1,188
Views
20
CrossRef citations to date
0
Altmetric
Review

Recent early clinical drug development for acute kidney injury

ORCID Icon, ORCID Icon, , , &
Pages 141-154 | Received 02 Jun 2016, Accepted 16 Dec 2016, Published online: 27 Dec 2016

References

  • Bedford M, Stevens PE, Wheeler TW, et al. What is the real impact of acute kidney injury? BMC Nephrol. 2014;15:95.
  • Linder A, Fjell C, Levin A, et al. Small acute increases in serum creatinine are associated with decreased long-term survival in the critically ill. Am J Respir Crit Care Med. 2014;189(9):1075–1081.
  • Doyle JF, Forni LG. Acute kidney injury: short-term and long-term effects. Crit Care. 2016;20(1):188 .
  • Chawla LS, Kimmel PL. Acute kidney injury and chronic kidney disease: an integrated clinical syndrome. Kidney Int. 2012;82(5):516–524.
  • Meybohm P, Bein B, Brosteanu O, et al. A multicenter trial of remote ischemic preconditioning for heart surgery. N Engl J Med. 2015;373(15):1397–1407.
  • Hausenloy DJ, Candilio L, Evans R, et al. Remote ischemic preconditioning and outcomes of cardiac surgery. N Engl J Med. 2015;373(15):1408–1417.
  • Zarbock A, Schmidt C, Van Aken H, et al. Effect of remote ischemic preconditioning on kidney injury among high-risk patients undergoing cardiac surgery: a randomized clinical trial. JAMA. 2015;313(21):2133–2141.
  • Weisbord SD, Palevsky PM. Design of clinical trials in acute kidney injury: lessons from the past and future directions. Semin Nephrol. 2016;36(1):42–52.
  • Ramos AM, Gonzalez-Guerrero C, Sanz A, et al. Designing drugs that combat kidney damage. Expert Opin Drug Discov. 2015;10(5):541–556.
  • Sanz AB, Sanchez-Nino MD, Martin-Cleary C, et al. Progress in the development of animal models of acute kidney injury and its impact on drug discovery. Expert Opin Drug Discov. 2013;8(7):879–895.
  • Cruz DN, Mehta RL. Acute kidney injury in 2013: breaking barriers for biomarkers in AKI--progress at last. Nat Rev Nephrol. 2014;10(2):74–76.
  • Peddi V, Ratner L, Cooper M, et al. Abstract #2967 Treatment with QPI-1002, a Short Interfering (SI) RNA for the prophylaxis of delayed graft function. World transplant congress. Am J Transplant. 2014;14(S3):152–155.
  • Wire B. Thrasos announces promising results for phase 2 THR-184 dose ranging clinical study for the prevention of acute kidney injury (AKI) [webpage]. 2016. [cited 2016 May 07]. Available from: http://www.businesswire.com/news/home/20160229005651/en/Thrasos-Announces-Promising-Results-Phase-2-THR-184
  • Bromberg J, Weir M, Gaber A., et al. BB-3 a hepatocyte growth factor like molecule, improves outcome in kidney transplant recipients with delayed graft function. American society of nephrology kidney week 2015; San Diego. J Am Soc Nephrol. 2015;852A. abstr no TH-OR101.
  • Thomas RA, Czopek A, Bellamy CO, et al. Hemin preconditioning upregulates heme oxygenase-1 in deceased donor renal transplant recipients: a randomized, controlled, phase IIB trial. Transplantation. 2016;100(1):176–183 .
  • Madhav Swaminathan DM, Chertow GM, Warnock DG, et al. ACT-AKI: a phase 2 multicenter, randomized, double-blind, placebo-controlled trial of AC607 for the treatment of acute kidney injury in cardiac surgery subjects kidney week 2014; November 2014; Philadelphia. J Am Soc Nephrol. 2014. abstr no HI-OR09.
  • McCullough PA, Bennett-Guerrero E, Chawla LS, et al. ABT-719 for the prevention of acute kidney injury in patients undergoing high-risk cardiac surgery: a randomized phase 2b clinical trial. J Am Heart Assoc. 2016;5(8):e003549.
