demonstrated no significant impact of ARB treatment duration on the SBP changes (estimate: 0.03; SE: 0.08; 95% CI, −0.14 to 0.19, p = 0.76; Supplementary Figure S2). A significant heterogeneity was observed among the studies (I2=95.04%).

Impact of SBP changes on eGFR

Meta-regression result demonstrated that there was no significant impact of SBP change on eGFR changes (estimate: 0.07; SE: 0.11: 95% CI, −0.14 to 0.28; p = 0.53; I2=8.96%; Supplementary Figure S3).

Risk of bias

The quality of the included studies was evaluated by the risk of bias assessment in the 22 full-text publications (Supplementary Figures S4 and S5). The overall risk of bias, across the six items of the Cochrane instrument, was judged to be low (Supplementary Figure S4). Within individual studies, however, two [Citation39,Citation41] had high-risk in random sequence generation and one [Citation39] in allocation concealment. In addition, in some studies, details of the methods for generating the random sequence [Citation36,Citation42,Citation44,Citation47–51,Citation53] and allocation concealment [Citation36,Citation41,Citation42,Citation44,Citation47–50,Citation55] were not provided, and outcome data were incomplete [Citation41,Citation44] (Supplementary Figure S5).

Discussion

The major observations of this meta-analysis of studies that investigated the BP and renal effects of ARB in patients with concomitant hypertension and CKD are the following: firstly, ARB given as monotherapy or prescribed in combination with CCBs/diuretics (HCTZ) significantly lowered BP; secondly, ARB induced a significant reduction in proteinuria; and thirdly, ARB were not associated with significant changes in eGFR.

There are two major components to slowing the rate of progression of CKD: (1) treatment of the underlying disease whenever possible, and (2) treatment of risk factors for progression, mainly systemic hypertension, which leads to glomerular hyperfiltration, and proteinuria [Citation56–62]. Treatment of hypertension in CKD patients is primarily aimed at achieving recommended BP targets by selecting the best class of drugs that have demonstrated antihypertensive efficacy and properties that may go above and beyond their BP-lowering effect. In this context, as proteinuria has been associated with the progression of renal disease in both nondiabetic and diabetic patients with CKD [Citation63,Citation64], the ability of hypertensive drugs to reduce proteinuria in addition to lowering BP, is another criteria for selecting drug treatments in CKD [Citation61]. Indeed, studies such as the Modification of Diet in Renal Disease Study (MDRD) [Citation65], and African American Study of Kidney Disease (AASK) [Citation66], demonstrated that higher baseline proteinuria was associated with a faster eGFR decline. Similarly, in the Ramipril Efficacy in Nephropathy (REIN) study, proteinuria was correlated with eGFR decline and progression of ESRD [Citation67]. Moreover, the RENAAL study conducted in patients with diabetic nephropathy reported baseline urine albumin-creatinine ratio as a strong independent predictor of ESRD [Citation68]. Furthermore, IDNT reported similar findings in diabetic nephropathy patients [Citation69]. For these many reasons, both European and American hypertension guidelines recommend the use of blockers of the renin-angiotensin system as first-line therapy in patients with hypertension and CKD [Citation11–12].

One objective of our meta-analysis was to evaluate the effect of ARB as monotherapy or in combination with other antihypertensive agents on systolic and diastolic BP in patients presenting strictly hypertension and CKD. Our results confirm that ARB, as monotherapy or in combination therapy, are effective in reducing both SBP and DBP with a slightly greater effect in patients treated for more than 1 year. Uncontrolled BP is a risk factor for worsening of renal function through an increase in the intraglomerular pressure and impaired glomerular filtration, leading ultimately to ESRD [Citation70–75]. Patients with CKD and a normal BP level have better preservation of GFR than hypertensive patients; lower BP targets (≤130/80 mmHg) are associated with better renal outcomes in patients with CKD and high proteinuria [Citation72]. Therefore, treatment of hypertensive patients with CKD is primarily aimed at BP control and limiting proteinuria to delay progression to ESRD [Citation75].

