Teprasiran, a Small Interfering RNA, for the Prevention of Acute Kidney Injury in High-Risk Patients Undergoing Cardiac Surgery: A Randomized Clinical Study

Matthias Thielmann, David Corteville, Gabor Szabo, Madhav Swaminathan, Andre Lamy, Lukas J Lehner, Craig D Brown, Nicolas Noiseux, Mohamed G Atta, Elizabeth C Squiers, Shai Erlich, Daniel Rothenstein, Bruce Molitoris, C David Mazer, Matthias Thielmann, David Corteville, Gabor Szabo, Madhav Swaminathan, Andre Lamy, Lukas J Lehner, Craig D Brown, Nicolas Noiseux, Mohamed G Atta, Elizabeth C Squiers, Shai Erlich, Daniel Rothenstein, Bruce Molitoris, C David Mazer

Abstract

Background: Acute kidney injury (AKI) affects up to 30% of patients undergoing cardiac surgery, leading to increased in-hospital and long-term morbidity and mortality. Teprasiran is a novel small interfering RNA that temporarily inhibits p53-mediated cell death that underlies AKI.

Methods: This prospective, multicenter, double-blind, randomized, controlled phase 2 trial evaluated the efficacy and safety of a single 10 mg/kg dose of teprasiran versus placebo (1:1), in reducing the incidence, severity, and duration of AKI after cardiac surgery in high-risk patients. The primary end point was the proportion of patients who developed AKI determined by serum creatinine by postoperative day 5. Other end points included AKI severity and duration using various prespecified criteria. To inform future clinical development, a composite end point of major adverse kidney events at day 90, including death, renal replacement therapy, and ≥25% reduction of estimated glomerular filtration rate was assessed. Both serum creatinine and serum cystatin-C were used for estimated glomerular filtration rate assessments.

Results: A total of 360 patients were randomly assigned in 41 centers; 341 dosed patients were 73±7.5 years of age (mean±SD), 72% were men, and median European System for Cardiac Operative Risk Evaluation score was 2.6%. Demographics and surgical parameters were similar between groups. AKI incidence was 37% for teprasiran- versus 50% for placebo-treated patients, a 12.8% absolute risk reduction, P=0.02; odds ratio, 0.58 (95% CI, 0.37-0.92). AKI severity and duration were also improved with teprasiran: 2.5% of teprasiran- versus 6.7% of placebo-treated patients had grade 3 AKI; 7% teprasiran- versus 13% placebo-treated patients had AKI lasting for 5 days. No significant difference was observed for the major adverse kidney events at day 90 composite in the overall population. No safety issues were identified with teprasiran treatment.

Conclusions: The incidence, severity, and duration of early AKI in high-risk patients undergoing cardiac surgery were significantly reduced after teprasiran administration. A phase 3 study with a major adverse kidney event at day 90 primary outcome that has recently completed enrollment was designed on the basis of these findings (NCT03510897). Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT02610283.

Keywords: RNA, small interfering; acute kidney injury; thoracic surgery.

Figures

Figure 1.
Figure 1.
Patient disposition. Patients were assigned in a 1:1 ratio to the teprasiran or placebo groups. Of the 360 patients randomly assigned, 19 patients (6 assigned to placebo and 13 assigned to teprasiran) were discontinued before dosing because of events arising during or after the surgery (eg, death, physician decision, withdrawal by subject, or hemodynamic instability). Of those dosed, 93.2% of the placebo group and 92.7% of the teprasiran group completed the day 90 visit. *Two subjects (1 subject in each treatment group) died after study day 90. Both subjects experienced SAEs with fatal outcomes that began before study day 90.
Figure 2.
Figure 2.
Subgroup analysis of the primary end point (modified intention-to-treat–baseline serum creatinine required analysis set). The forest plot presents, for each subgroup, acute kidney injury odds ratio (OR) between treatments by a dot and its 95% CI by a horizontal line. The size of the dot varies proportionally with the total (Nteprasiran+Nplacebo) sample size of each subgroup. On the left side of the figure, the subgroup sample size by treatment is presented with the corresponding acute kidney injury rate. The column on the right lists the odds ratio and its 95% CI; the x-axis of the plot is in log scale. P values for odds ratio interaction between subgroups are as follows: age=0.17, CPB time=0.98, diabetes=0.34, eGFR=0.71, procedure=0.43. CPB indicates cardiopulmonary bypass; and eGFR, estimated glomerular filtration rate. *Diabetes requiring insulin treatment.
Figure 3.
Figure 3.
Serum creatinine and serum cystatin C dynamics over time. Serum cystatin C values over time per cohort are presented as absolute (A) and change from baseline (BL; B). After a decrease in values on day 0 (day of surgery), an increase in serum cystatin C is observed in the acute postoperative period starting on day 1 in both treatment cohorts. The observed increase is maintained through day 90. Serum creatinine (sCr) values over time per cohort as absolute (C) and change from baseline (D). A rapid increase is observed on day 0 (day of surgery), reaching peak values on days 2 to 3 and decreasing to near presurgery values by day 5. sCr values at day 90 closely resemble baseline sCr values.

