Octreotide-LAR in later-stage autosomal dominant polycystic kidney disease (ALADIN 2): A randomized, double-blind, placebo-controlled, multicenter trial

Norberto Perico, Piero Ruggenenti, Annalisa Perna, Anna Caroli, Matias Trillini, Sandro Sironi, Antonio Pisani, Eleonora Riccio, Massimo Imbriaco, Mauro Dugo, Giovanni Morana, Antonio Granata, Michele Figuera, Flavio Gaspari, Fabiola Carrara, Nadia Rubis, Alessandro Villa, Sara Gamba, Silvia Prandini, Monica Cortinovis, Andrea Remuzzi, Giuseppe Remuzzi, ALADIN 2 Study Group, Norberto Perico, Piero Ruggenenti, Annalisa Perna, Anna Caroli, Matias Trillini, Sandro Sironi, Antonio Pisani, Eleonora Riccio, Massimo Imbriaco, Mauro Dugo, Giovanni Morana, Antonio Granata, Michele Figuera, Flavio Gaspari, Fabiola Carrara, Nadia Rubis, Alessandro Villa, Sara Gamba, Silvia Prandini, Monica Cortinovis, Andrea Remuzzi, Giuseppe Remuzzi, ALADIN 2 Study Group

Abstract

Background: Autosomal dominant polycystic kidney disease (ADPKD) is the most frequent genetically determined renal disease. In affected patients, renal function may progressively decline up to end-stage renal disease (ESRD), and approximately 10% of those with ESRD are affected by ADPKD. The somatostatin analog octreotide long-acting release (octreotide-LAR) slows renal function deterioration in patients in early stages of the disease. We evaluated the renoprotective effect of octreotide-LAR in ADPKD patients at high risk of ESRD because of later-stage ADPKD.

Methods and findings: We did an internally funded, parallel-group, double-blind, placebo-controlled phase III trial to assess octreotide-LAR in adults with ADPKD with glomerular filtration rate (GFR) 15-40 ml/min/1.73 m2. Participants were randomized to receive 2 intramuscular injections of 20 mg octreotide-LAR (n = 51) or 0.9% sodium chloride solution (placebo; n = 49) every 28 days for 3 years. Central randomization was 1:1 using a computerized list stratified by center and presence or absence of diabetes or proteinuria. Co-primary short- and long-term outcomes were 1-year total kidney volume (TKV) (computed tomography scan) growth and 3-year GFR (iohexol plasma clearance) decline. Analyses were by modified intention-to-treat. Patients were recruited from 4 Italian nephrology units between October 11, 2011, and March 20, 2014, and followed up to April 14, 2017. Baseline characteristics were similar between groups. Compared to placebo, octreotide-LAR reduced median (95% CI) TKV growth from baseline by 96.8 (10.8 to 182.7) ml at 1 year (p = 0.027) and 422.6 (150.3 to 695.0) ml at 3 years (p = 0.002). Reduction in the median (95% CI) rate of GFR decline (0.56 [-0.63 to 1.75] ml/min/1.73 m2 per year) was not significant (p = 0.295). TKV analyses were adjusted for age, sex, and baseline TKV. Over a median (IQR) 36 (24 to 37) months of follow-up, 9 patients on octreotide-LAR and 21 patients on placebo progressed to a doubling of serum creatinine or ESRD (composite endpoint) (hazard ratio [HR] [95% CI] adjusted for age, sex, baseline serum creatinine, and baseline TKV: 0.307 [0.127 to 0.742], p = 0.009). One composite endpoint was prevented for every 4 treated patients. Among 63 patients with chronic kidney disease (CKD) stage 4, 3 on octreotide-LAR and 8 on placebo progressed to ESRD (adjusted HR [95% CI]: 0.121 [0.017 to 0.866], p = 0.036). Three patients on placebo had a serious renal cyst rupture/infection and 1 patient had a serious urinary tract infection/obstruction, versus 1 patient on octreotide-LAR with a serious renal cyst infection. The main study limitation was the small sample size.

Conclusions: In this study we observed that in later-stage ADPKD, octreotide-LAR slowed kidney growth and delayed progression to ESRD, in particular in CKD stage 4.

