Electrolyte Blood Press. 2022 Dec;20(2):39-48. English.
Published online Dec 30, 2022.
Copyright © 2022 Korean Society for Electrolyte and Blood Pressure Research
Review

Blood Pressure Control in Patients with Diabetic Kidney Disease

Yaeni Kim,1 Won Kim,2 Jwa-Kyung Kim,3 Ju Young Moon,4 Samel Park,5 Cheol Whee Park,1 Hoon Suk Park,6 Sang Heon Song,7 Tae-Hyun Yoo,8 So-Young Lee,9 Eun Young Lee,5 Jeonghwan Lee,10 Kyubok Jin,11 Dae Ryong Cha,12 Jin Joo Cha,12 Sang Youb Han,13 and On behalf of the Korean Diabetic Kidney Disease Working Group
    • 1Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University, Seoul, Republic of Korea.
    • 2Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju, Republic of Korea.
    • 3Department of Internal Medicine & Kidney Research Institute, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea.
    • 4Division of Nephrology, Department of Internal Medicine, Kyung Hee University, Seoul, Republic of Korea.
    • 5Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Republic of Korea.
    • 6Division of Nephrology, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, The Catholic University, Seoul, Republic of Korea.
    • 7Department of Internal Medicine & Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea.
    • 8Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, Republic of Korea.
    • 9Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea.
    • 10Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea.
    • 11Department of Internal Medicine, Keimyung University School of Medicine, Keimyung University Kidney Institute, Daegu, Republic of Korea.
    • 12Department of Internal Medicine, Korea University Ansan Hospital, Ansan, Republic of Korea.
    • 13Department of Internal Medicine, Inje University College of Medicine, Ilsan-Paik Hospital, Goyang, Republic of Korea.
Received October 26, 2022; Revised November 21, 2022; Accepted December 05, 2022.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Diabetic kidney disease (DKD) is the most common cause of end-stage kidney disease. Blood pressure (BP) control can reduce the risks of cardiovascular (CV) morbidity, mortality, and kidney disease progression. Recently, the Kidney Disease: Improving Global Outcomes (KDIGO) guidelines have suggested the implementation of a more intensive BP control with a target systolic BP (SBP) of <120 mmHg based on the evidence that the CV benefits obtained is outweighed by the kidney injury risk associated with a lower BP target. However, an extremely low BP level may paradoxically aggravate renal function and CV outcomes. Herein, we aimed to review the existing literature regarding optimal BP control using medications for DKD.

Keywords
Blood pressure; Diabetes; Kidney; Mortality

INTRODUCTION

Diabetic kidney disease (DKD) is the most common cause of end-stage kidney disease (ESKD) worldwide1). According to the Korean ESKD registry (Korean Renal Data System, KORDS), its incidence has exponentially increased from 10% in 1985 to 49.8% in 20202). The initial clinical manifestations of DKD include glomerular hyperfiltration, albuminuria, and arterial hypertension, which eventually lead to a lower estimated glomerular filtration rate (eGFR)3). The progression of chronic kidney disease (CKD) is considerably faster in patients with diabetes than that in patients without diabetes4).

The renin-angiotensin-aldosterone system(RAAS) is the most important treatment target. RAAS activation increases the glomerular capillary hydraulic pressure and disrupts the renal vascular autoregulation, causing further renal damage5). Hence, rigorous control of RAAS activation is crucial for blood pressure (BP) and urinary albumin level control in DKD management.

Recent clinical studies have shown that strict BP control can reduce cardiovascular (CV) morbidity and mortality rates, as well as the progression of kidney diseases. However, a considerable decrease in BP levels may paradoxically aggravate the renal damage6, 7). Accordingly, when determining the optimal BP control for the prevention of CV events and all-cause death, the renal function status and risk of ESKD should be taken into consideration. Currently, the target BP for patients with diabetes is frequently used, as no randomized controlled trials (RCTs) have determined the target BP for patients with DKD.

A multidisciplinary approach is recommended to delay the deterioration of renal function in patients with DKD. Although this study examined the antihypertensive medications used in patients with DKD, lifestyle modifications, including a low-salt diet, exercise, smoking cessation, and weight control, require prioritization. In addition, recent clinical trials on the use of sodium-glucose cotransporter 2 inhibitors, finerenone, and selective endothelin A receptor antagonists have shown promising results. Sodium-glucose cotransporter 2 inhibitors and finerenone have been recommended in the latest Kidney Disease Improving Global Outcomes (KDIGO) guidelines8). Although these agents are beyond the scope of this review, their use may attenuate the progression of renal disease in patients with type 2 diabetes. Hence, we aimed to review the existing literature on optimal BP control using medications in these patients.

