Skip to main content
Clinical Journal of the American Society of Nephrology : CJASN logoLink to Clinical Journal of the American Society of Nephrology : CJASN
editorial
. 2014 Apr 10;9(5):831–836. doi: 10.2215/CJN.02480314

Novel Treatments of Autosomal Dominant Polycystic Kidney Disease

Rex L Mahnensmith 1,
PMCID: PMC4011435  PMID: 24721886

Autosomal dominant polycystic kidney disease (ADPKD) is considered the most common inherited form of kidney disease across all ethnic types (13). Present understanding holds that mutations of responsible genes, either PKD1 or PKD2, occur during embryogenesis and variably combine with “second hit” mutations within renal tubular epithelial cells that alter the intracellular level of polycystins, leading to epithelial cyst formations early in life (26). Cyst pathogenesis includes disordered nephron epithelial cell proliferations resulting in regional nephron bulging, accelerated apoptosis, loss of epithelial cell polarity, epithelial cell dedifferentiation, and eventual “pinching off” of the altered nephron regions into isolated cysts (3,6). Vasopressin-mediated fluid transport into cysts coupled with direct stimulation of cystic growths are considered central pathogenic growth variables (24).

Ultimately, multiple cysts slowly expand through adulthood, compressing functioning nephrons and blood vessels and causing progressive loss of renal function. Inflammation and fibrosis accompany renal deterioration. Approximately 50% of affected individuals experience advanced kidney failure by age 60 years (3). Whether a patient is afflicted with PKD1 or PKD2 mutations, heterogeneity of somatic mutations and individual variables, such as high BP, hemorrhagic events, and infections, alter disease progression (3). Thus, ADPKD presents a significant effect on health care systems worldwide.

Pharmacologic targets for intervention are based on known pathogenic variables. In an experimental model of polycystic kidney disease (PKD), genetic knockout of vasopressin production yielded rodents that remained relatively free of cysts until a vasopressin receptor agonist was administered exogenously, and then cysts appeared (4). In similar animal models of genetic PKD showing progressive cyst growth, vasopressin interruption using V2 antagonists slows cyst formations and enlargement (79). Even forced-water ingestion sufficient to naturally suppress secreted vasopressin can slow cyst formation and enlargement (10). These observations led to emphatic recommendations that patients with ADPKD aim to ingest >3 liters of noncaffeinated fluid per day, with the hope that natural vasopressin suppression would attenuate cyst growth (11,12). In addition to vasopressin-mediated fluid transport into cysts, other studies have demonstrated that the disordered nephron epithelial cell proliferation is related to activation of mammalian target of rapamycin (mTOR), a protein kinase that regulates polycystin pathways, cell proliferation, and protein synthesis (13,14). In experimental models of ADPKD, mTOR inhibition slows cyst enlargement (13,14). Additional evidence has established that cAMP mediates the progression of cystic disease in ADPKD by stimulating nephron cell proliferation and vasopressin-mediated fluid transport into cysts (1517) This provides rationale for therapies targeting cAMP, including somatostatin analogs, which can thwart renal and liver cyst growth in experimental models (1719).

On the basis of emerging understanding of complex cellular pathogenic pathways for ADPKD, hope has emerged regarding clinicians’ ability to modify the clinical course of ADPKD (3). For decades, ADPKD has been passively observed by the doctor. BP control has been deemed essential, as is avoidance of trauma, urinary stone formation, and infection, but little else could alter the biology of ADPKD or its clinical course. Routine cyst drainage or cyst decapitations proved of little help (20). Angiotensin inhibition has received emphasis as having unique benefit owing to known activation of the renin-angiotensin-aldosterone system in ADPKD (2125). One well done study demonstrated that angiotensin-converting enzyme inhibition prevented decreased creatinine clearance over a 5-year period in children and young adults with ADPKD (25). Additional proof of unique benefit resides with final analysis of the HALT Progression of Polycystic Kidney Disease trial (26).

In this context, four novel therapies with strong molecular rationales have entered into clinical trials as potentially modifying ADPKD: vasopressin-receptor inhibitors, mTOR inhibitors, somatostatin analogs, and statins. A significant factor propelling these trials is the now-accepted total kidney volume (TKV) imaging technology by magnetic resonance imaging (MRI), which was developed through the Consortium of Radiologic Imaging Studies in Polycystic Kidney Disease Group (CRISP) that identified total polycystic kidney volume (TKV), assessed best through MRI, as tightly associated with renal dysfunction and progression of ADPKD (27,28). CRISP also demonstrated that changes in TKV can be accurately detected over a period of months, making change in TKV an acceptable marker of disease progression in clinical trials, along with measured GFRs or serum creatinine change as principal meaningful end points.

