Treatment of diabetic kidney disease: current and future targets

Mi-Kyung Kim, Mi-Kyung Kim

Abstract

Diabetic kidney disease (DKD) is a leading cause of end-stage renal disease in Korea and worldwide, and is a risk factor for the development of cardiovascular complications. The conventional treatments for DKD are control of blood glucose and blood pressure levels by inhibiting the renin-angiotensin system. However, the prevalence of DKD continues to increase and additional therapies are required to prevent or ameliorate the condition. Many drugs have been, or are being, developed to target the molecular mechanisms in play in DKD. This review focuses on DVD treatment, considering current and emerging therapeutic targets and the clinical trial-based evidence.

Keywords: Diabetes; Kidney; Therapeutics.

Conflict of interest statement

No potential conflict of interest relevant to this article was reported.

Figures

Figure 1.
Figure 1.
Current and emerging treatments for diabetic kidney disease. DPP-4, dipeptidyl peptidase-4; SGLT2, sodium-glucose cotransporter 2; NOX, NADPH oxidase; PKC, protein kinase C; AGE, advanced glycation endproduct; RAGE, receptor for advanced glycation endproduct; ACE-I, angiotensin converting enzyme-inhibitor; ARB, angiotensin II receptor blocker; Nrf2, nuclear factor-like 2; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; JAK/STAT, Janus kinase-signal transducer and activator transcription factor; CCR, C-C chemokine receptor; TGF-β, transforming growth factor β; VEGF, vascular endothelial growth factor; MCP-1, monocyte chemotactic protein 1; ECM, extracellular matrix.