Renal hemodynamics in diabetic kidney disease: The relationship between albuminuria, hypertension, and renal function

In patients with type 1 diabetes, hypertension develops concurrently with albuminuria or overt nephropathy; in patients with type 2 diabetes, hypertension often precedes albuminuria, and the eGFR declines4, 9). Hypertension coexists with other CV risk factors, such as insulin resistance, dyslipidemia, and obesity. These risk factors can further exacerbate systemic hypertension and may be a cause as well as a consequence of the deterioration of renal function. Systemic hypertension increases intraglomerular pressure, inducing hyperfiltration and proteinuria5). Furthermore, the overproduction of vasoactive factors disrupting renal vascular autoregulation worsens glomerular hyperfiltration, in which RAAS activation plays a crucial role10, 11). The local production of angiotensin II induces intraglomerular hypertension, proteinuria, and inflammatory pathways, which contribute to the development of the pathognomonic features of DKD, such as glomerular hypertrophy and sclerosis, tubulointerstitial inflammation, and fibrosis12).

An increase in urinary albumin excretion (UAE) is an early renal manifestation of generalized vascular dysfunction and may serve as an indicator of renal and CV risks13). The UAE rate, even within the normal range, is associated with changes in BP levels, development of hypertension, and renal vascular resistance, which are particularly evident in the presence of renal dysfunction14, 15). An increase in albuminuria is a predictor of ESKD progression15). Recent studies have confirmed that an increase in the UAE rate leads to poor CV and renal outcomes, while reduction of albuminuria attained through optimal BP control is associated with a favorable prognosis16, 17). Therefore, the reduction of albuminuria may serve as a treatment target because alterations in urinary albumin levels may reflect parallel changes in both CV and renal risks18).

Patients with diabetes and normoalbuminuria may present with different clinical manifestations. CKD may present heterogeneously in patients with type 1 and type 2 diabetes, with differing histological patterns and variations in the extent of fibrosis and ischemia involving the tubules, glomeruli, and interstitium19). Albuminuria may be absent or minimal in a significant proportion of patients with diabetes with declining renal function20). Higher BP levels deteriorate renal function regardless of the albuminuria status21). For instance, the United Kingdom Prospective Diabetes Study (UKPDS) showed that each 10 mmHg increase in mean BP caused a 15% increase in the hazard ratios for CKD development or a twofold increase in the serum creatinine levels in patients with normoalbuminuria22). Although the renal outcomes in patients with diabetes with traditional “albuminuric” phenotype have improved by maximal RAAS inhibition and BP lowering, the utility of corresponding treatments when albuminuria is not present remains unclear. For patients with non-classic phenotypes of DKD presenting with minimal albuminuria, the appropriate antihypertensive treatment and antihypertensive drug combinations should be determined based on the target BP values18).

Albuminuria is generally recognized as an indicator of and a treatment target for slowing the progression of kidney disease, and renoprotection is achieved by the reduction of albuminuria23). The United States Food and Drug Administration recently accredited the changes in albuminuria as the target marker for kidney disease progression in various clinical trials24). In addition, persistent hypertension may cause albuminuria, which may precede and even predict the development of hypertension, particularly in patients with low eGFR14). Considering the bidirectional relationship between hypertension and albuminuria, the optimal BP may require the reduction of albuminuria and tolerable decline in eGFR in patients with DKD.

Adequate measurement of blood pressure: Standardized versus routine office blood pressure measurement

In the 2021 KDIGO BP guidelines, the standardized office BP measurement was recommended over routine office measurement8). This recommendation was also previously incorporated in the 2017 American College of Cardiology/American Heart Association guidelines25). As BP levels based on office measurements tend to be higher than standardized BP measurements26), overtreatment and hypotensive events may be possible.

Several considerations should be taken into account when performing the standardized office BP measurements, that is, from the preparation to the actual measurement8). For instance, the patients are required to empty their bladders and refrain from consuming caffeine, exercising, and smoking for at least 30 min before the BP measurement. Additionally, patients should attempt to relax for more than 5 minutes while seated in a chair with back support and place both feet on the ground. Neither the patient nor the observer may talk during the 5-minute rest period and throughout the procedure. The cuff is placed directly on the skin, away from the patient’s clothing. During the measurement, adequate arm support is provided, and an adequately sized cuff, with a bladder length that covered 80% of the patient’s arm circumference should be placed on the upper arm of the patient. BP measurements are taken once in both arms, and subsequent measurements are taken in the arm with a higher BP level. A minimum of two BP measurements at 1- to 2-minute intervals were required, and the average of the measurements are recorded. Both systolic BP (SBP) and diastolic BP levels are recorded along with the antihypertension medication that the patient recently took before the BP measurement.

Notably, the American Diabetes Association (ADA) consensus panel has indicated that a 24-h ambulatory BP measurement is relevant in identifying the at-risk subgroups of patients with diabetes27). This may be due to the fact that masked hypertension and nocturnal non-dipping BP status are possible confounding variables in the relationship between office BP measurement and DKD progression28). Furthermore, standing or seated BP should be used as a target for treatment as some patients with diabetes and hypertension may develop autonomic neuropathy manifested as orthostatic hypotension and impaired systemic hemodynamics29).