  • Pickkers P, Heemskerk S, Schouten J, et al. Alkaline phosphatase for treatment of sepsis-induced acute kidney injury: a prospective randomized double-blind placebo-controlled trial. Crit Care. 2012;16(1):R14.
  • Bulger EM, Maier RV, Sperry J, et al. A novel drug for treatment of necrotizing soft-tissue infections: a randomized clinical trial. JAMA Surgery. 2014;149(6):528–536.
  • AtoxBio. Clinical data: study ATB-201: recovery from AKI in subset of patients with AKI during first 7 days [webpage]. [cited 2016 Aug 12]. Available from: http://www.atoxbio.com/clinical-data-7/
  • Vaseva AV, Marchenko ND, Ji K, et al. p53 opens the mitochondrial permeability transition pore to trigger necrosis. Cell. 2012;149(7):1536–1548.
  • Thompson JD, Kornbrust DJ, Foy JW, et al. Toxicological and pharmacokinetic properties of chemically modified siRNAs targeting p53 RNA following intravenous administration. Nucleic Acid Ther. 2012;22(4):255–264.
  • Ying Y, Kim J, Westphal SN, et al. Targeted deletion of p53 in the proximal tubule prevents ischemic renal injury. J Am Soc Nephrol. 2014;25(12):2707–2716. .
  • Molitoris BA, Dagher PC, Sandoval RM, et al. siRNA targeted to p53 attenuates ischemic and cisplatin-induced acute kidney injury. J Am Soc Nephrol. 2009;20(8):1754–1764. .
  • Bramlage CP, Tampe B, Koziolek M, et al. Bone morphogenetic protein (BMP)-7 expression is decreased in human hypertensive nephrosclerosis. BMC Nephrol. 2010;11:31.
  • Hanai J. [BMP in the kidney: signaling, function and pathophysiological significance]. Clin Calcium. 2007;17(5):704–710.
  • Leeuwis JW, Nguyen TQ, Chuva de Sousa Lopes SM, et al. Direct visualization of Smad1/5/8-mediated transcriptional activity identifies podocytes and collecting ducts as major targets of BMP signalling in healthy and diseased kidneys. J Pathol. 2011;224(1):121–132.
  • Wang Z, Zhao J, Zhang J, et al. Protective effect of BMP-7 against aristolochic acid-induced renal tubular epithelial cell injury. Toxicol Lett. 2010;198(3):348–357.
  • Miyazono K, Maeda S, Imamura T. BMP receptor signaling: transcriptional targets, regulation of signals, and signaling cross-talk. Cytokine Growth Factor Rev. 2005;16(3):251–263.
  • Simic P, Vukicevic S. Bone morphogenetic proteins in development and homeostasis of kidney. Cytokine Growth Factor Rev. 2005;16(3):299–308.
  • Zeisberg M, Shah AA, Kalluri R. Bone morphogenic protein-7 induces mesenchymal to epithelial transition in adult renal fibroblasts and facilitates regeneration of injured kidney. J Biol Chem. 2005;280(9):8094–8100.
  • Zeisberg M, Kalluri R. Reversal of experimental renal fibrosis by BMP7 provides insights into novel therapeutic strategies for chronic kidney disease. Pediatr Nephrol. 2008;23(9):1395–1398.
  • Gould SE, Day M, Jones SS, et al. BMP-7 regulates chemokine, cytokine, and hemodynamic gene expression in proximal tubule cells. Kidney Int. 2002;61(1):51–60.
  • Sugimoto H, LeBleu VS, Bosukonda D, et al. Activin-like kinase 3 is important for kidney regeneration and reversal of fibrosis. Nat Med. 2012;18(3):396–404 .
  • Wetzel P, Haag J, Câmpean V, et al. Bone morphogenetic protein-7 expression and activity in the human adult normal kidney is predominantly localized to the distal nephron. Kidney Int. 2006;70(4):717–723.
  • Liu Y. Hepatocyte growth factor and the kidney. Curr Opin Nephrol Hypertens. 2002;11(1):23–30.