References

    1. Park CW. Diabetic kidney disease: from epidemiology to clinical perspectives. Diabetes Metab J. 2014;38:252–260.
    1. Ahn JH, Yu JH, Ko SH, et al. Prevalence and determinants of diabetic nephropathy in Korea: Korea National Health and Nutrition Examination Survey. Diabetes Metab J. 2014;38:109–119.
    1. Gregg EW, Li Y, Wang J, et al. Changes in diabetes-related complications in the United States, 1990-2010. N Engl J Med. 2014;370:1514–1523.
    1. Rhee EJ. Diabetes in Asians. Endocrinol Metab (Seoul) 2015;30:263–269.
    1. Parving HH, Lewis JB, Ravid M, Remuzzi G, Hunsicker LG, DEMAND investigators Prevalence and risk factors for microalbuminuria in a referred cohort of type II diabetic patients: a global perspective. Kidney Int. 2006;69:2057–2063.
    1. Macisaac RJ, Ekinci EI, Jerums G. Markers of and risk factors for the development and progression of diabetic kidney disease. Am J Kidney Dis. 2014;63(2 Suppl 2):S39–S62.
    1. Kim Y, Park CW. New therapeutic agents in diabetic nephropathy. Korean J Intern Med. 2017;32:11–25.
    1. Muskiet MH, Smits MM, Morsink LM, Diamant M. The gut-renal axis: do incretin-based agents confer renoprotection in diabetes? Nat Rev Nephrol. 2014;10:88–103.
    1. Diabetes Control and Complications Trial Research Group. Nathan DM, Genuth S, et al. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. N Engl J Med. 1993;329:977–986.
    1. UK Prospective Diabetes Study (UKPDS) Group Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33) Lancet. 1998;352:837–853.
    1. ADVANCE Collaborative Group. Patel A, MacMahon S, et al. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med. 2008;358:2560–2572.
    1. Stratton IM, Adler AI, Neil HA, et al. Association of glycaemia with macrovascular and microvascular complications of type 2 diabetes (UKPDS 35): prospective observational study. BMJ. 2000;321:405–412.
    1. Zoungas S, Chalmers J, Ninomiya T, et al. Association of HbA1c levels with vascular complications and death in patients with type 2 diabetes: evidence of glycaemic thresholds. Diabetologia. 2012;55:636–643.
    1. Korean Diabetes Association . Seoul: Korean Diabetes Association; c2011. Treatment guideline for diabetes: 2015 [Internet] [cited 2017 Jun 14]. Available from: .
    1. American Diabetes Association 5: Glycemic targets. Diabetes Care. 2016;39 Suppl 1:S39–S46.
    1. National Kidney Foundation KDOQI clinical practice guideline for diabetes and CKD: 2012 update. Am J Kidney Dis. 2012;60:850–886.
    1. Penno G, Garofolo M, Del Prato S. Dipeptidyl peptidase-4 inhibition in chronic kidney disease and potential for protection against diabetes-related renal injury. Nutr Metab Cardiovasc Dis. 2016;26:361–373.
    1. Panchapakesan U, Pollock C. The role of dipeptidyl peptidase: 4 inhibitors in diabetic kidney disease. Front Immunol. 2015;6:443.
    1. Solini A. Role of SGLT2 inhibitors in the treatment of type 2 diabetes mellitus. Acta Diabetol. 2016;53:863–870.
    1. Gilbert RE. Sodium-glucose linked transporter-2 inhibitors: potential for renoprotection beyond blood glucose lowering? Kidney Int. 2014;86:693–700.
    1. Zhong J, Rao X, Rajagopalan S. An emerging role of dipeptidyl peptidase 4 (DPP4) beyond glucose control: potential implications in cardiovascular disease. Atherosclerosis. 2013;226:305–314.
    1. Tiruppathi C, Miyamoto Y, Ganapathy V, Roesel RA, Whitford GM, Leibach FH. Hydrolysis and transport of proline-containing peptides in renal brush-border membrane vesicles from dipeptidyl peptidase IV-positive and dipeptidyl peptidase IV-negative rat strains. J Biol Chem. 1990;265:1476–1483.
    1. Mentlein R. Dipeptidyl-peptidase IV (CD26): role in the inactivation of regulatory peptides. Regul Pept. 1999;85:9–24.
    1. Sharkovska Y, Reichetzeder C, Alter M, et al. Blood pressure and glucose independent renoprotective effects of dipeptidyl peptidase-4 inhibition in a mouse model of type-2 diabetic nephropathy. J Hypertens. 2014;32:2211–2223.