Target blood pressure for patients with diabetic kidney disease

For the past two decades, the target BP levels in patients with and without albuminuria have been <130/80 mmHg and <140/90 mmHg, respectively30). Recent results from the Systolic Blood Pressure Intervention Trial (SPRINT) challenge this traditional school of thought7). This study demonstrated favorable results of intensive BP control, with a target SBP of <120 mmHg (mean BP, 121.4 mmHg), compared with that of the conventional target of <140 mmHg (mean BP, 136.2 mmHg). Adopting the results from the SPRINT, the KDIGO 2021 guidelines updated their recommendation to a target SBP of <120 mmHg in patients with CKD8). However, given the negative results of intensive BP control from several key studies6, 31, 32), other guidelines do not subscribe to the use of strict target BP levels and still recommend different targets for patients with CKD (Table 1)6, 31, 32).

Table 1
Guideline comparisons of goal BP for diabetic patients with hypertension

Nevertheless, mounting evidence appears to support the efficacy of intensive BP control in reducing adverse CV events and all-cause mortality rates, in patients with CKD with and without diabetes. In previous studies, a BP target of >140 mmHg in patients with CKD with diabetes was regarded as suboptimal33, 34, 35, 36). The UKPDS also noted favorable outcomes with tighter BP control (mean BP, 144/82 mmHg vs. mean BP, 154/87 mmHg) in terms of diabetes-related outcomes, mortality, stroke, and microvascular complications33). Additionally, two studies that conducted posthoc analyses, the Reduction of Endpoints in Non-Insulin-Dependent Diabetes Mellitus with the Angiotensin II Antagonist Losartan (RENAAL) study and the Irbesartan Diabetic Nephropathy Trial (IDNT), showed an association between lower BP levels and improved renal outcomes37, 38). In a meta-analysis of 123 trials that included 613,815 participants, the risks of major CV events and all-cause mortality were notably lower with every 10 mmHg reduction in SBP39). A pooled analysis of four multicenter RCTs including 4,983 patients with CKD also reported that an SBP target of <130 mmHg was associated with decreased all-cause mortality and CV outcomes compared with the standard BP target of <140 mmHg, excluding patients with an eGFR of ≥60 mL/min/1.73 m2 and who had undergone intensive glycemic control40).

Several studies have demonstrated the efficacy of intensive BP control. The Action in Diabetes and Vascular Disease: Preterax and Diamicron Modified Release Controlled Evaluation (ADVANCE) trial involving 11,140 patients with type 2 diabetes confirmed that the risk of significant renal events was reduced with a lower BP level (mean SBP, 134.7 vs. 140.3 mmHg), which was driven by the reduced risks of developing both micro- and macroalbuminuria41). In this study, progressively lower numbers of renal events were observed in patients with increasingly lower SBP levels (<110 mmHg). In the SPRINT, which is the most important clinical study investigating the effectiveness of intensive BP control, intensive BP control had beneficial effects on patients with a risk of experiencing serious adverse effects, such as hypotension, syncope, electrolyte abnormalities, and kidney injury.

However, results on the evaluation of the efficacy and safety of intensive SBP reduction to <120 mmHg were contradicting. In the Action to Control Cardiovascular Risk in Diabetes (ACCORD) trials involving patients with type 2 diabetes, strict SBP control to <120 mmHg (mean BP, 119.3 mmHg) had no association with CV and renal protection compared with the standard SBP target of <140 mmHg (mean BP, 133.5 mmHg), except for the risk of stroke6). Furthermore, interventions that substantially lowered the SBP level to <120 mmHg were associated with an increased risk of adverse kidney events, such as higher serum creatinine levels or an eGFR of <30 mL/min/1.73 m2 6). The corresponding SBP intervention in the SPRINT also demonstrated a higher incidence of adverse renal outcomes in those without diabetes7). Beddhu et al. compared the intensive and standard SBP groups in both SPRINT and ACCORD trial42); the intensive intervention group showed an increased risk of incident CKD regardless of the status of type 2 diabetes. Interestingly, the absolute risk was higher in patients with diabetes than in those without diabetes. The Appropriate Blood Control in Diabetes study also examined the effect of maintaining a lower BP target of <130/80 mmHg (mean, 128 mmHg) for 5 years on the preservation of renal function compared with that of achieving the standard target of <140/90 mmHg (mean: 137 mmHg) in patients who are normotensive with type 2 diabetes; however, no evidence was found to support the efficacy of intensive BP control43).

In addition to safety issues, some studies have shown a U-shaped association between SBP and the risk of mortality. An observational subgroup analysis in the International Verapamil SR-Trandolapril study showed that an SBP level of <110 mmHg showed a significantly increased risk of all-cause mortality compared with SBP levels of 125-130 mmHg44). Several cohort studies have also suggested the risks associated with a considerable BP reduction45, 46, 47). In a previous meta-analysis, BP-lowering treatments reduced the incidence of CV events, particularly in high-risk patients with various comorbidities, but they had no proportional effects in patients with a lower baseline SBP of <130 mmHg39). According to these results, the international guidelines that defined the optimal BP values have recently been revised, with recommendations including an SBP target of <130 mmHg for high-risk patients, such as those with diabetes or CKD25, 48, 49, 50).