Vasopressin V2 Receptor Antagonist Intervention

Vasopressin V2 receptor antagonists block vasopressin-mediated fluid transport into ADPKD cysts. The vasopressin V2 receptor antagonist, tolvaptan, has been evaluated in a phase 3, double-blind, placebo-controlled randomized trial over 3 years in 1445 patients with ADPKD aged 18–50 years with TKV>750 ml and estimated creatinine clearance>60 ml/min (29). The primary outcome was the annual rate of change in TKV, measured by volumetric MRI. Secondary end points included the rate of kidney function decline measured as reciprocal of serum creatinine and a composite of time to clinical progression defined as worsening kidney function, kidney pain, hypertension, and albuminuria. Over the 3-year period, the mean increase in TKV in the tolvaptan group was 2.8% per year compared with 5.5% per year in the placebo group (P<0.001). Analysis of secondary composite end points of time to clinical progression favored tolvaptan over placebo (44 versus 50 events per 100 follow-up years; P=0.01), with lower rates of worsening kidney function (2 versus 5 events per 100 person-years of follow-up; P<0.001) and less kidney pain (5 versus 7 events per 100 person-years of follow-up; P=0.01). Tolvaptan users demonstrated a slower decline in kidney function by reciprocal creatinine tracking (P<0.001). These are positive findings. However, tolvaptan use was associated with higher discontinuation (15.4% versus 5% with placebo) ascribed mostly to polyuria, nocturia, polydipsia, and liver toxicity, the latter prompting regulatory concern (30).

Tolvaptan remains under study in various trials in ADPKD patients for further evaluations of dose-related benefits versus adversity. On the basis of animal data and the human trial data to date, V2 antagonist intervention provides rational hope to ADPKD families and clinicians. The patient will experience a larger urine volume and must match this urine flow with sufficient daily fluid intake. Close monitoring of serum creatinine and liver functions is essential. The high monthly expense of tolvaptan may prove to be a negative factor from a cost-effectiveness perspective (31).

mTOR Pathway Inhibition

Six clinical trials evaluating mTOR inhibitors in adults with ADPKD have now been published. Five trials evaluated sirolimus (rapamycin), including the newest pilot trial by Braun et al. appearing in this issue of CJASN (3236). One trial evaluated everolimus (37). These trials are summarized in Table 1. Patient numbers per trial varied, with follow-up ranging from 6 to 24 months. The dose of mTOR inhibitor and baseline TKV and renal function also varied across the trials (Table 1). A small benefit or no benefit in TKV change is exhibited from mTOR inhibitor use (Table 1). In the four sirolimus trials, excluding the Braun trial (3235), the change in GFR did not reach a statistically significant difference between sirolimus versus control groups. Sirolimus associated with significant side effects in all trials, including stomatitis, acne, increased infections, increased proteinuria, and hyperlipidemia. In fact, dropout from the sirolimus group reached 29% in the Perico et al. trial (3235). Walz et al. evaluated everolimus over 24 months in a larger number of participants across a wide range of disease stages (37). The group receiving everolimus exhibited a slower increase in TKV compared with placebo, but exhibited a greater decline in eGFR over the same time frame. Everolimus exhibited higher rates of untoward side effects and dropout reached 25% of enrollees.

Table 1.

Trials evaluating mTOR inhibitors

Trial Patients Studied (n) Pharmacologic Treatment TKV (ml) GFR (ml/min)
Soliman et al., 2009 (32)a
 Treatment 8 Sirolimus 1 mg/d×6 mo 2845±443 → 3221±311 (P>0.05) 32±11 → 44±19 (P>0.05)
 Control 8 2667±399 → 3590±354 (P<0.05) 41±20 → 35±18 (P>0.05)
Perico et al., 2010 (33)b
 Treatment 21 Sirolimus 3 mg/d×6 mo dose adjusted to serum concentration 1914±1051 → 1960±1095 (P<0.05) 75±23 → 73±25 (P>0.05)
 Control Cross-over 1907±1107 → 1977±1133 (P=0.002) 73±22 → 74±24 (P≥0.05)
 Difference P=0.45 P>0.05
Serra et al., 2010 (34)c
 Treatment 50 Sirolimus 2 mg/d×18 mo 907 (507–1330) → 1007 (670–1593) (P>0.05) 92±20 → 92±21 (P>0.05)
 Control 50 1003 (574–1422) → 1123 (582–1731) (P>0.05) 91±17 → 87±20 (P>0.05)
 Difference P=0.26 P=0.07
Stallone et al., 2012 (35)d
 High-dose treatment 19 Sirolimus 3 mg/d×18 mo 1493±672 → 1508±674 (P=0.02) 61±17 → 66±24 (P>0.05)
 Low-dose treatment 18 Sirolimus 1 mg/d×18 mo 1712±634 → 1726±628 (P>0.05) 62±14 → 63±26 (P>0.05)
 Control 18 Doses adjusted to serum concentration 1869±668 → 1905±605 (P=0.003) C: 62±13 → 59±15 (P=0.01)
Walz et al., 2010 (37)e
 Treatment 213 Everolimus 2.5 mg bid×24 mo Mean TKV 230 (CI, 172 to 288) Mean eGFR 8.9 ml/min
 Cross-over 216 Mean TKV 301 (CI, 248 to 354) Mean eGFR 7.7 ml/min
 Control P=0.06 P=0.15
Braun et al., 2014 (36)
 Low-dose treatment 10 Sirolimus titrated to 2–5 ng/ml×12 mo 2479±1965 → 2115+1035 (P>0.05) 70±27 → 78+35 (P<0.05)
 High-dose treatment 10 Sirolimus titrated to 5–8 ng/ml×12 mo 1718±932 → 1537±864 (P>0.05) 72±25 → 74±34 (P>0.05)
 Control 10 Placebo 2072±1184 → 2059±1236 (P>0.05) 73±20 → 62±15 (P<0.05)