    1. Yang J, Campitelli J, Hu G, Lin Y, Luo J, Xue C. Increase in DPP-IV in the intestine, liver and kidney of the rat treated with high fat diet and streptozotocin. Life Sci. 2007;81:272–279.
    1. Pala L, Mannucci E, Pezzatini A, et al. Dipeptidyl peptidase-IV expression and activity in human glomerular endothelial cells. Biochem Biophys Res Commun. 2003;310:28–31.
    1. Mega C, de Lemos ET, Vala H, et al. Diabetic nephropathy amelioration by a low-dose sitagliptin in an animal model of type 2 diabetes (Zucker diabetic fatty rat) Exp Diabetes Res. 2011;2011:162092.
    1. Marques C, Mega C, Goncalves A, et al. Sitagliptin prevents inflammation and apoptotic cell death in the kidney of type 2 diabetic animals. Mediators Inflamm. 2014;2014:538737.
    1. Liu WJ, Xie SH, Liu YN, et al. Dipeptidyl peptidase IV inhibitor attenuates kidney injury in streptozotocin-induced diabetic rats. J Pharmacol Exp Ther. 2012;340:248–255.
    1. Jung GS, Jeon JH, Choe MS, et al. Renoprotective effect of gemigliptin, a dipeptidyl peptidase-4 inhibitor, in streptozotocin-induced type 1 diabetic mice. Diabetes Metab J. 2016;40:211–221.
    1. Kodera R, Shikata K, Takatsuka T, et al. Dipeptidyl peptidase-4 inhibitor ameliorates early renal injury through its anti-inflammatory action in a rat model of type 1 diabetes. Biochem Biophys Res Commun. 2014;443:828–833.
    1. Kanasaki K, Shi S, Kanasaki M, et al. Linagliptin-mediated DPP-4 inhibition ameliorates kidney fibrosis in streptozotocin-induced diabetic mice by inhibiting endothelial-to-mesenchymal transition in a therapeutic regimen. Diabetes. 2014;63:2120–2131.
    1. Gangadharan Komala M, Gross S, Zaky A, Pollock C, Panchapakesan U. Saxagliptin reduces renal tubulointerstitial inflammation, hypertrophy and fibrosis in diabetes. Nephrology (Carlton) 2016;21:423–431.
    1. Hattori S. Sitagliptin reduces albuminuria in patients with type 2 diabetes. Endocr J. 2011;58:69–73.
    1. Fujita H, Taniai H, Murayama H, et al. DPP-4 inhibition with alogliptin on top of angiotensin II type 1 receptor blockade ameliorates albuminuria via up-regulation of SDF-1alpha in type 2 diabetic patients with incipient nephropathy. Endocr J. 2014;61:159–166.
    1. White WB, Cannon CP, Heller SR, et al. Alogliptin after acute coronary syndrome in patients with type 2 diabetes. N Engl J Med. 2013;369:1327–1335.
    1. Scirica BM, Bhatt DL, Braunwald E, et al. Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N Engl J Med. 2013;369:1317–1326.
    1. Groop PH, Cooper ME, Perkovic V, Emser A, Woerle HJ, von Eynatten M. Linagliptin lowers albuminuria on top of recommended standard treatment in patients with type 2 diabetes and renal dysfunction. Diabetes Care. 2013;36:3460–3468.
    1. Katz PM, Leiter LA. The role of the kidney and SGLT2 inhibitors in type 2 diabetes. Can J Diabetes. 2015;39 Suppl 5:S167–S175.
    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. Vallon V, Gerasimova M, Rose MA, et al. SGLT2 inhibitor empagliflozin reduces renal growth and albuminuria in proportion to hyperglycemia and prevents glomerular hyperfiltration in diabetic Akita mice. Am J Physiol Renal Physiol. 2014;306:F194–F204.
    1. De Nicola L, Gabbai FB, Liberti ME, Sagliocca A, Conte G, Minutolo R. Sodium/glucose cotransporter 2 inhibitors and prevention of diabetic nephropathy: targeting the renal tubule in diabetes. Am J Kidney Dis. 2014;64:16–24.
    1. Chilton R, Tikkanen I, Cannon CP, et al. Effects of empagliflozin on blood pressure and markers of arterial stiffness and vascular resistance in patients with type 2 diabetes. Diabetes Obes Metab. 2015;17:1180–1193.
    1. Adler AI, Stratton IM, Neil HA, et al. Association of systolic blood pressure with macrovascular and microvascular complications of type 2 diabetes (UKPDS 36): prospective observational study. BMJ. 2000;321:412–419.
    1. Patel A, ADVANCE Collaborative Group. MacMahon S, 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. American Diabetes Association 9: Microvascular complications and foot care. Diabetes Care. 2016;39 Suppl 1:S72–S80.