References

    1. Huen SC, Parikh CR. Predicting acute kidney injury after cardiac surgery: a systematic review. Ann Thorac Surg. 2012;93:337–347. doi: 10.1016/j.athoracsur.2011.09.010
    1. Lamy A, Devereaux PJ, Prabhakaran D, Taggart DP, Hu S, Paolasso E, Straka Z, Piegas LS, Akar AR, Jain AR, et al. ; CORONARY Investigators. Off-pump or on-pump coronary-artery bypass grafting at 30 days. N Engl J Med. 2012;366:1489–1497. doi: 10.1056/NEJMoa1200388
    1. Wang Y, Bellomo R. Cardiac surgery-associated acute kidney injury: risk factors, pathophysiology and treatment. Nat Rev Nephrol. 2017;13:697–711. doi: 10.1038/nrneph.2017.119
    1. Meersch M, Schmidt C, Hoffmeier A, Van Aken H, Wempe C, Gerss J, Zarbock A. Prevention of cardiac surgery-associated AKI by implementing the KDIGO guidelines in high risk patients identified by biomarkers: the PrevAKI randomized controlled trial. Intensive Care Med. 2017;43:1551–1561. doi: 10.1007/s00134-016-4670-3
    1. Hobson C, Ozrazgat-Baslanti T, Kuxhausen A, Thottakkara P, Efron PA, Moore FA, Moldawer LL, Segal MS, Bihorac A. Cost and mortality associated with postoperative acute kidney injury. Ann Surg. 2015;261:1207–1214. doi: 10.1097/SLA.0000000000000732
    1. Ozrazgat-Baslanti T, Thottakkara P, Huber M, Berg K, Gravenstein N, Tighe P, Lipori G, Segal MS, Hobson C, Bihorac A. Acute and chronic kidney disease and cardiovascular mortality after major surgery. Ann Surg. 2016;264:987–996. doi: 10.1097/SLA.0000000000001582
    1. Korenkevych D, Ozrazgat-Baslanti T, Thottakkara P, Hobson CE, Pardalos P, Momcilovic P, Bihorac A. The pattern of longitudinal change in serum creatinine and 90-day mortality after major surgery. Ann Surg. 2016;263:1219–1227. doi: 10.1097/SLA.0000000000001362
    1. Rimes-Stigare C, Ravn B, Awad A, Torlén K, Martling CR, Bottai M, Mårtensson J, Bell M. Creatinine- and cystatin c-based incidence of chronic kidney disease and acute kidney disease in AKI survivors. Crit Care Res Pract. 2018;2018:7698090. doi: 10.1155/2018/7698090
    1. James MT, Bhatt M, Pannu N, Tonelli M. Long-term outcomes of acute kidney injury and strategies for improved care. Nat Rev Nephrol. 2020;16:193–205. doi: 10.1038/s41581-019-0247-z
    1. Ranjan A, Iwakuma T. Non-canonical cell death induced by p53. Int J Mol Sci. 2016;17:E2068. doi: 10.3390/ijms17122068
    1. Jiang L, Sheikh MS, Huang Y. Decision making by p53: life versus death. Mol Cell Pharmacol. 2010;2:69–77
    1. Komarov PG, Komarova EA, Kondratov RV, Christov-Tselkov K, Coon JS, Chernov MV, Gudkov AV. A chemical inhibitor of p53 that protects mice from the side effects of cancer therapy. Science. 1999;285:1733–1737. doi: 10.1126/science.285.5434.1733
    1. Tang C, Ma Z, Zhu J, Liu Z, Liu Y, Liu Y, Cai J, Dong Z. P53 in kidney injury and repair: mechanism and therapeutic potentials. Pharmacol Ther. 2019;195:5–12. doi: 10.1016/j.pharmthera.2018.10.013
    1. McLaren BK, Zhang PL, Herrera GA. P53 protein is a reliable marker in identification of renal tubular injury. Appl Immunohistochem Mol Morphol. 2004;12:225–229. doi: 10.1097/00129039-200409000-00007
    1. Kim MG, Yang J, Ko YS, Lee HY, Oh SW, Cho WY, Jo SK. Impact of aging on transition of acute kidney injury to chronic kidney disease. Sci Rep. 2019;9:18445. doi: 10.1038/s41598-019-54585-1