A few limitations exist in the current RCTs. These involve diverse groups of patients but largely exclude patients with CKD or those with diabetes and advanced CKD. Notably, the SPRINT reported a mean eGFR of 48 mL/min/1.73 m2, mostly involving patients with CKD stage G3a; however, the study only included those in the prediabetes stage, which was present in 36.5% of the patients7). Another important study, the ACCORD trial, excluded patients with a serum creatinine level of >1.5 mg/dL. Meanwhile, the BP measurements in the SPRINT were performed under ideal conditions; that is, the patients were resting in a quiet room for 5 minutes without an observer, which may have induced a reduction in BP levels compared with that performed in other clinical trials. However, these conditions may not be applicable in a real clinical setting. The KDIGO guidelines suggest that an SBP target of <120 mmHg obtained using non-standardized BP measurement methods may be potentially harmful. Therefore, caution should be taken when applying the updated guidelines of intensive BP control in patients with DKD.

Blood pressure lowering on albuminuria

The optimal goal for BP control in patients with CKD includes the prevention of CV events and all-cause death as well as the prevention of incident CKD and attenuation of CKD progression. Although intensive BP control fails to preserve the GFR, it has been shown to reduce the risk of albuminuria. The ACCORD trial found that intensive BP control considerably reduced the risk of macroalbuminuria, even though a higher risk of CKD was noted. In the ADVANCE trial, a reduction in the risk of microalbuminuria and macroalbuminuria was achieved through active BP control41, 51). Furthermore, when albuminuria was reduced, independent of the BP status or antihypertensive regimen used, the increased risks of CV and renal outcomes were attenuated in patients with CKD who did not have diabetes16, 52).

However, the results of these studies, particularly the short-term data, should be interpreted with caution because albuminuria reduction may also result from GFR reduction53). These studies showed that lowering the BP levels may result in the simultaneous reduction of albuminuria and eGFR. Nevertheless, considering that various markers of kidney injury did not significantly increase despite the increase in serum creatinine levels, decreased eGFR may only reflect alterations in hemodynamics achieved by intensive BP control54, 55). Therefore, a reasonable decrease in the eGFR followed by a gradual recovery may be acceptable, as their effects on the CV outcomes of albuminuria reduction related to BP lowering are favorable.

Strategies to control blood pressure in patients with diabetic kidney disease

Results from patients with and without diabetes have shown that RAAS blockade provides a renoprotective effect, along with a reduction in proteinuria and attenuation of eGFR decline. In the African American Study of Kidney Disease and Hypertension trial, the angiotensin-converting enzyme inhibitor (ACEi)-treated group showed a slower eGFR decline than those who received other treatments56). In the RENAAL and IDNT studies involving patients with type 2 diabetes and CKD, the use of angiotensin II receptor blockers (ARBs) attenuated the progression of CKD57, 58). However, combination therapy with ACEis and ARBs is not recommended as it causes a greater decline in renal function and hyperkalemia59, 60, 61). Hyperkalemia related to RAAS blockade frequently occurs in patients with diabetes and a novel potassium binder patiromer can be used in those with CKD stages 3 to 4, and on RAAS blockade (ACEi/ARB, spironolactone)62). For patients with albuminuria or CV diseases, ACEis and ARBs are the first-line antihypertensive medications.

However, the efficacy of ACEi and ARB as first-line therapy in the absence of albuminuria remains unclear. In the Bergamo Nephrologic Diabetes Complications Trial, ACEi prevented the onset of microalbuminuria in patients with type 2 diabetes but with normal urinary albumin excretion, representing those with early-stage CKD without microalbuminuria63). The Randomized Olmesartan and Diabetes Microalbuminuria Prevention study also showed delayed onset of microalbuminuria in similar conditions64); however, significant fatal CV events occurred in the treatment group. Bangalo et al. also reported that no evidence was found regarding the superiority of RAAS blockade over other BP medications65). In the absence of albuminuria, maintaining a BP level of <130 mmHg but above 120 mmHg may be prudent66). Lower BP targets may be appropriate when the potential for kidney injury with intensive BP control is minimal and reversible, and the benefits against adverse CV events outweigh those of renal events.

With accumulating evidence of various pleiotropic effects conferred with respect to cardio and renoprotection, SGLT2i and GLP1RA have emerged as a game changer in the management strategy for DKD. Study results of various clinical trials were accepted by renowned societies including the ADA, the European Association for the Study of Diabetes and the KDIGO, recommending SGLT2i and GLP1RA as first-line therapies for patients with DKD67, 68). The Dapagliflozin and Prevention of Adverse Outcomes in Chronic Kidney Disease (DAPA-CKD) trial demonstrated that risk of kidney failure or death from CV or renal causes was reduced with dapagliflozin and was even effective in patients with CKD without diabetes69). Apart from their significant impact on lowering adverse CV events and death70, 71, 72), BP can be reduced by 5 mmHg by these drugs73, 74).