DBP has been reported to be significantly correlated with the progression of renal failure in patients with chronic glomerulonephritis; the progression rate of renal failure in patients with DBP <90 mmHg was significantly lower than that in patients with DBP >90 mmHg [Citation76]. In another study, it was reported that the improvement in DBP from 93 to 90 mmHg in patients with renal failure was associated with retardation in the progression of chronic renal failure [Citation77]. Furthermore, DBP <90 mmHg was associated with a slower rate and risk of progression to ESRD [Citation78,Citation79]. In another study, nisoldipine demonstrated a beneficial effect on the progression of renal failure in patients with renal insufficiency; in this study, there was no significant difference in SBP, but a significant reduction in DBP from 90 to 85 mmHg (P < 0.03) was reported [Citation79]. Therefore, the control of DBP (<90 mmHg) may be as important as SBP to preserve renal function or to retard the progression of chronic renal failure. The results of our meta-analysis demonstrated that ARB significantly reduced DBP in patients with hypertension and CKD, highlighting their potential benefits in these patients. However, one has to mention that the ideal systolic and diastolic BP to target in CKD remains controversial. Indeed, SBP levels below 110-120 mmHg have been associated with a worsening of renal function and with an increased mortality in patients with moderate to severe CKD, suggesting a J-curve [Citation81–83].

Proteinuria has been reported to be an independent factor in renal disease progression. Clinical trials have shown renoprotective effects of proteinuria reduction and suggest that antiproteinuric treatment maximizes renoprotection [Citation18,Citation62–64]. Results from the MDRD revealed a tight association between proteinuria and rate of eGFR decline [Citation65]. Renal protection achieved by lowering of BP depended primarily on the level of initial proteinuria [Citation67]. Secondary analysis of data from IDNT confirmed that baseline proteinuria is an important risk factor for renal failure in patients with type 2 diabetes and overt nephropathy [Citation69]. In the present meta-analysis, ARB as monotherapy at recommended and at maximal doses were found to reduce proteinuria in patients with hypertension and CKD. ARB monotherapy for 8 weeks to 1 year or >1 year demonstrated a significant reduction in proteinuria in patients with hypertension and CKD. Further, ARB in combination with other antihypertensive drugs (CCBs/diuretics) provided an even greater decrease in proteinuria. Treatment with ARB in combination with diuretics for >1 year demonstrated greater reduction in proteinuria than that for a treatment duration <1 year. These results suggest that the beneficial antiproteinuric effect of ARB in patients with hypertension and CKD is preserved and even amplified over time. ARB and ACEIs are known to be more effective than other antihypertensive drugs in reducing proteinuria mainly because they lower both systemic BP and the intraglomerular pressure vasodilating the efferent arterioles [Citation57]. The antiproteinuric effect of ARB has been demonstrated in patients with diabetic and nondiabetic CKD and is dose-dependent even when using very high doses, which do not lower BP more than conventional doses [Citation41,Citation84–87]. Thus, the Diovan Reduction Of Proteinuria (DROP) study reported that valsartan at its higher dose (640 mg/day) provided a greater reduction in microalbuminuria than the lower dose (160 mg/day) in hypertensive patients with type 2 diabetes mellitus and urinary albumin excretion rate of 20 − 700 µg/min [Citation87]. In the Strategies for Management of ART (SMART) trial, patients who received candesartan (128 mg/day) showed a significantly greater reduction (mean difference 33%) in proteinuria at 30 weeks than those who received 16 mg/day [Citation84] and very high doses of irbesartan (up to 900 mg/day) were found to more effective in lowering proteinuria than lower doses of 300 mg/day [Citation85]. A previous meta-analysis suggested that ARB treatment improved proteinuria over the short-to-medium term and prevented the progression of proteinuria/albuminuria over the medium term [Citation88]. Another meta-analysis compared the effects of monotherapy and combination therapy with RAAS blockers on proteinuria and concluded that the ARB reduced proteinuria, independent of the degree of proteinuria and underlying disease [Citation18].

Serum creatinine and the derived calculation of eGFR and CrCl are the common markers used to define CKD and to evaluate the progression of CKD towards ESRD. It has been well demonstrated that there is an acute renal hemodynamic effect following initiation of the RAAS blockade, which leads to an acute reduction in GFR. This phenomenon is due to the reduction in systemic and intraglomerular pressures. This initial decline is inversely correlated with renal function decline during the long-term follow-up; the greater the acute GFR fall, the slower the rate of long-term GFR decline and the risk of death [Citation89,Citation90]. The same is true for the early decline in proteinuria [Citation91]. In this meta-analysis, ARB as monotherapy or in combination with other antihypertensive agents did not significantly influence serum creatinine, eGFR, or CrCl, which may trigger the question of some stabilization of renal function. However, the impact of ARB on eGFR is highly variable. Further, in this meta-analysis, we did not observe any significant impact of the decrease in SBP on eGFR changes among patients with hypertension and CKD; However, one has to acknowledge that most studies were of too short duration to assess long-term changes in eGFR. Several large clinical trials performed essentially in patients with type 2 diabetes and CKD have demonstrated the ability of ARB to retard the progression towards ESRD without any significant impact on total mortality [Citation92].