    1. Taler SJ, Agarwal R, Bakris GL, et al. KDOQI US commentary on the 2012 KDIGO clinical practice guideline for management of blood pressure in CKD. Am J Kidney Dis. 2013;62:201–213.
    1. Kitada M, Kanasaki K, Koya D. Clinical therapeutic strategies for early stage of diabetic kidney disease. World J Diabetes. 2014;5:342–356.
    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. Brem AS, Morris DJ, Gong R. Aldosterone-induced fibrosis in the kidney: questions and controversies. Am J Kidney Dis. 2011;58:471–479.
    1. Hou J, Xiong W, Cao L, Wen X, Li A. Spironolactone addon for preventing or slowing the progression of diabetic nephropathy: a meta-analysis. Clin Ther. 2015;37:2086–2103.e10.
    1. Pitt B, Kober L, Ponikowski P, et al. Safety and tolerability of the novel non-steroidal mineralocorticoid receptor antagonist BAY 94-8862 in patients with chronic heart failure and mild or moderate chronic kidney disease: a randomized, double-blind trial. Eur Heart J. 2013;34:2453–2463.
    1. Bakris GL, Agarwal R, Chan JC, et al. Effect of finerenone on albuminuria in patients with diabetic nephropathy: a randomized clinical trial. JAMA. 2015;314:884–894.
    1. Gagliardini E, Zoja C, Benigni A. Et and diabetic nephropathy: preclinical and clinical studies. Semin Nephrol. 2015;35:188–196.
    1. Mann JF, Green D, Jamerson K, et al. Avosentan for overt diabetic nephropathy. J Am Soc Nephrol. 2010;21:527–535.
    1. Perez-Gomez MV, Sanchez-Nino MD, Sanz AB, et al. Horizon 2020 in diabetic kidney disease: the clinical trial pipeline for add-on therapies on top of renin angiotensin system blockade. J Clin Med. 2015;4:1325–1347.
    1. Shintani T, Klionsky DJ. Autophagy in health and disease: a double-edged sword. Science. 2004;306:990–995.
    1. White E. Deconvoluting the context-dependent role for autophagy in cancer. Nat Rev Cancer. 2012;12:401–410.
    1. Masini M, Bugliani M, Lupi R, et al. Autophagy in human type 2 diabetes pancreatic beta cells. Diabetologia. 2009;52:1083–1086.
    1. Kume S, Koya D. Autophagy: a novel therapeutic target for diabetic nephropathy. Diabetes Metab J. 2015;39:451–460.
    1. Kim SI, Na HJ, Ding Y, Wang Z, Lee SJ, Choi ME. Autophagy promotes intracellular degradation of type I collagen induced by transforming growth factor (TGF)-β1. J Biol Chem. 2012;287:11677–11688.
    1. Yamahara K, Kume S, Koya D, et al. Obesity-mediated autophagy insufficiency exacerbates proteinuria-induced tubulointerstitial lesions. J Am Soc Nephrol. 2013;24:1769–1781.
    1. Mizushima N, Yamamoto A, Matsui M, Yoshimori T, Ohsumi Y. In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. Mol Biol Cell. 2004;15:1101–1111.
    1. Bromann PA, Korkaya H, Courtneidge SA. The interplay between Src family kinases and receptor tyrosine kinases. Oncogene. 2004;23:7957–7968.
    1. Thomas SM, Brugge JS. Cellular functions regulated by Src family kinases. Annu Rev Cell Dev Biol. 1997;13:513–609.
    1. Kopetz S, Shah AN, Gallick GE. Src continues aging: current and future clinical directions. Clin Cancer Res. 2007;13:7232–7236.
    1. Mima A, Matsubara T, Arai H, et al. Angiotensin II-dependent Src and Smad1 signaling pathway is crucial for the development of diabetic nephropathy. Lab Invest. 2006;86:927–939.
    1. Taniguchi K, Xia L, Goldberg HJ, et al. Inhibition of Src kinase blocks high glucose-induced EGFR transactivation and collagen synthesis in mesangial cells and prevents diabetic nephropathy in mice. Diabetes. 2013;62:3874–3886.
    1. Yan Y, Ma L, Zhou X, et al. Src inhibition blocks renal interstitial fibroblast activation and ameliorates renal fibrosis. Kidney Int. 2016;89:68–81.
    1. Zhou D, Liu Y. Therapy for kidney fibrosis: is the Src kinase a potential target? Kidney Int. 2016;89:12–14.
    1. Seo HY, Jeon JH, Jung YA, et al. Fyn deficiency attenuates renal fibrosis by inhibition of phospho-STAT3. Kidney Int. 2016;90:1285–1297.

Source: PubMed

3
Tilaa