    1. Zhang D, Liu Y, Wei Q, Huo Y, Liu K, Liu F, Dong Z. Tubular p53 regulates multiple genes to mediate AKI. J Am Soc Nephrol. 2014;25:2278–2289. doi: 10.1681/ASN.2013080902
    1. Christophorou MA, Ringshausen I, Finch AJ, Swigart LB, Evan GI. The pathological response to DNA damage does not contribute to p53-mediated tumour suppression. Nature. 2006;443:214–217. doi: 10.1038/nature05077
    1. Gudkov AV, Komarova EA. Pathologies associated with the p53 response. Cold Spring Harb Perspect Biol. 2010;2:a001180. doi: 10.1101/cshperspect.a001180
    1. Molitoris BA, Dagher PC, Sandoval RM, Campos SB, Ashush H, Fridman E, Brafman A, Faerman A, Atkinson SJ, Thompson JD, et al. . siRNA targeted to p53 attenuates ischemic and cisplatin-induced acute kidney injury. J Am Soc Nephrol. 2009;20:1754–1764. doi: 10.1681/ASN.2008111204
    1. Imamura R, Isaka Y, Sandoval RM, Ori A, Adamsky S, Feinstein E, Molitoris BA, Takahara S. Intravital two-photon microscopy assessment of renal protection efficacy of siRNA for p53 in experimental rat kidney transplantation models. Cell Transplant. 2010;19:1659–1670. doi: 10.3727/096368910X516619
    1. Demirjian S, Ailawadi G, Polinsky M, Bitran D, Silberman S, Shernan SK, Burnier M, Hamilton M, Squiers E, Erlich S, et al. . Safety and tolerability study of an intravenously administered small interfering ribonucleic acid (siRNA) post on-pump cardiothoracic surgery in patients at risk of acute kidney injury. Kidney Int Rep. 2017;2:836–843. doi: 10.1016/j.ekir.2017.03.016
    1. Levey AS, Coresh J, Tighiouart H, Greene T, Inker LA. Strengths and limitations of estimated and measured GFR. Nat Rev Nephrol. 2019;15:784. doi: 10.1038/s41581-019-0213-9
    1. Sandilands EA, Dhaun N, Dear JW, Webb DJ. Measurement of renal function in patients with chronic kidney disease. Br J Clin Pharmacol. 2013;76:504–515. doi: 10.1111/bcp.12198
    1. von Scholten BJ, Persson F, Svane MS, Hansen TW, Madsbad S, Rossing P. Effect of large weight reductions on measured and estimated kidney function. BMC Nephrol. 2017;18:52. doi: 10.1186/s12882-017-0474-0
    1. Brondén B, Eyjolfsson A, Blomquist S, Dardashti A, Ederoth P, Bjursten H. Evaluation of cystatin C with iohexol clearance in cardiac surgery. Acta Anaesthesiol Scand. 2011;55:196–202. doi: 10.1111/j.1399-6576.2010.02361.x
    1. Zhu J, Yin R, Wu H, Yi J, Luo L, Dong G, Jing H. Cystatin C as a reliable marker of renal function following heart valve replacement surgery with cardiopulmonary bypass. Clin Chim Acta. 2006;374:116–121. doi: 10.1016/j.cca.2006.06.001
    1. Jonsson AS, Flodin M, Hansson LO, Larsson A. Estimated glomerular filtration rate (eGFRCystC) from serum cystatin C shows strong agreement with iohexol clearance in patients with low GFR. Scand J Clin Lab Invest. 2007;67:801–809. doi: 10.1080/00365510701397538
    1. Kervella D, Lemoine S, Sens F, Dubourg L, Sebbag L, Guebre-Egziabher F, Bonnefoy E, Juillard L. Cystatin C versus creatinine for GFR estimation in CKD due to heart failure. Am J Kidney Dis. 2017;69:321–323. doi: 10.1053/j.ajkd.2016.09.016