Similar to other meta-analyses, this review was limited by the data (both quantity and data type) availability and accessibility. Rigid inclusion criteria were used for this meta-analysis that minimized the potential of bias during the selection process. Most of the studies included in this meta-analysis had small sample sizes and were conducted over a short duration (except five studies of 3 years duration), which limited our ability to evaluate the effects of ARB on CV events, ESRD, and deaths. The included studies were also of different treatment durations, which could have affected the treatment outcome. Different ARB were used in different studies, which increased the level of heterogeneity in this meta-analysis. Studies reporting the effect of ARB in combination with other antihypertensive agents were limited. Given the lack of consistency across studies in the collection of data, it has not been possible to analyze the relationships between BP, proteinuria, and eGFR.

In conclusion, this meta-analysis examined the effect of ARB on BP, proteinuria and renal function in patients with hypertension and CKD. ARB, as a monotherapy or in combination with other antihypertensive agents, demonstrated effective BP reduction and improved proteinuria in this patient population but without a clear effect on renal disease progression. These data suggest that ARB fulfill two important criteria to be recommended as first line therapy in hypertension and CKD. Few RCTs have shown the ability of ARB to retard the progression of nephropathy in type 2 diabetes but they did not affect total mortality significantly.

Supplemental material

Supplementary_File_ARBs_and_CKD_and_hypertension.docx

Download MS Word (988.9 KB)

Acknowledgments

The authors thank Jitendriya Mishra (Novartis Healthcare Pvt. Ltd., Hyderabad, India) for providing medical writing assistance on this manuscript.

Disclosure statement

MB has received conference fees and research supports from Menarini, Sankyo, Sanofi, Novartis, and Servier. GB is an employee of Novartis and is therefore entitled to receive Novartis stocks and stock options. PB is a Novartis consultant entitled to get Novartis shares and participate to stock option plans. The remaining authors declare that they have no conflict of interest.

Additional information

Funding

The analysis was funded by Novartis Pharma AG, Basel, Switzerland.