  • Liu Y, Tolbert EM, Lin L, et al. Up-regulation of hepatocyte growth factor receptor: an amplification and targeting mechanism for hepatocyte growth factor action in acute renal failure. Kidney Int. 1999;55(2):442–453.
  • Liu Y, Sun AM, Dworkin LD. Hepatocyte growth factor protects renal epithelial cells from apoptotic cell death. Biochem Biophys Res Commun. 1998;246(3):821–826.
  • Franquesa M, Riera M, Herrero-Fresneda I, et al. Tubular epithelial cells transfected with hHGF counteracts monocyte chemotactic protein-1 up-regulation after hypoxia/reoxygenation insult. Transplant Proc. 2009;41(6):2069–2072.
  • Adachi E, Hirose-Sugiura T, Kato Y, et al. Pharmacokinetics and pharmacodynamics following intravenous administration of recombinant human hepatocyte growth factor in rats with renal injury. Pharmacology. 2014;94(3–4):190–197.
  • Dai C, Yang J, Liu Y. Single injection of naked plasmid encoding hepatocyte growth factor prevents cell death and ameliorates acute renal failure in mice. J Am Soc Nephrol. 2002;13(2):411–422.
  • Homsi E, Janino P, Amano M, et al. Endogenous hepatocyte growth factor attenuates inflammatory response in glycerol-induced acute kidney injury. Am J Nephrol. 2009;29(4):283–291.
  • Yamasaki N, Nagano T, Mori-Kudo I, et al. Hepatocyte growth factor protects functional and histological disorders of HgCl(2)-induced acute renal failure mice. Nephron. 2002;90(2):195–205.
  • Zhou D, Tan RJ, Lin L, et al. Activation of hepatocyte growth factor receptor, c-met, in renal tubules is required for renoprotection after acute kidney injury. Kidney Int. 2013;84(3):509–520 .
  • Narayan P, Duan B, Jiang K, et al. Late intervention with the small molecule BB3 mitigates postischemic kidney injury. Am J Physiol Renal Physiol. 2016;311(2):F352–61. .
  • Tracz MJ, Alam J, Nath KA. Physiology and pathophysiology of heme: implications for kidney disease. J Am Soc Nephrol. 2007;18(2):414–420.
  • Shiraishi F, Curtis LM, Truong L, et al. Heme oxygenase-1 gene ablation or expression modulates cisplatin-induced renal tubular apoptosis. Am J Physiol Renal Physiol. 2000;278(5):F726–36.
  • Tracz MJ, Juncos JP, Croatt AJ, et al. Deficiency of heme oxygenase-1 impairs renal hemodynamics and exaggerates systemic inflammatory responses to renal ischemia. Kidney Int. 2007;72(9):1073–1080.
  • Tracz MJ, Juncos JP, Grande JP, et al. Renal hemodynamic, inflammatory, and apoptotic responses to lipopolysaccharide in HO-1-/- mice. Am J Pathol. 2007;170(6):1820–1830.
  • Blydt-Hansen TD, Katori M, Lassman C, et al. Gene transfer-induced local heme oxygenase-1 overexpression protects rat kidney transplants from ischemia/reperfusion injury. J Am Soc Nephrol. 2003;14(3):745–754.
  • Karch J, Kanisicak O, Brody MJ, et al. Necroptosis interfaces with MOMP and the MPTP in mediating cell death. PLoS One. 2015;10(6):e0130520.
  • Xu Y, Huang S, Liu Z-G, et al. Poly(ADP-ribose) polymerase-1 signaling to mitochondria in necrotic cell death requires RIP1/TRAF2-mediated JNK1 activation. J Biol Chem. 2006;281(13):8788–8795.
  • Linkermann A, Brasen JH, Darding M, et al. Two independent pathways of regulated necrosis mediate ischemia-reperfusion injury. Proc Natl Acad Sci USA. 2013;110(29):12024–12029.
  • Ying Y, Padanilam BJ. Regulation of necrotic cell death: p53, PARP1 and cyclophilin D-overlapping pathways of regulated necrosis? Cell Mol Life Sci. 2016;73(11–12):2309–2324.
  • Umbro I, Gentile G, Tinti F, et al. Recent advances in pathophysiology and biomarkers of sepsis-induced acute kidney injury. J Infect. 2016;72(2):131–142.
