The STAGED-PKD 2-Stage Adaptive Study With a Patient Enrichment Strategy and Treatment Effect Modeling for Improved Study Design Efficiency in Patients With ADPKD

Ronald D Perrone, Ali Hariri, Pascal Minini, Curie Ahn, Arlene B Chapman, Shigeo Horie, Bertrand Knebelmann, Michal Mrug, Albert C M Ong, York P C Pei, Vicente E Torres, Vijay Modur, Ronald T Gansevoort, Ronald D Perrone, Ali Hariri, Pascal Minini, Curie Ahn, Arlene B Chapman, Shigeo Horie, Bertrand Knebelmann, Michal Mrug, Albert C M Ong, York P C Pei, Vicente E Torres, Vijay Modur, Ronald T Gansevoort

Abstract

Rationale & objective: Venglustat, a glucosylceramide synthase inhibitor, inhibits cyst growth and reduces kidney failure in mouse models of autosomal dominant polycystic kidney disease (ADPKD). STAGED-PKD aims to determine the safety and efficacy of venglustat and was designed using patient enrichment for progression to end-stage kidney disease and modeling from prior ADPKD trials.

Study design: STAGED-PKD is a 2-stage, international, double-blind, randomized, placebo-controlled trial in adults with ADPKD (Mayo Class 1C-1E) and estimated glomerular filtration rate (eGFR) 45-<90 mL/min/1.73 m2 at risk of rapidly progressive disease. Enrichment for rapidly progressing patients was identified based on retrospective analysis of total kidney volume (TKV) and eGFR slope from the combined Consortium for Radiologic Imaging Studies of Polycystic Kidney Disease and HALT Progression of Polycystic Kidney Disease A studies.

Setting & participants: Target enrollment in stages 1 and 2 was 240 and 320 patients, respectively.

Interventions: Stage 1 randomizes patients 1:1:1 to venglustat 8 mg or 15 mg once daily or placebo. Stage 2 randomizes patients 1:1 to placebo or venglustat, with the preferred dose based on stage 1 safety data.

Outcomes: Primary endpoints are TKV growth rate over 18 months in stage 1 and eGFR slope over 24 months in stage 2. Secondary endpoints include: annualized rate of change in eGFR from baseline to 18 months (stage 1); annualized rate of change in TKV based on magnetic resonance imaging from baseline to 18 months (stage 2); and safety, tolerability, pain, and fatigue (stages 1 and 2).

Limitations: If stage 1 is unsuccessful, patients enrolled in the trial may develop drug-related adverse events that can have long-lasting effects.

Conclusions: Modeling allows the design and powering of a 2-stage combined study to assess venglustat's impact on TKV growth and eGFR slope. Stage 1 TKV assessment via a nested approach allows early evaluation of efficacy and increased efficiency of the trial design by reducing patient numbers and trial duration.

Funding: This study was funded by Sanofi.

Trial registration: STAGED-PKD has been registered at ClinicalTrials.gov with study number NCT03523728.

Keywords: Autosomal dominant polycystic kidney disease; modeling; study design; total kidney volume; venglustat.

© 2022 The Authors.