References

  • Horowitz B, Miskulin D, Zager P. Epidemiology of hypertension in CKD. Adv Chronic Kidney Dis. 2015;22:88–95.
  • Judd E, Calhoun DA. Management of hypertension in CKD: beyond the guidelines. Adv Chronic Kidney Dis. 2015;22:116–122.
  • National Kidney Foundation. K/DOQI clinical practice guidelines for chronic kidney disease: evaluation, classification, and stratification. Am J Kidney Dis. 2002;39:S76–S110.
  • Locatelli F, Marcelli D, Comelli M, et al. Proteinuria and blood pressure as causal components of progression to end-stage renal failure. Northern Italian Cooperative Study Group. Nephrol Dial Transplant. 1996;11:461–467.
  • Culleton BF, Larson MG, Parfrey PS, et al. Proteinuria as a risk factor for cardiovascular disease and mortality in older people: a prospective study. Am J Med. 2000;109:1–8.
  • Chronic Kidney Disease Prognosis Consortium, Matsushita K, van der Velde M, et al. Association of estimated glomerular filtration rate and albuminuria with all-cause and cardiovascular mortality in general population cohorts: a collaborative meta-analysis. Lancet. 2010;375:2073–2081.
  • Rashidi A, Sehgal AR, Rahman M, et al. The case for chronic kidney disease, diabetes mellitus, and myocardial infarction being equivalent risk factors for cardiovascular mortality in patients older than 65 years. Am J Cardiol. 2008;102:1668–1673.
  • Wenzel RR. Renal protection in hypertensive patients: selection of antihypertensive therapy. Drugs. 2005;65(Suppl 2):29–39.
  • Wheeler DC, Becker GJ. Summary of KDIGO guideline. What do we really know about management of blood pressure in patients with chronic kidney disease? Kidney Int. 2013;83:377–383.
  • Stevens PE, Levin A. Evaluation and management of chronic kidney disease: synopsis of the kidney disease: improving global outcomes 2012 clinical practice guideline. Ann Intern Med. 2013;158:825–830.
  • Williams B, Mancia G, Spiering W. et al. 2018 ESC/ESH Guidelines for the management of arterial hypertension: the task force for the management of arterial hypertension of the European Society of Cardiology and the European Society of Hypertension. J Hypertens. 2018;36:1953–2041.
  • Whelton PK, Carey RM, Aronow WS, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the prevention, detection, evaluation, and management of high blood pressure in adults. J Am Coll Cardiol. 2018;71:2199–2269.
  • Campbell RC, Ruggenenti P, Remuzzi G. Halting the progression of chronic nephropathy. J Am Soc Nephrol. 2002;13(Suppl 3):S190–S195.
  • Jafar TH, Schmid CH, Landa M, et al. Angiotensin-converting enzyme inhibitors and progression of non-diabetic renal disease. A meta-analysis of patient-level data. Ann Intern Med. 2001;135:73–87.
  • Viazzi F, Leoncini G, Pontremoli R. Antihypertensive treatment and renal protection: the role of drugs inhibiting the renin-angiotensin-aldosterone system High Blood Press Cardiovasc Prev. 2013;20:273–82.
  • Ruilope LM. Chronic kidney disease: Blood pressure control in CKD – still a matter of debate. Nat Rev Nephrol. 2013;9:572–573.
  • Ruggenenti P, Cravedi P, Remuzzi G. The RAAS in the pathogenesis and treatment of diabetic nephropathy. Nat Rev Nephrol. 2010;6:319–330.
  • Kunz R, Friedrich C, Wolbers M, et al. Meta-analysis: effect of monotherapy and combination therapy with inhibitors of the renin angiotensin system on proteinuria in renal disease. Ann Intern Med. 2008;148:30–48.
  • Hilgers KF, Mann JF. ACE inhibitors versus AT(1) receptor antagonists in patients with chronic renal disease. J Am Soc Nephrol. 2002;13:1100–1108.
  • Gansevoort RT, de Zeeuw D, de Jong PE. Is the antiproteinuric effect of ACE inhibition mediated by interference in the renin-angiotensin system? Kidney Int. 1994;45:861–867.
  • Remuzzi A, Perico N, Sangalli F, et al. ACE inhibition and ANG II receptor blockade improve glomerular size-selectivity in IgA nephropathy. Am J Physiol. 1999;276:F457–F466.
  • Parving HH, Lehnert H, Bröchner-Mortensen J, et al. The effect of Irbesartan on the development of diabetic nephropathy in patients with type 2 diabetes. N Engl J Med. 2001;345:870–878.
  • Lewis E, Hunsicker LG, Clarke WR, et al. Renoprotective effect of the angiotensin-receptor antagonist irbesartan in patients with nephropathy due to type 2 diabetes. New Engl J Med. 2001;345:851–860.