A highly selective endothelin A receptor (ETAR) antagonist, atrasentan can reduce albuminuria even with its short-term use in patients with DKD75). The long-term use of atrasentan was evaluated in 2,648 patients with type 2 diabetes and overt albuminuria in the Study of Diabetic Nephropathy with Atrasentan76). Composite adverse kidney outcome of doubling serum creatinine or kidney failure with replacement therapy was greatly reduced from atrasentan therapy. Its potential in proteinuria reduction should also be acknowledged with evidence from outcome of those with primary focal segmental glomerulosclerosis when used in combination with a dual ETAR antagonist (sparsentan) and ARB in a recent Phase 2 study77). It is also reported that BP is significantly lowered by ETAR antagonists77, 78).

Lastly, mineralocorticoid receptor antagonists (MRAs) provide cardioprotection and renoprotection79, 80). A first-generation nonselective MRA, spironolactone, was first known to provide renoprotection in non-diabetic patients with CKD by proteinuria reduction and eGFR preservation81). Then, finerenone, a next-generation selective MRA, proved to be a potential measure for DKD management showing its effective risk reduction in terms of CKD progression and CV events development in patients with CKD and diabetes in the Finerenone in Reducing Kidney Failure and Disease Progression in Diabetic Kidney Disease study82). People with resistant hypertension can even lower their BP with MRAs as well83, 84).

A recent retrospective study acknowledged the importance of consistent BP control53). Only 28% of patients achieved the target BP, with those remaining showing an increased risk of DKD (odds ratio 1.38) and albuminuria (odds ratio 1.47) within a period of 4 years. Therefore, adherence to the medication regimen is essential. Currently, several guidelines emphasize the non- or suboptimal adherence to antihypertensive medications as a hindrance to achieving consistent BP target.

CONCLUSIONS

The optimal BP control goal in patients with DKD should include the attenuation of renal functional deterioration and improvement of CV outcomes. Despite substantial attempts to address these factors, the optimal BP level in patients with DKD remains unknown. The recently updated KDIGO 2021 guidelines recommend the implementation of intensive BP control, with a target SBP level of <120 mmHg, based on the evidence that the CV benefits obtained is outweighed by the kidney injury risk associated with a lower BP target. However, different BP targets may be necessary, based on age, type of diabetes, and CKD stages. Lesser aggressive treatment strategies may be used in older and frail patients with non-albuminuric renal impairment, based on the paradoxical J-curve relationship between BP reduction and renal and CV morbidity85). However, future trials are needed to clarify other uncertainties. Well-designed RCTs may be able to evaluate the effects of intensive BP control through the use of various interventions in diverse patient populations, including patients with CKD with and without diabetes, those with a high CV risk or proteinuria, and those with early- and late-stage CKD. These trials may ultimately better determine the cutoff BP target values in these patients.

Acknowledgement

The authors declare that they have no conflict of interest. This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (2022R1A2C1005430)