  • Bentala H, Verweij WR, Huizinga-Van der Vlag A, et al. Removal of phosphate from lipid A as a strategy to detoxify lipopolysaccharide. Shock (Augusta, Ga). 2002;18(6):561–566.
  • Peters E, Geraci S, Heemskerk S, et al. Alkaline phosphatase protects against renal inflammation through dephosphorylation of lipopolysaccharide and adenosine triphosphate. Br J Pharmacol. 2015;172(20):4932–4945.
  • Bauerle JD, Grenz A, Kim JH, et al. Adenosine generation and signaling during acute kidney injury. J Am Soc Nephrol. 2011;22(1):14–20.
  • Kaempfer R, Arad G, Levy R, et al. CD28: direct and critical receptor for superantigen toxins. Toxins. 2013;5(9):1531–1542.
  • Ramachandran G, Kaempfer R, Chung CS, et al. CD28 homodimer interface mimetic peptide acts as a preventive and therapeutic agent in models of severe bacterial sepsis and gram-negative bacterial peritonitis. J Infect Dis. 2015;211(6):995–1003.
  • Bragadottir G, Redfors B, Ricksten SE. Effects of levosimendan on glomerular filtration rate, renal blood flow, and renal oxygenation after cardiac surgery with cardiopulmonary bypass: a randomized placebo-controlled study. Crit Care Med. 2013;41(10):2328–2335.
  • Grossini E, Molinari C, Pollesello P, et al. Levosimendan protection against kidney ischemia/reperfusion injuries in anesthetized pigs. J Pharmacol Exp Ther. 2012;342(2):376–388.
  • Zager RA, Johnson AC, Lund S, et al. Levosimendan protects against experimental endotoxemic acute renal failure. Am J Physiol Renal Physiol. 2006;290(6):F1453–62.
  • Yilmaz MB, Grossini E, Silva Cardoso JC, et al. Renal effects of levosimendan: a consensus report. Cardiovasc Drugs Ther. 2013;27(6):581–590.
  • Shi WY, Li S, Collins N, et al. Peri-operative levosimendan in patients undergoing cardiac surgery: an overview of the evidence. Heart Lung Circ. 2015;24(7):667–672.
  • Niu ZZ, Wu SM, Sun WY, et al. Perioperative levosimendan therapy is associated with a lower incidence of acute kidney injury after cardiac surgery: a meta-analysis. J Cardiovasc Pharmacol. 2014;63(2):107–112 .
  • Belletti A, Benedetto U, Biondi-Zoccai G, et al. The effect of vasoactive drugs on mortality in patients with severe sepsis and septic shock. A network meta-analysis of randomized trials. J Crit Care. 2016;37:91–98.
  • Morelli A, De Castro S, Teboul JL, et al. Effects of levosimendan on systemic and regional hemodynamics in septic myocardial depression. Intensive Care Med. 2005;31(5):638–644.
  • Orme RM, Perkins GD, McAuley DF, et al. An efficacy and mechanism evaluation study of levosimendan for the prevention of acute oRgan dysfunction in sepsis (LeoPARDS): protocol for a randomized controlled trial. Trials. 2014;15:199.
  • Humphreys BD, Bonventre JV. Mesenchymal stem cells in acute kidney injury. Annu Rev Med. 2008;59(1):311–325.
  • Kunter U, Rong S, Boor P, et al. Mesenchymal stem cells prevent progressive experimental renal failure but maldifferentiate into glomerular adipocytes. J Am Soc Nephrol. 2007;18(6):1754–1764.
  • Tögel F, Hu Z, Weiss K, et al. Administered mesenchymal stem cells protect against ischemic acute renal failure through differentiation-independent mechanisms. Am J Physiol Ren Physiol. 2005;289(1):F31–F42 .
  • Bersenev A Cell therapy clinical trial failures in 2014 [webpage]. Cells trials: current trends in cell therapy. 2015. [cited 2016 May 15]. Available from: http://celltrials.info/2015/01/04/failures-2014/
  • Star RA, Rajora N, Huang J, et al. Evidence of autocrine modulation of macrophage nitric oxide synthase by alpha-melanocyte-stimulating hormone. Proc Natl Acad Sci USA. 1995;92(17):8016–8020.