  • Brenner BM, Cooper ME, de Zeeuw D, et al. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med. 2001;345:861–869.
  • 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;54:2978–2986.
  • Dasgupta K, Quinn RR, Zarnke KB, et al. The 2014 Canadian Hypertension Education Program recommendations for blood pressure measurement, diagnosis, assessment of risk, prevention, and treatment of hypertension. Can J Cardiol. 2014;30:485–501.
  • Shimamoto K; Ando K; Fujita T, et al. The Japanese Society of Hypertension Guidelines for the Management of Hypertension (JSH 2014). Hypertens Res. 2014;37:253–390.
  • Hou FF, Zhang X, Zhang GH, et al. Efficacy and safety of benazepril for advanced chronic renal insufficiency. N Engl J Med. 2006;354:131–140.
  • Yasuda T, Endoh M, Suzuki D, et al. Effects of valsartan on progression of kidney disease in Japanese hypertensive patients with advanced, predialysis, chronic kidney disease: Kanagawa Valsartan Trial (KVT). Hypertens Res. 2013;36:240–246.
  • Casas JP, Chua W, Loukogeorgakis S, et al. Effect of inhibitors of the renin-angiotensin system and other antihypertensive drugs on renal outcomes: systematic review and meta-analysis. Lancet. 2005;366:2026–2033.
  • Palmer SC, Mavridis D, Navarese E et al. Comparative efficacy and safety of blood pressure-lowering agents in adults with diabetes and kidney disease: a network meta-analysis. Lancet. 2015;385:2047–2056
  • Liberati A, Altman DG, Tetzlaff J, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. J Clin Epidemiol. 2009;62:e1–e34.
  • Higgins JPT, Green S (eds). Cochrane handbook for systematic reviews of interventions version 5.1.0 [updated March 2011]. The Cochrane Collaboration; 2011. http://handbook.cochrane.org.
  • Borenstein M, Hedges LV, Higgins JPT, Rothstein HR (eds). Introduction to Meta-analysis. Chichester (West Sussex, UK): John Wiley & Sons, Inc; 2009.
  • Morris SB, DeShon RP. Combining effect size estimates in meta-analysis with repeated measures and independent-groups designs. Psychol Methods. 2002;7:105–125.
  • Zinellu A, Sotgia S, Mangoni AA, et al. Effects of ramipril and telmisartan on plasma concentrations of low molecular weight and protein thiols and carotid intima media thickness in patients with chronic kidney disease. Dis Markers. 2016; 2016:1821596.
  • Nakamura T, Inoue T, Suzuki T, et al. Comparison of renal and vascular protective effects between telmisartan and amlodipine in hypertensive patients with chronic kidney disease with mild renal insufficiency. Hypertens Res. 2008;31:841–850.
  • Nakamura T, Kanno Y, Takenaka T, et al. An angiotensin receptor blocker reduces the risk of congestive heart failure in elderly hypertensive patients with renal insufficiency. Hypertens Res. 2005;28:415–423.
  • Sharma AM, Hollander A, Köster J. Telmisartan in patients with mild/moderate hypertension and chronic kidney disease. Clin Nephrol. 2005;63:250–257.
  • Hou FF, Xie D, Zhang X, et al. Renoprotection of Optimal Antiproteinuric Doses (ROAD) Study: a randomized controlled study of benazepril and losartan in chronic renal insufficiency. J Am Soc Nephrol. 2007;18:1889–1898.
  • Aranda P, Segura J, Ruilope LM, et al. Long-term renoprotective effects of standard versus high doses of telmisartan in hypertensive nondiabetic nephropathies. Am J Kidney Dis. 2005;46:1074–1079.
  • Uneda K, Tamura K, Wakui H, et al. Comparison of direct renin inhibitor and angiotensin II receptor blocker on clinic and ambulatory blood pressure profiles in hypertension with chronic kidney disease. Clin Exp Hypertens. 2016;38:738–743.
  • Kaneshiro Y, Ichihara A, Sakoda M, et al. Add-on benefits of amlodipine and thiazide in nondiabetic chronic kidney disease stage 1/2 patients treated with valsartan. Kidney Blood Press Res. 2009;32:51–58.
  • Antlanger M, Bernhofer S, Kovarik JJ, et al. Effects of direct renin inhibition versus angiotensin II receptor blockade on angiotensin profiles in non-diabetic chronic kidney disease. Ann Med. 2017;49:525–533.
  • Woo KT, Choong HL, Wong KS, et al. Aliskiren and losartan trial in non-diabetic chronic kidney disease. J Renin Angiotensin Aldosterone Syst. 2014;15:515–522.