    1. Dardashti A, Nozohoor S, Grubb A, Bjursten H. Shrunken pore syndrome is associated with a sharp rise in mortality in patients undergoing elective coronary artery bypass grafting. Scand J Clin Lab Invest. 2016;76:74–81. doi: 10.3109/00365513.2015.1099724
    1. Mooney JF, Croal BL, Cassidy S, Lee VW, Chow CK, Cuthbertson BH, Hillis GS. Relative value of cystatin C and creatinine-based estimates of glomerular filtration rate in predicting long-term mortality after cardiac surgery: a cohort study. BMJ Open. 2019;9:e029379. doi: 10.1136/bmjopen-2019-029379
    1. Ray KK, Wright RS, Kallend D, Koenig W, Leiter LA, Raal FJ, Bisch JA, Richardson T, Jaros M, Wijngaard PLJ, et al. ; ORION-10 and ORION-11 Investigators. Two phase 3 trials of inclisiran in patients with elevated LDL cholesterol. N Engl J Med. 2020;382:1507–1519. doi: 10.1056/NEJMoa1912387
    1. Billings FT, Hendricks PA, Schildcrout JS, Shi Y, Petracek MR, Byrne JG, Brown NJ. High-dose perioperative atorvastatin and acute kidney injury following cardiac surgery: a randomized clinical trial. JAMA. 2016;315:877–888
    1. Menting TP, Wever KE, Hendriks EJ, Van der Vliet DJA, Rovers MM, Warle MC, Menting TP. Ischaemic preconditioning for the reduction of renal ischaemia reperfusion injury. Cochrane Database of Syst Rev. 2017;3:CD010777.
    1. Bailey M, McGuinness S, Haase M, Haase-Fielitz A, Parke R, Hodgson CL, Forbes A, Bagshaw SM, Bellomo R. Sodium bicarbonate and renal function after cardiac surgery: a prospectively planned individual patient meta-analysis. Anesthesiology. 2015;122:294–306. doi: 10.1097/ALN.0000000000000547
    1. Bove T, Zangrillo A, Guarracino F, Alvaro G, Persi B, Maglioni E, Galdieri N, Comis M, Caramelli F, Pasero DC, et al. . Effect of fenoldopam on use of renal replacement therapy among patients with acute kidney injury after cardiac surgery: a randomized clinical trial. JAMA. 2014;312:2244–2253. doi: 10.1001/jama.2014.13573
    1. Bragadottir G, Redfors B, Ricksten SE. Mannitol increases renal blood flow and maintains filtration fraction and oxygenation in postoperative acute kidney injury: a prospective interventional study. Crit Care. 2012;16:R159. doi: 10.1186/cc11480
    1. McCullough PA, Bennett-Guerrero E, Chawla LS, Beaver T, Mehta RL, Molitoris BA, Eldred A, Ball G, Lee HJ, Houser MT, 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:e003549. doi: 10.1161/JAHA.116.003549
    1. Ederoth P, Grins E, Dardashti A, Brondén B, Metzsch C, Erdling A, Nozohoor S, Mokhtari A, Hansson MJ, Elmér E, et al. . Ciclosporin to Protect Renal function In Cardiac Surgery (CiPRICS): a study protocol for a double-blind, randomised, placebo-controlled, proof-of-concept study. BMJ Open. 2016;6:e012299. doi: 10.1136/bmjopen-2016-012299
    1. Swaminathan M, Stafford-Smith M, Chertow GM, Warnock DG, Paragamian V, Brenner RM, Lellouche F, Fox-Robichaud A, Atta MG, Melby S, et al. ; ACT-AKI investigators. Allogeneic mesenchymal stem cells for treatment of AKI after cardiac surgery. J Am Soc Nephrol. 2018;29:260–267. doi: 10.1681/ASN.2016101150
    1. Himmelfarb J, Chertow GM, McCullough PA, Mesana T, Shaw AD, Sundt TM, Brown C, Cortville D, Dagenais F, de Varennes B, et al. . Perioperative THR-184 and AKI after cardiac surgery. J Am Soc Nephrol. 2018;29:670–679. doi: 10.1681/ASN.2017020217

Source: PubMed

3
Abonner