  • Chiao H, Kohda Y, McLeroy P, et al. Alpha-melanocyte-stimulating hormone protects against renal injury after ischemia in mice and rats. J Clin Invest. 1997;99(6):1165–1172.
  • Doi K, Hu X, Yuen PST, et al. AP214, an analogue of α-melanocyte-stimulating hormone, ameliorates sepsis-induced acute kidney injury and mortality. Kidney Int. 2008;73(11):1266–1274.
  • Simmons MN, Subramanian V, Crouzet S, et al. Alpha-melanocyte stimulating hormone analogue AP214 protects against ischemia induced acute kidney injury in a porcine surgical model. J Urol. 2010;183(4):1625–1629.
  • Vanden Berghe T, Linkermann A, Jouan-Lanhouet S, et al. Regulated necrosis: the expanding network of non-apoptotic cell death pathways. Nat Rev Mol Cell Biol. 2014;15(2):135–147.
  • Linkermann A. Nonapoptotic cell death in acute kidney injury and transplantation. Kidney Int. 2016;89(1):46–57 .
  • Kim J, Padanilam BJ. Loss of poly(ADP-ribose) polymerase 1 attenuates renal fibrosis and inflammation during unilateral ureteral obstruction. Am J Physiol Ren Physiol. 2011;301(2):F450–F9.
  • Linkermann A, Brasen JH, Himmerkus N, et al. Rip1 (receptor-interacting protein kinase 1) mediates necroptosis and contributes to renal ischemia/reperfusion injury. Kidney Int. 2012;81(8):751–761.
  • Lau A, Wang S, Jiang J, et al. RIPK3-mediated necroptosis promotes donor kidney inflammatory injury and reduces allograft survival. Am J Transplant. 2013;13(11):2805–2818.
  • Linkermann A, Skouta R, Himmerkus N, et al. Synchronized renal tubular cell death involves ferroptosis. Proc Natl Acad Sci USA. 2014;111(47):16836–16841.
  • Jiang L, Kon N, Li T, et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 2015;520(7545):57–62.
  • Mulay SR, Desai J, Kumar SV, et al. Cytotoxicity of crystals involves RIPK3-MLKL-mediated necroptosis. Nat Commun. 2016;7:10274.
  • Mulay SR, Linkermann A, Anders HJ. Necroinflammation in kidney disease. J Am Soc Nephrol. 2016;27(1):27–39.
  • Shutinoski B, Alturki NA, Rijal D, et al. K45A mutation of RIPK1 results in poor necroptosis and cytokine signaling in macrophages, which impacts inflammatory responses in vivo. Cell Death Differ. 2016;23:1628–1637.
  • Najjar M, Saleh D, Zelic M, et al. RIPK1 and RIPK3 kinases promote cell-death-independent inflammation by toll-like receptor 4. Immunity. 2016;45(1):46–59.
  • Birch RE, Schwiebert EM, Peppiatt-Wildman CM, et al. Emerging key roles for P2X receptors in the kidney. Front Physiol. 2013;4:262.
  • Yan Y, Bai J, Zhou X, et al. P2X7 receptor inhibition protects against ischemic acute kidney injury in mice. Am J Physiol Cell Physiol. 2015;308(6):C463–72.
  • Fremeaux-Bacchi V, Legendre CM. The emerging role of complement inhibitors in transplantation. Kidney Int. 2015;88(5):967–973.
  • Patel H, Smith RAG, Sacks SH, et al. Therapeutic strategy with a membrane-localizing complement regulator to increase the number of usable donor organs after prolonged cold storage. J Am Soc Nephrol. 2006;17(4):1102–1111.
  • Leaf DE, Waikar SS. End points for clinical trials in acute kidney injury. Am J Kidney Dis. 2016. doi:10.1053/j.ajkd.2016.05.033. [Epub ahead of print]
  • de Caestecker M, Humphreys BD, Liu KD, et al. Bridging translation by improving preclinical study design in AKI. J Am Soc Nephrol. 2015;26(12):2905–2916.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.