  • Takenaka T, Seto T, Okayama M, et al. Long-term effects of calcium antagonists on augmentation index in hypertensive patients with chronic kidney disease: a randomized controlled study. Am J Nephrol. 2012;35:416–423.
  • Nakamura T, Inoue T, Sugaya T, et al. Renoprotective effect of telmisartan in patients with chronic kidney disease. Clin Exp Hypertens. 2008;30:662–672.
  • Crowe AV, Howse M, Vinjamuri S, et al. The antiproteinuric effect of losartan is systemic blood pressure dependent. Nephrol Dial Transplant. 2003;18:2160–2164.
  • Matsuda H, Hayashi K, Saruta T. Distinct time courses of renal protective action of angiotensin receptor antagonists and ACE inhibitors in chronic renal disease. J Hum Hypertens. 2003;17:271–276.
  • Matsuda H, Hayashi K, Homma K, et al. Differing anti-proteinuric action of candesartan and losartan in chronic renal disease. Hypertens Res. 2003;26:875–880.
  • Plum J, Bunten B, Nemeth R, et al. Effects of the angiotensin II antagonist valsartan on blood pressure, proteinuria, and renal hemodynamics in patients with chronic renal failure and hypertension. J Am Soc Nephrol. 1998;9:2223–2234.
  • Tsygankova OV, Platonov D, Bondareva ZG, et al. Do early and late nephroprotective effects differ with different inhibitors of renin-angiotensin-aldosteron system in chronic heart failure patients? Eur Heart J. 2012;33(suppl. 1):808–809.
  • Nakamura T, Fujiwara N, Kawagoe Y, et al. Effects of telmisartan and enalapril on renoprotection in patients with mild to moderate chronic kidney disease. Eur J Clin Invest. 2010;40:790–796.
  • Sowers J, Giles T, Ofili E, et al. Intensive treatment with combination amlodipine/valsartan vs moderate therapy for hypertensive patients with diabetes or CKD uncontrolled on ARB monotherapy. J Hypertens. 2010;28(Suppl A):E295 (abstract PP.17.149).
  • Praga M, Fernández Andrade C, Luño J, et al. Antiproteinuric efficacy of losartan in comparison with amplodipine in non-diabetic proteinuric renal diseases: a double-blind, randomized clinical trial. Nephrol Dial Transplant. 2003;18:1806–1813.
  • Levey AS, de Jong PE, Coresh J, et al. The definition, classification, and prognosis of chronic kidney disease: a KDIGO Controversies Conference report. Kidney Int. 2011;80:17–28.
  • Remuzzi G, Ruggenenti P, Perico N. Chronic renal diseases: renoprotective benefits of renin-angiotensin system inhibition. Ann Intern Med. 2002;136:604–615.
  • Sarafidis PA, Khosla N, Bakris GL. Antihypertensive therapy in the presence of proteinuria. Am J Kidney Dis. 2007;49:12–26.
  • Weir MR. Progressive renal and cardiovascular disease: optimal treatment strategies. Kidney Int. 2002;62:1482–1492.
  • Henry TY. Progression of chronic renal failure. Arch Intern Med. 2003;163:1417–1429.
  • Perico N, Benigni A, Remuzzi G. Present and future drug treatments for chronic kidney diseases: evolving targets in renoprotection. Nat Rev Drug Discov. 2008;7:936–953.
  • Cheung AK, Rahman M, Reboussin DM, et al. Effects of Intensive BP Control in CKD. J Am Soc Nephrol. 2017;28:2812–2823.
  • van den Belt SM, Heerspink HJL, Gracchi V, et al. Early proteinuria lowering by angiotensin-converting enzyme inhibition predicts renal survival in children with CKD. J Am Soc Nephrol. 2018;29:2225–2233.
  • Heerspink HJ, Kröpelin TF, Hoekman J, et al. Reducing albuminuria as surrogate endpoint (REASSURE) consortium. J Am Soc Nephrol. 2015;26:2055–2064.
  • Peterson JC, Adler S, Burkart JM, et al. Blood pressure control, proteinuria, and the progression of renal disease. The Modification of Diet in Renal Disease Study. Ann Intern Med. 1995;123:754–762.
  • Wright TJ Jr, Bakris G, Greene T, et al. Effect of blood pressure lowering and antihypertensive drug class on progression of hypertensive kidney disease: results from the AASK trial. JAMA. 2002;288:2421–2431.
  • The GISEN Group (Gruppo Italiano di Studi Epidemiologici in Nefrologia). 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. 1997;349:1857–1863.
  • Keane WF, Zhang Z, Lyle PA, et al. Risk scores for predicting outcomes in patients with type 2 diabetes and nephropathy: the RENAAL study. Clin J Am Soc Nephrol. 2006;1:761–767.