Trial registration: ClinicalTrials.gov NCT01377246; EudraCT: 2011-000138-12.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Trial profile.
Fig 1. Trial profile.
CT, computed tomography; ESRD, end-stage renal disease; GFR, glomerular filtration rate; octreotide-LAR, octreotide long-acting release; TKV, total kidney volume.
Fig 2. Absolute changes in total kidney…
Fig 2. Absolute changes in total kidney volume from baseline to 1-year and 3-year follow-up.
Absolute changes in total kidney volume from baseline to 1-year (primary short-term outcome) and 3-year follow-up in patients randomized to either placebo or octreotide-LAR. Data are reported as median and interquartile range. P values from non-parametric ANCOVA adjusted for age, sex and baseline total kidney volume. Analysis performed including all non-missing data. octreotide-LAR, octreotide long-acting release.
Fig 3. Kaplan–Meier curves for the secondary…
Fig 3. Kaplan–Meier curves for the secondary composite endpoint of doubling of serum creatinine or ESRD.
Kaplan–Meier curves show the proportion of patients who reached the composite endpoint of doubling of serum creatinine or ESRD in the placebo and octreotide-LAR groups during the 3-year study period. *Adjusted by age, sex, and baseline serum creatinine.°Adjusted by age, sex, and baseline serum creatinine and total kidney volume. ESRD, end-stage renal disease; HR, hazard ratio; octreotide-LAR, octreotide long-acting release.
Fig 4. Kaplan–Meier curves for the secondary…
Fig 4. Kaplan–Meier curves for the secondary composite endpoint of doubling of serum creatinine or ESRD and for the single endpoint of ESRD in patients with CKD stage 4.
Kaplan–Meier curves show the proportion of patients with CKD stage 4 who reached (A) the composite endpoint of doubling of serum creatinine or ESRD or (B) ESRD considered as a single endpoint (secondary outcomes) in the placebo and octreotide-LAR groups during the 3-year study period. *Adjusted by age, sex, and baseline serum creatinine.°Adjusted by age, sex, and baseline serum creatinine and total kidney volume. CKD, chronic kidney disease; ESRD, end-stage renal disease; HR, hazard ratio; octreotide LAR, octreotide long-acting release.