Figures

Figure 1
Figure 1
GSL biosynthesis in ADPKD and potential effect of the GCS inhibitor venglustat. Adapted from Natoli et al. Loss of polycystin function disrupts the TSC complex, and consequently suppression of Rheb is inactivated, leading to activation of mTORC1. mTORC1 activation increases de novo ceramide synthesis. Polycystin dysregulation also activates mTORC2, which also promotes de novo ceramide synthesis and increases GL-1 by increasing GCS production. Beyond polycystin disruption, other factors may impact GSL accumulation in ADPKD, including growth factor activation (eg, EGF 1 or IGF 1) of mTORC2 or cytokine- and ROS-mediated activation of sphingomyelinase activity. Red boxes show molecules that are upregulated in cystic kidneys compared with normal kidneys. Yellow box overlay indicates GSL accumulation that could be attenuated/stopped with a GCS inhibitor. Abbreviations: ADPKD, autosomal dominant polycystic kidney disease; EGF, epidermal growth factor; IGF, insulin-like growth factor; GCS, glucosylceramide synthase; GL-1, glucosylceramide; GM3, monosialodihexosylganglioside; GSL, glycosphingolipid; mTORC, mammalian target of rapamycin complex; Rheb, Ras homolog enriched in brain; ROS, reactive oxygen species; RTK, receptor tyrosine kinase; TNF, tumor necrosis factor; TSC, tuberous sclerosis.
Figure 2
Figure 2
Class-level data from the CRISP and HALT-PKD A studies. Based on retrospective analysis of 2 historical studies. For each class, mean TKV growth rate is plotted against mean eGFR rate of decline. The size of each bubble is proportional to the sample size. Abbreviations: CRISP, Consortium for Radiologic Imaging Studies of Polycystic Kidney Disease; eGFR, estimated glomerular filtration rate; HALT-PKD, HALT Progression of Polycystic Kidney Disease; STAGED-PKD, study to assess glucosylceramide synthase inhibitor efficacy in ADPKD; TKV, total kidney volume.
Figure 3
Figure 3
Correlation between TKV growth and eGFR decline based on individual-level data from CRISP and HALT-PKD A. Predicted eGFR rate of decline based on a model predicting eGFR at time t as a function of TKV growth rate and adjusted for baseline eGFR and age. Abbreviations: CRISP, Consortium for Radiologic Imaging Studies of Polycystic Kidney Disease; eGFR, estimated glomerular filtration rate; HALT-PKD, HALT Progression of Polycystic Kidney Disease; TKV, total kidney volume.
Figure 4
Figure 4
Study design and key steps in STAGED-PKD stage 1 (A) and stage 2 (B). ∗To ensure adequate representation of patients across the spectrum of eGFR, a minimum of 20% of patients are enrolled in each of the following categories: ≥45-<60 mL/min/1.73 m2; ≥60-<75 mL/min/1.73 m2; and ≥75-<90 mL/min/1.73 m2; †Highest dose determined to be well tolerated in stage 1. Abbreviations: ADPKD, autosomal dominant polycystic kidney disease; eGFR, estimated glomerular filtration rate; ROW, rest of the world; STAGED-PKD, study to assess glucosylceramide synthase inhibitor efficacy in ADPKD.