References

    1. Jha V, Garcia-Garcia G, Iseki K, et al. Chronic kidney disease: Global dimension and perspectives. Lancet 2013;382:260–272.
    1. Hong YA, Ban TH, Kang CY, et al. Trends in epidemiologic characteristics of end-stage renal disease from 2019 korean renal data system (kords). Kidney Res Clin Pract 2021;40:52–61.
    1. Reidy K, Kang HM, Hostetter T, Susztak K. Molecular mechanisms of diabetic kidney disease. J Clin Invest 2014;124:2333–2340.
    1. Molitch ME, DeFronzo RA, Franz MJ, et al. Nephropathy in diabetes. Diabetes Care 27 2004;Suppl 1:S79–S83.
    1. Van Buren PN, Toto R. Hypertension in diabetic nephropathy: Epidemiology, mechanisms, and management. Adv Chronic Kidney Dis 2011;18:28–41.
    1. Cushman WC, Evans GW, Byington RP, et al. Effects of intensive blood-pressure control in type 2 diabetes mellitus. N Engl J Med 2010;362:1575–1585.
    1. Wright JT Jr, Williamson JD, Whelton PK, et al. A randomized trial of intensive versus standard blood-pressure control. N Engl J Med 2015;373:2103–2116.
    1. Kdigo 2021 clinical practice guideline for the management of blood pressure in chronic kidney disease. Kidney Int 2021;99:S1–S87.
    1. Nørgaard K, Feldt-Rasmussen B, Borch-Johnsen K, Saelan H, Deckert T. Prevalence of hypertension in type 1 (insulin-dependent) diabetes mellitus. Diabetologia 1990;33:407–410.
    1. Koszegi S, Molnar A, Lenart L, et al. Raas inhibitors directly reduce diabetes-induced renal fibrosis via growth factor inhibition. J Physiol 2019;597:193–209.
    1. Viazzi F, Bonino B, Cappadona F, Pontremoli R. Renin-angiotensin-aldosterone system blockade in chronic kidney disease: Current strategies and a look ahead. Intern Emerg Med 2016;11:627–635.
    1. Fioretto P, Mauer M. Histopathology of diabetic nephropathy. Semin Nephrol 2007;27:195–207.
    1. Leoncini G, Sacchi G, Viazzi F, et al. Microalbuminuria identifies overall cardiovascular risk in essential hypertension: An artificial neural network-based approach. J Hypertens 2002;20:1315–1321.
    1. Xu H, Huang X, Risérus U, et al. Urinary albumin excretion, blood pressure changes and hypertension incidence in the community: Effect modification by kidney function. Nephrol Dial Transplant 2014;29:1538–1545.
    1. Viazzi F, Leoncini G, Conti N, et al. Microalbuminuria is a predictor of chronic renal insufficiency in patients without diabetes and with hypertension: The magic study. Clin J Am Soc Nephrol 2010;5:1099–1106.
    1. Viazzi F, Muiesan ML, Schillaci G, et al. Changes in albuminuria and cardiovascular risk under antihypertensive treatment: A systematic review and meta-regression analysis. J Hypertens 2016;34:1689–1697.
    1. Heerspink HJ, Kröpelin TF, Hoekman J, de Zeeuw D. Drug-induced reduction in albuminuria is associated with subsequent renoprotection: A meta-analysis. J Am Soc Nephrol 2015;26:2055–2064.
    1. Viazzi F, Leoncini G, Grassi G, Pontremoli R. Antihypertensive treatment and renal protection: Is there a j-curve relationship? J Clin Hypertens (Greenwich) 2018;20:1560–1574.
    1. Fioretto P, Mauer M. Diabetic nephropathy: Diabetic nephropathy-challenges in pathologic classification. Nat Rev Nephrol 2010;6:508–510.
    1. Viazzi F, Piscitelli P, Giorda C, et al. Association of kidney disease measures with risk of renal function worsening in patients with hypertension and type 2 diabetes. J Diabetes Complications 2017;31:419–426.
    1. Ekinci EI, Jerums G, Skene A, et al. Renal structure in normoalbuminuric and albuminuric patients with type 2 diabetes and impaired renal function. Diabetes Care 2013;36:3620–3626.
    1. Retnakaran R, Cull CA, Thorne KI, Adler AI, Holman RR. Risk factors for renal dysfunction in type 2 diabetes: U.K. Prospective diabetes study 74. Diabetes 2006;55:1832–1839.
    1. Heerspink HJL, Greene T, Tighiouart H, et al. Change in albuminuria as a surrogate endpoint for progression of kidney disease: A meta-analysis of treatment effects in randomised clinical trials. Lancet Diabetes Endocrinol 2019;7:128–139.
    1. Levey AS, Gansevoort RT, Coresh J, et al. Change in albuminuria and gfr as end points for clinical trials in early stages of ckd: A scientific workshop sponsored by the national kidney foundation in collaboration with the us food and drug administration and european medicines agency. Am J Kidney Dis 2020;75:84–104.
    1. 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: A report of the american college of cardiology/american heart association task force on clinical practice guidelines. Hypertension 2018;71:e13–e115.
    1. Ahmad FS, Chan C, Rosenman MB, et al. Validity of cardiovascular data from electronic sources: The multi-ethnic study of atherosclerosis and healthlnk. Circulation 2017;136:1207–1216.
    1. Tuttle KR, Bakris GL, Bilous RW, et al. Diabetic kidney disease: A report from an ada consensus conference. Am J Kidney Dis 2014;64:510–533.
    1. Pogue V, Rahman M, Lipkowitz M, et al. Disparate estimates of hypertension control from ambulatory and clinic blood pressure measurements in hypertensive kidney disease. Hypertension 2009;53:20–27.
    1. Bonino B, Leoncini G, De Cosmo S, et al. Antihypertensive treatment in diabetic kidney disease: The need for a patient-centered approach. Medicina (Kaunas) 2019;55