References

    1. Ong AC, Devuyst O, Knebelmann B, Walz G. Autosomal dominant polycystic kidney disease: the changing face of clinical management. Lancet. 2015;385:1993–2002. 10.1016/S0140-6736(15)60907-2
    1. Spithoven EM, Kramer A, Meijer E, Orskov B, Wanner C, Caskey F, et al. Analysis of data from the ERA-EDTA Registry indicates that conventional treatments for chronic kidney disease do not reduce the need for renal replacement therapy in autosomal dominant polycystic kidney disease. Kidney Int. 2014;86:1244–52. 10.1038/ki.2014.120
    1. Torres VE, Harris PC. Strategies targeting cAMP signaling in the treatment of polycystic kidney disease. J Am Soc Nephrol. 2014;25:18–32. 10.1681/ASN.2013040398
    1. Belibi FA, Reif G, Wallace DP, Yamaguchi T, Olsen L, Li H, et al. Cyclic AMP promotes growth and secretion in human polycystic kidney epithelial cells. Kidney Int. 2004;66:964–73. 10.1111/j.1523-1755.2004.00843.x
    1. Grantham JJ, Mulamalla S, Swenson-Fields KI. Why kidneys fail in autosomal dominant polycystic kidney disease. Nat Rev Nephrol. 2011;7:556–66. 10.1038/nrneph.2011.109
    1. Pawlikowski M, Melen-Mucha G. Perspectives of new potential therapeutic applications of somatostatin analogs. Neuro Endocrinol Lett. 2003;24:21–7.
    1. Silva P, Schenermann M, Gard-Weiss T, Epstein FH. Somatostatin inhibits CNP-induced stimulation of shark rectal gland. Bull Mt Desert Isl Biol Lab Salisb Cove Maine. 1991;40:25–29.
    1. Reubi JC, Horisberger U, Studer UE, Waser B, Laissue JA. Human kidney as target for somatostatin: high affinity receptors in tubules and vasa recta. J Clin Endocrinol Metab. 1993;77:1323–8. 10.1210/jcem.77.5.7915721
    1. Lamberts SW, van der Lely AJ, de Herder WW, Hofland LJ. Octreotide. N Engl J Med. 1996;334:246–54. 10.1056/NEJM199601253340408
    1. Susini C, Buscail L. Rationale for the use of somatostatin analogs as antitumor agents. Ann Oncol. 2006;17:1733–42. 10.1093/annonc/mdl105
    1. Ruggenenti P, Remuzzi A, Ondei P, Fasolini G, Antiga L, Ene-Iordache B, et al. Safety and efficacy of long-acting somatostatin treatment in autosomal-dominant polycystic kidney disease. Kidney Int. 2005;68:206–16. 10.1111/j.1523-1755.2005.00395.x
    1. Caroli A, Antiga L, Cafaro M, Fasolini G, Remuzzi A, Remuzzi G, et al. Reducing polycystic liver volume in ADPKD: effects of somatostatin analogue octreotide. Clin J Am Soc Nephrol. 2010;5:783–9. 10.2215/CJN.05380709
    1. Masyuk TV, Masyuk AI, Torres VE, Harris PC, Larusso NF. Octreotide inhibits hepatic cystogenesis in a rodent model of polycystic liver disease by reducing cholangiocyte adenosine 3’,5’-cyclic monophosphate. Gastroenterol. 2007;132:1104–16. 10.1053/j.gastro.2006.12.039
    1. Caroli A, Perico N, Perna A, Antiga L, Brambilla P, Pisani 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;382:1485–95. 10.1016/S0140-6736(13)61407-5
    1. Ravine D, Gibson RN, Walker RG, Sheffield LJ, Kincaid-Smith P, Danks DM. Evaluation of ultrasonographic diagnostic criteria for autosomal dominant polycystic kidney disease 1. Lancet. 1994;343:824–7.
    1. Gaspari F, Perico N, Ruggenenti P, Mosconi L, Amuchastegui CS, Guerini E, et al. Plasma clearance of nonradioactive iohexol as a measure of glomerular filtration rate. J Am Soc Nephrol. 1995;6:257–63.
    1. Perico N, Antiga L, Caroli A, Ruggenenti P, Fasolini G, Cafaro M, et al. Sirolimus therapy to halt the progression of ADPKD. J Am Soc Nephrol. 2010;21:1031–40. 10.1681/ASN.2009121302
    1. Ruggenenti P, Gentile G, Perico N, Perna A, Barcella L, Trillini M, et al. Effect of sirolimus on disease progression in patients with autosomal dominant polycystic kidney disease and CKD stages 3b-4. Clin J Am Soc Nephrol. 2016;11:785–94. 10.2215/CJN.09900915
    1. Sharma K, Caroli A, Quach LV, Petzold K, Bozzetto M, Serra AL, et al. Kidney volume measurement methods for clinical studies on autosomal dominant polycystic kidney disease. PLoS ONE. 2017;12(5):e0178488 10.1371/journal.pone.0178488
    1. Chapman AB, Bost JE, Torres VE, Guay-Woodford L, Bae KT, Landsittel D, et al. Volume and functional outcomes in autosomal dominant polycystic kidney disease. Clin J Am Soc Nephrol. 2012;7:479–86. 10.2215/CJN.09500911
    1. Pisani A, Sabbatini M, Imbriaco M, Riccio E, Rubis N, Prinster A, et al. Long-term effects of octreotide on liver volume in patients with polycystic kidney and liver disease. Clin Gastroenterol Hepatol. 2016;14:1022–30.e4. 10.1016/j.cgh.2015.12.049
    1. Randomised placebo-controlled trial of effect of ramipril on decline in glomerular filtration rate and risk of terminal renal failure in proteinuric, non-diabetic nephropathy. The GISEN Group (Gruppo Italiano di Studi Epidemiologici in Nefrologia). Lancet. 1997;349:1857–63.
    1. Peduzzi P, Concato J, Feinstein AR, Holford TR. Importance of events per independent variable in proportional hazards regression analysis. II. Accuracy and precision of regression estimates. J Clin Epidemiol. 1995;48:1503–10.
    1. Zink RC, Koch GG. NParCov3: a SAS/IML macro for nonparametric randomization-based analysis of covariance. J Stat Softw. 2012;50:1–17. 10.18637/jss.v050.i03