References

    1. Gabow P.A. Autosomal dominant polycystic kidney disease. N Engl J Med. 1993;329(5):332–342.
    1. Grantham J.J., Torres V.E. The importance of total kidney volume in evaluating progression of polycystic kidney disease. Nat Rev Nephrol. 2016;12(11):667–677.
    1. Merrill A.H., Jr. Sphingolipid and glycosphingolipid metabolic pathways in the era of sphingolipidomics. Chem Rev. 2011;111(10):6387–6422.
    1. Natoli T.A., Modur V., Ibraghimov-Beskrovnaya O. Glycosphingolipid metabolism and polycystic kidney disease. Cell Signal. 2020;69
    1. Peterschmitt M.J., Crawford N.P.S., Gaemers S.J.M., Ji A.J., Sharma J., Pham T.T. Pharmacokinetics, pharmacodynamics, safety, and tolerability of oral venglustat in healthy volunteers. Clin Pharmacol Drug Dev. 2021;10(1):86–98.
    1. US Food and Drug Administration Orphan drug designations and approvals – venglustat. Published May 1, 2018.
    1. European Medicines Agency Public summary of opinion on orphan designation – venglustat for the treatment of autosomal polycystic kidney disease. Published February 25, 2019.
    1. Smith K.A., Thompson A.M., Baron D.A., Broadbent S.T., Lundstrom G.H., Perrone R.D. Addressing the need for clinical trial end points in autosomal dominant polycystic kidney disease: a report from the polycystic kidney disease outcomes consortium (PKDOC) Am J Kidney Dis. 2019;73(4):533–541.
    1. Irazabal M.V., Rangel L.J., Bergstralh E.J., et al. Imaging classification of autosomal dominant polycystic kidney disease: a simple model for selecting patients for clinical trials. J Am Soc Nephrol. 2015;26(1):160–172.
    1. Yu A.S.L., Shen C., Landsittel D.P., et al. Long-term trajectory of kidney function in autosomal-dominant polycystic kidney disease. Kidney Int. 2019;95(5):1253–1261.
    1. Bae K.T., Zhou W., Shen C., et al. Growth pattern of kidney cyst number and volume in autosomal dominant polycystic kidney disease. Clin J Am Soc Nephrol. 2019;14(6):823–833.
    1. Chapman A.B., Bost J.E., Torres V.E., et al. Kidney volume and functional outcomes in autosomal dominant polycystic kidney disease. Clin J Am Soc Nephrol. 2012;7(3):479–486.
    1. PKD Consortium Clinical Path Institute Qualification of total kidney volume as a prognostic biomarker for use in clinical trials evaluating patients with Autosomal Dominant Polycystic Kidney Disease (ADPKD) Published March 20, 2014.
    1. US Food and Drug Administration FDA facilitates the use of surrogate endpoints in drug development. Published November 5, 2018.
    1. US Food and Drug Administration Table of surrogate endpoints that were the basis of drug approval or licensure. Published 2018.
    1. Grantham J.J., Chapman A.B., Torres V.E. Volume progression in autosomal dominant polycystic kidney disease: the major factor determining clinical outcomes. Clin J Am Soc Nephrol. 2006;1(1):148–157.
    1. Schrier R.W., Abebe K.Z., Perrone R.D., et al. Blood pressure in early autosomal dominant polycystic kidney disease. N Engl J Med. 2014;371(24):2255–2266.
    1. Torres V.E., Abebe K.Z., Chapman A.B., et al. Angiotensin blockade in late autosomal dominant polycystic kidney disease. N Engl J Med. 2014;371(24):2267–2276.
    1. Torres V.E., Chapman A.B., Devuyst O., et al. Tolvaptan in patients with autosomal dominant polycystic kidney disease. N Engl J Med. 2012;367(25):2407–2418.
    1. Torres V.E., Chapman A.B., Devuyst O., et al. Tolvaptan in later-stage autosomal dominant polycystic kidney disease. N Engl J Med. 2017;377(20):1930–1942.
    1. Perrone R.D., Chapman A.B., Oberdhan D., et al. The NOCTURNE randomized trial comparing 2 tolvaptan formulations. Kidney Int Rep. 2020;5(6):801–812.
    1. Ku E., Xie D., Shlipak M., et al. Change in measured GFR versus eGFR and CKD outcomes. J Am Soc Nephrol. 2016;27(7):2196–2204.
    1. Levey A.S., Gansevoort R.T., 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(1):84–104.
    1. Natoli T.A., Smith L.A., Rogers K.A., et al. Inhibition of glucosylceramide accumulation results in effective blockade of polycystic kidney disease in mouse models. Nat Med. 2010;16(7):788–792.
    1. Chapman A.B. Autosomal dominant polycystic kidney disease: time for a change? J Am Soc Nephrol. 2007;18(5):1399–1407.
    1. Bretz F., Maurer W., Brannath W., Posch M. A graphical approach to sequentially rejective multiple test procedures. Stat Med. 2009;28(4):586–604.
    1. Irazabal M.V., Blais J.D., Perrone R.D., et al. Prognostic enrichment design in clinical trials for autosomal dominant polycystic kidney disease: the TEMPO 3:4 clinical trial. Kidney Int Rep. 2016;1(4):213–220.
    1. Caroli A., Perico N., Perna A., et al. Effect of longacting somatostatin analogue on kidney and cyst growth in autosomal dominant polycystic kidney disease (ALADIN): a randomised, placebo-controlled, multicentre trial. Lancet. 2013;382(9903):1485–1495.
    1. Meijer E., Visser F.W., van Aerts R.M.M., et al. Effect of lanreotide on kidney function in patients with autosomal dominant polycystic kidney disease: the DIPAK 1 randomized clinical trial. JAMA. 2018;320(19):2010–2019.
    1. Perico N., Ruggenenti P., Perna A., et al. Octreotide-LAR in later-stage autosomal dominant polycystic kidney disease (ALADIN 2): a randomized, double-blind, placebo-controlled, multicenter trial. PLoS Med. 2019;16(4)
    1. Chow S.C., Lin M. Analysis of two-stage adaptive seamless trial design. Pharm Anal Acta. 2015;6(3)
    1. Barnawi R.A., Attar R.Z., Alfaer S.S., Safdar O.Y. Is the light at the end of the tunnel nigh? A review of ADPKD focusing on the burden of disease and tolvaptan as a new treatment. Int J Nephrol Renovasc Dis. 2018;11:53–67.

Source: PubMed

3
구독하다