  • Atkins RC, Briganti EM, Lewis JB, et al. Proteinuria reduction and progression to renal failure in patients with type 2 diabetes mellitus and overt nephropathy. Am J Kidney Dis. 2005;45:281–287.
  • Hamrahian SM. Management of hypertension in patients with chronic kidney disease. Curr Hypertens Rep. 2017;19:43.
  • Klag MJ, Whelton PK, Randall BL, et al. Blood pressure and end-stage renal disease in men. 1996;334:13–18.
  • Anderson AH, Yang W, Townsend RR, et al. Time-updated systolic blood pressure and the progression of chronic kidney disease: a cohort study. Ann Intern Med. 2015;162:258–265.
  • Sim JJ, Shi J, Kovesdy CP, et al. Impact of achieved blood pressures on mortality risk and end-stage renal disease among a large, diverse hypertension population. J Am Coll Cardiol. 2014; 64:588–597.
  • Agarwal R. Blood pressure components and the risk for end-stage renal disease and death in chronic kidney disease. Clin J Am Soc Nephrol. 2009;4:830–837.
  • Kidney Disease Outcomes Quality Initiative (K/DOQI). K/DOQI clinical practice guidelines on hypertension and antihypertensive agents in chronic kidney disease. Am J Kidney Dis. 2004;43(5 Suppl 1):S1–S290.
  • Shimamatsu K, Onoyama K, Harada A, et al. Effect of blood pressure on the progression rate of renal impairment in chronic glomerulonephritis. J Clin Hypertens. 1985;1:239–244.
  • Bergström J, Alvestrand A, Bucht H, et al. Progression of chronic renal failure in man is retarded with more frequent clinical follow-ups and better blood pressure control. Clin Nephrol. 1985;25:1–6.
  • Brazy PC, Stead WW, Fitzwilliam JF. Progression of renal insufficiency: role of blood pressure. Kidney Int. 1989;35:670–674.
  • Peralta CA, Norris KC, Li S, et al. Blood pressure components and end-stage renal disease in persons with chronic kidney disease: the Kidney Early Evaluation Program (KEEP).Arch Intern Med. 2012;172:41–47.
  • Eliahou HE, Cohen D, Hellberg B, Ben-David A, et al. Effect of the calcium channel blocker nisoldipine on the progression of chronic renal failure in man. Am J Nephrol. 1988;8:285–290.
  • Kovesdy CP, Trivedi BK, Kalantar-Zadeh K, et al. Association of low blood pressure with increased mortality in patients with moderate to severe chronic kidney disease. Nephrol Dial Transplant. 2006;21:1257–1262.
  • Upadhyay A, Earley A, Haynes SM, et al. Systematic review: blood pressure target in chronic kidney disease and proteinuria as an effect modifier. Ann Intern Med. 2011;154:541–548.
  • Flynn C, Bakris GL. Blood pressure targets for patients with diabetes or kidney disease. Curr Hypertens Rep. 2011;13:452–455.
  • Schmieder RE, Klingbeil AU, Fleischmann EH, et al. Additional antiproteinuric effect of ultrahigh dose candesartan: a double-blind, randomized, prospective study. J Am Soc Nephrol. 2005;16:3038–3045.
  • Rossing K, Schjoedt KJ, Jensen BR, et al. Enhanced renoprotective effects of ultrahigh doses of irbesartan in patients with type 2 diabetes and microalbuminuria. Kidney Int. 2005;68:1190–1198.
  • Burgess E, Muirhead N, Rene de Cotret P, et al. Supramaximal dose of candesartan in proteinuric renal disease. J Am Soc Nephrol. 2009;20:893–900.
  • Hollenberg NK, Parving HH, Viberti G, et al. Albuminuria response to very high-dose valsartan in type 2 diabetes mellitus. J Hypertens. 2007;25:1921–1926.
  • Takagi H, Yamamoto H, Iwata K, et al. Effects of telmisartan on proteinuria or albuminuria: a meta-analysis of randomized trials. Int J Cardiol. 2013;167:1443–1449.
  • Holtkamp FA, de Zeeuw D, Thomas MC, 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;80:282–287.
  • Coresh J, Turin TC, Matsushita K, et al. Decline in estimated glomerular filtration rate and subsequent risk of end-stage renal disease and mortality. JAMA. 2014;311:2518–2531.
  • Wapstra FH, Navis G, de Jong PE, et al. Prognostic value of the short-term antiproteinuric response to ACE inhibition for prediction of GFR decline in patients with nondiabetic renal disease. Exp Nephrol. 1996;4:47–52.
  • Strippoli GF, Craig M, Deeks JJ, et al. Effects of angiotensin converting enzyme inhibitors and angiotensin II receptor antagonists on mortality and renal outcomes in diabetic nephropathy: systematic review. BMJ. 2004;329:828.