    1. KDIGO 2012 clinical practice guideline for the evaluation and management of chronic kidney disease. Summary of recommendation statements. Kidney Int Suppl. 2013;3:5–14. 10.1038/kisup.2012.77
    1. Irazabal MV, Rangel LJ, Bergstralh EJ, Osborn SL, Harmon AJ, Sundsbak JL, et al. Imaging classification of autosomal dominant polycystic kidney disease: a simple model for selecting patients for clinical trials. J Am Soc Nephrol. 2015;26:160–72. 10.1681/ASN.2013101138
    1. Brouhard BH, LaGrone LF, Richards GE, Travis LB. Somatostatin limits rise in glomerular filtration rate after a protein meal. J Pediatr. 1987;110:729–34.
    1. Vora J, Owens DR, Luzio S, Atiea J, Ryder R, Hayes TM. Renal response to intravenous somatostatin in insulin-dependent diabetic patients and normal subjects. J Clin Endocrinol Metab. 1987;64:975–9. 10.1210/jcem-64-5-975
    1. Gines A, Salmeron JM, Gines P, Jimenez W, Salo J, Piera C, et al. Effects of somatostatin on renal function in cirrhosis. Gastroenterology. 1992;103:1868–74.
    1. Colao A, Petersenn S, Newell-Price J, Findling JW, Gu F, Maldonado M, et al. A 12-month phase 3 study of pasireotide in Cushing’s disease. N Engl J Med. 2012;366:914–24. 10.1056/NEJMoa1105743
    1. Ruggenenti P, Cravedi P, Remuzzi G. Mechanisms and treatment of CKD. J Am Soc Nephrol. 2012;23:1917–28. 10.1681/ASN.2012040390
    1. Jafar TH, Stark PC, Schmid CH, Landa M, Maschio G, Marcantoni C, et al. Proteinuria as a modifiable risk factor for the progression of non-diabetic renal disease. Kidney Int. 2001;60:1131–40. 10.1046/j.1523-1755.2001.0600031131.x
    1. Hebert LA, Greene T, Levey A, Falkenhain ME, Klahr S. High urine volume and low urine osmolality are risk factors for faster progression of renal disease. Am J Kidney Dis. 2003;41:962–71.
    1. Devuyst O, Chapman AB, Gansevoort RT, Higashihara E, Perrone RD, Torres VE, et al. Urine osmolality, response to tolvaptan, and outcome in autosomal dominant polycystic kidney disease: results from the TEMPO 3:4 trial. J Am Soc Nephrol. 2017;28:1592–602. 10.1681/ASN.2016040448
    1. Gabow PA, Kaehny WD, Johnson AM, Duley IT, Manco-Johnson M, Lezotte DC, et al. The clinical utility of renal concentrating capacity in polycystic kidney disease. Kidney Int. 1989;35:675–80.
    1. Juul KV, Bichet DG, Nielsen S, Norgaard JP. The physiological and pathophysiological functions of renal and extrarenal vasopressin V2 receptors. Am J Physiol Renal Physiol. 2014;306:F931–40. 10.1152/ajprenal.00604.2013
    1. Meijer E, Visser FW, van Aerts RMM, Blijdorp CJ, Casteleijn NF, D’Agnolo HMA, et al. Effect of lanreotide on kidney function in patients with autosomal dominant polycystic kidney disease: the DIPAK 1 randomized clinical trial. JAMA. 2018;320:2010–9. 10.1001/jama.2018.15870
    1. Lantinga MA, D’Agnolo HM, Casteleijn NF, de Fijter JW, Meijer E, Messchendorp AL, et al. Hepatic cyst infection during use of the somatostatin analog lanreotide in autosomal dominant polycystic kidney disease: an interim analysis of the randomized open-label multicenter DIPAK-1 study. Drug Saf. 2017;40:153–67. 10.1007/s40264-016-0486-x
    1. Hogan MC, Masyuk TV, Page LJ, Kubly VJ, Bergstralh EJ, Li X, et al. Randomized clinical trial of long-acting somatostatin for autosomal dominant polycystic kidney and liver disease. J Am Soc Nephrol. 2010;21:1052–61. 10.1681/ASN.2009121291
    1. McKeage K, Cheer S, Wagstaff AJ. Octreotide long-acting release (LAR): a review of its use in the management of acromegaly. Drugs. 2003;63:2473–99. 10.2165/00003495-200363220-00014
    1. Broder MS, Beenhouwer D, Strosberg JR, Neary MP, Cherepanov D. Gastrointestinal neuroendocrine tumors treated with high dose octreotide-LAR: a systematic literature review. World J Gastroenterol. 2015;21:1945–55. 10.3748/wjg.v21.i6.1945
    1. King BF, Reed JE, Bergstralh EJ, Sheedy PF 2nd, Torres VE. Quantification and longitudinal trends of kidney, renal cyst, and renal parenchyma volumes in autosomal dominant polycystic kidney disease. J Am Soc Nephrol. 2000;11:1505–11.
    1. Lambers Heerspink HJ, Perkovic V, de Zeeuw D. Is doubling of serum creatinine a valid clinical ‘hard’ endpoint in clinical nephrology trials? Nephron Clin Pract. 2011;119:c195–9. 10.1159/000327614
    1. Pocock SJ. Clinical trials: a practical approach. Chichester: John Wiley & Sons; 1983.
    1. Solazzo A, Testa F, Giovanella S, Busutti M, Furci L, Carrera P, et al. The prevalence of autosomal dominant polycystic kidney disease (ADPKD): a meta-analysis of European literature and prevalence evaluation in the Italian province of Modena suggest that ADPKD is a rare and underdiagnosed condition. PLoS ONE. 2018;13(1):e0190430 10.1371/journal.pone.0190430
    1. Spinelli L, Pisani A, Giugliano G, Trimarco B, Riccio E, Visciano B, et al. Left ventricular dysfunction in ADPKD and effects of octreotide-LAR: a cross-sectional and longitudinal substudy of the ALADIN trial. Int J Cardiol. 2019;275:145–51. 10.1016/j.ijcard.2018.10.063

Source: PubMed

3
Subscribe