    1. Mancia G, Fagard R, Narkiewicz K, et al. 2013 esh/esc guidelines for the management of arterial hypertension: The task force for the management of arterial hypertension of the european society of hypertension (esh) and of the european society of cardiology (esc). Eur Heart J 2013;34:2159–2219.
    1. Lonn EM, Bosch J, López-Jaramillo P, et al. Blood-pressure lowering in intermediate-risk persons without cardiovascular disease. N Engl J Med 2016;374:2009–2020.
    1. Ruggenenti P, Perna A, Loriga G, et al. Blood-pressure control for renoprotection in patients with non-diabetic chronic renal disease (rein-2): Multicentre, randomised controlled trial. Lancet 2005;365:939–946.
    1. Uk prospective diabetes study group. Tight blood pressure control and risk of macrovascular and microvascular complications in type 2 diabetes: Ukpds 38. Bmj 1998;317:703–713.
    1. Hansson L, Zanchetti A, Carruthers SG, et al. Effects of intensive blood-pressure lowering and low-dose aspirin in patients with hypertension: Principal results of the hypertension optimal treatment (hot) randomised trial. Hot study group. Lancet 1998;351:1755–1762.
    1. Curb JD, Pressel SL, Cutler JA, et al. Effect of diuretic-based antihypertensive treatment on cardiovascular disease risk in older diabetic patients with isolated systolic hypertension. Systolic hypertension in the elderly program cooperative research group. Jama 1996;276:1886–1892.
    1. Tuomilehto J, Rastenyte D, Birkenhäger WH, et al. Effects of calcium-channel blockade in older patients with diabetes and systolic hypertension. Systolic hypertension in europe trial investigators. N Engl J Med 1999;340:677–684.
    1. Bakris GL, Weir MR, Shanifar S, et al. Effects of blood pressure level on progression of diabetic nephropathy: Results from the renaal study. Arch Intern Med 2003;163:1555–1565.
    1. Pohl MA, Blumenthal S, Cordonnier DJ, et al. Independent and additive impact of blood pressure control and angiotensin ii receptor blockade on renal outcomes in the irbesartan diabetic nephropathy trial: Clinical implications and limitations. J Am Soc Nephrol 2005;16:3027–3037.
    1. Ettehad D, Emdin CA, Kiran A, et al. Blood pressure lowering for prevention of cardiovascular disease and death: A systematic review and meta-analysis. Lancet 2016;387:957–967.
    1. Malhotra R, Nguyen HA, Benavente O, et al. Association between more intensive vs less intensive blood pressure lowering and risk of mortality in chronic kidney disease stages 3 to 5: A systematic review and meta-analysis. JAMA Intern Med 2017;177:1498–1505.
    1. de Galan BE, Perkovic V, Ninomiya T, et al. Lowering blood pressure reduces renal events in type 2 diabetes. J Am Soc Nephrol 2009;20:883–892.
    1. Beddhu S, Greene T, Boucher R, et al. Intensive systolic blood pressure control and incident chronic kidney disease in people with and without diabetes mellitus: Secondary analyses of two randomised controlled trials. Lancet Diabetes Endocrinol 2018;6:555–563.
    1. Schrier RW, Estacio RO, Esler A, Mehler P. Effects of aggressive blood pressure control in normotensive type 2 diabetic patients on albuminuria, retinopathy and strokes. Kidney Int 2002;61:1086–1097.
    1. Cooper-DeHoff RM, Gong Y, Handberg EM, et al. Tight blood pressure control and cardiovascular outcomes among hypertensive patients with diabetes and coronary artery disease. Jama 2010;304:61–68.
    1. Kovesdy CP, Lu JL, Molnar MZ, et al. Observational modeling of strict vs conventional blood pressure control in patients with chronic kidney disease. JAMA Intern Med 2014;174:1442–1449.
    1. Sim JJ, Shi J, Kovesdy CP, Kalantar-Zadeh K, Jacobsen SJ. 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.
    1. Navaneethan SD, Schold JD, Jolly SE, et al. Blood pressure parameters are associated with all-cause and cause-specific mortality in chronic kidney disease. Kidney Int 2017;92:1272–1281.
    1. Lee HY, Shin J, Kim GH, et al. 2018 korean society of hypertension guidelines for the management of hypertension: Part ii-diagnosis and treatment of hypertension. Clin Hypertens 2019;25:20
    1. Hur KY, Moon MK, Park JS, et al. 2021 clinical practice guidelines for diabetes mellitus of the korean diabetes association. Diabetes Metab J 2021;45:461–481.
    1. Erratum 10. Cardiovascular disease and risk management: Standards of medical care in diabetes-2022. Diabetes care 2022;45(suppl. 1):S144-s174. Diabetes Care 2022;45:1296
    1. Patel A, MacMahon S, Chalmers J, et al. Effects of a fixed combination of perindopril and indapamide on macrovascular and microvascular outcomes in patients with type 2 diabetes mellitus (the advance trial): A randomised controlled trial. Lancet 2007;370:829–840.
    1. Savarese G, Dei Cas A, Rosano G, et al. Reduction of albumin urinary excretion is associated with reduced cardiovascular events in hypertensive and/or diabetic patients. A meta-regression analysis of 32 randomized trials. Int J Cardiol 2014;172:403–410.
    1. De Cosmo S, Viazzi F, Piscitelli P, et al. Blood pressure status and the incidence of diabetic kidney disease in patients with hypertension and type 2 diabetes. J Hypertens 2016;34:2090–2098.
    1. Nadkarni GN, Chauhan K, Rao V, et al. Effect of intensive blood pressure lowering on kidney tubule injury: Findings from the accord trial study participants. Am J Kidney Dis 2019;73:31–38.
    1. Rocco MV, Sink KM, Lovato LC, et al. Effects of intensive blood pressure treatment on acute kidney injury events in the systolic blood pressure intervention trial (sprint). Am J Kidney Dis 2018;71:352–361.
    1. Agodoa LY, Appel L, Bakris GL, et al. Effect of ramipril vs amlodipine on renal outcomes in hypertensive nephrosclerosis: A randomized controlled trial. Jama 2001;285:2719–2728.
    1. 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.
    1. Parving HH, Lehnert H, Bröchner-Mortensen J, Gomis R, Andersen S, Arner P. The effect of irbesartan on the development of diabetic nephropathy in patients with type 2 diabetes. N Engl J Med 2001;345:870–878.
    1. Fried LF, Emanuele N, Zhang JH, et al. Combined angiotensin inhibition for the treatment of diabetic nephropathy. N Engl J Med 2013;369:1892–1903.
    1. Mann JF, Schmieder RE, McQueen M, et al. Renal outcomes with telmisartan, ramipril, or both, in people at high vascular risk (the ontarget study): A multicentre, randomised, double-blind, controlled trial. Lancet 2008;372:547–553.
    1. Parving HH, Brenner BM, McMurray JJ, et al. Cardiorenal end points in a trial of aliskiren for type 2 diabetes. N Engl J Med 2012;367:2204–2213.
    1. Bakris GL, Pitt B, Weir MR, et al. Effect of patiromer on serum potassium level in patients with hyperkalemia and diabetic kidney disease: The amethyst-dn randomized clinical trial. Jama 2015;314:151–161.
    1. Ruggenenti P, Fassi A, Ilieva AP, et al. Preventing microalbuminuria in type 2 diabetes. N Engl J Med 2004;351:1941–1951.
    1. Haller H, Ito S, Izzo JL Jr, et al. Olmesartan for the delay or prevention of microalbuminuria in type 2 diabetes. N Engl J Med 2011;364:907–917.
    1. Bangalore S, Fakheri R, Toklu B, Messerli FH. Diabetes mellitus as a compelling indication for use of renin angiotensin system blockers: Systematic review and meta-analysis of randomized trials. Bmj 2016;352:i438
    1. Hildebrand AM, Garg AX. Blood pressure targets in chronic kidney disease: Does proteinuria dictate how low we go? Cmaj 2013;185:941–942.
    1. Davies MJ, D'Alessio DA, Fradkin J, et al. Management of hyperglycemia in type 2 diabetes, 2018. A consensus report by the american diabetes association (ada) and the european association for the study of diabetes (easd). Diabetes Care 2018;41:2669–2701.
    1. Kdigo 2020 clinical practice guideline for diabetes management in chronic kidney disease. Kidney Int 2020;98:S1–S115.
    1. Heerspink HJL, Stefánsson BV, Correa-Rotter R, et al. Dapagliflozin in patients with chronic kidney disease. N Engl J Med 2020;383:1436–1446.
    1. Neal B, Perkovic V, Mahaffey KW, et al. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med 2017;377:644–657.
    1. Wanner C, Inzucchi SE, Lachin JM, et al. Empagliflozin and progression of kidney disease in type 2 diabetes. N Engl J Med 2016;375:323–334.
    1. Wiviott SD, Raz I, Bonaca MP, et al. Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2019;380:347–357.
    1. Wang B, Zhong J, Lin H, et al. Blood pressure-lowering effects of glp-1 receptor agonists exenatide and liraglutide: A meta-analysis of clinical trials. Diabetes Obes Metab 2013;15:737–749.
    1. Majewski C, Bakris GL. Blood pressure reduction: An added benefit of sodium-glucose cotransporter 2 inhibitors in patients with type 2 diabetes. Diabetes Care 2015;38:429–430.
    1. Kohan DE, Pritchett Y, Molitch M, et al. Addition of atrasentan to renin-angiotensin system blockade reduces albuminuria in diabetic nephropathy. J Am Soc Nephrol 2011;22:763–772.
    1. Heerspink HJL, Parving HH, Andress DL, et al. Atrasentan and renal events in patients with type 2 diabetes and chronic kidney disease (sonar): A double-blind, randomised, placebo-controlled trial. Lancet 2019;393:1937–1947.
    1. Trachtman H, Nelson P, Adler S, et al. Duet: A phase 2 study evaluating the efficacy and safety of sparsentan in patients with fsgs. J Am Soc Nephrol 2018;29:2745–2754.
    1. Raichlin E, Prasad A, Mathew V, et al. Efficacy and safety of atrasentan in patients with cardiovascular risk and early atherosclerosis. Hypertension 2008;52:522–528.
    1. Pitt B, Zannad F, Remme WJ, et al. The effect of spironolactone on morbidity and mortality in patients with severe heart failure. Randomized aldactone evaluation study investigators. N Engl J Med 1999;341:709–717.
    1. Pitt B, Remme W, Zannad F, et al. Eplerenone, a selective aldosterone blocker, in patients with left ventricular dysfunction after myocardial infarction. N Engl J Med 2003;348:1309–1321.
    1. Bianchi S, Bigazzi R, Campese VM. Long-term effects of spironolactone on proteinuria and kidney function in patients with chronic kidney disease. Kidney Int 2006;70:2116–2123.
    1. Bakris GL, Agarwal R, Anker SD, et al. Effect of finerenone on chronic kidney disease outcomes in type 2 diabetes. N Engl J Med 2020;383:2219–2229.
    1. Williams B, MacDonald TM, Morant S, et al. Spironolactone versus placebo, bisoprolol, and doxazosin to determine the optimal treatment for drug-resistant hypertension (pathway-2): A randomised, double-blind, crossover trial. Lancet 2015;386:2059–2068.
    1. Krieger EM, Drager LF, Giorgi DMA, et al. Spironolactone versus clonidine as a fourth-drug therapy for resistant hypertension: The rehot randomized study (resistant hypertension optimal treatment). Hypertension 2018;71:681–690.
    1. Jung HH. Hypertension management in patients with chronic kidney disease in the post-sprint era. Electrolyte Blood Press 2021;19:19–28.

Metrics
Share
Tables

1 / 1

PERMALINK