Data are presented as the mean±SD or median (interquartile range). CI, confidence interval; CKD-EPI, Chronic Kidney Disease Epidemiology Collaboration; MDRD, Modification of Diet in Renal Disease; mTOR, mammalian target of rapamycin; TKV, total kidney volume.

a

GFR by creatinine clearance.

b

GFR by iohexol clearance.

c

eGFR by CKD-EPI equation.

d

eGFR by MDRD equation.

e

eGFR by re-expressed MDRD equation.

Disappointment emerged from these published trials because the data did not produce evident benefit and adversity was substantial. In perspective, the trials were short, the drug dosing varied, and the stage of disease studied varied substantially. Rather than abandon mTOR therapy for ADPKD, Granthum et al. called for longer-duration, better structured trials (38).

It was in this context that Braun et al. proposed and executed a new pilot trial with 30 patients of low-dose versus standard-dose sirolimus with tracking of GFR by radiolabeled iothalamate clearance (being more precise) in addition to TKV (assessed by noncontrast computed tomography imaging) and blood levels of sirolimus (36). The investigators reasoned that radiolabeled iothalamate clearance measurements may reveal a treatment effect that might be missed by either traditional creatinine clearance or eGFR calculations from a serum creatinine measurement. They also desired precise dosing of sirolimus titrated to achieve target serum sirolimus levels. They observed that low-dose sirolimus use was associated with a significant increase in GFR measured by radiolabeled iothalamate, but concordant GFR estimated from the Chronic Kidney Disease Epidemiology Collaboration equation did not exhibit a significant increase. This discordance confirmed value in precise GFR tracking. There was no TKV change observed. Higher-dose sirolimus demonstrated no measured GFR or TKV change. These observations are meaningful in that they pave the way for the larger trial hoped for by Granthum et al. using radiolabeled iothalamate as the more precise GFR tracking tool (38). Shortcomings do exist in this pilot, and these are well addressed by the authors. Notwithstanding these, we should commend the effort and encourage the proposed larger trial that this pilot supports.

Somatostatin Analogs

Four published trials examine the possible benefit of regular injection of a somatostatin analog, which aims at inhibition of cAMP production in renal cells and resultant cyst development and growth (3942). Essential data are summarized in Table 2. Three small studies focused on polycystic liver disease and assessed effect on TKV as secondary end points; their data suggested marginal benefit on TKV (3941). A fourth randomized controlled trial evaluated 70 ADPKD patients using octreotide over 3 years (42). In this study, TKV increased less in the octreotide group compared with placebo; this difference, although persistent in direction, was no longer statistically significant at 3 years. Decline in GFR, measured by radiolabeled iothalamate clearance, was slower in the octreotide group compared with placebo, but this difference also was not statistically significant at 3 years. Octreotide associated with diarrhea and cholecystitis but showed little adversity otherwise. The researchers expressed optimism, stating that “because of their very good risk–benefit profile, even in life-long therapy, somatostatin analogs are so far the only viable option for long-term treatment of this disorder” (42). Shortcomings included insufficient number of years of follow-up and a relatively small number of participants. Together, these four trials do not compel routine clinical use of a somatostatin analog in ADPKD, but they do provide hope and sufficient rationale for larger, longer randomized trials.

Table 2.

Trials evaluating somatostatin analogs

Trial Patients Studied (n) Pharmacologic Treatment TKV (ml) GFR (ml/min)
Ruggenenti et al., 2005 (39)a
 Treatment 12 Octreotide LAR 40 mg every mo×6 mo 2551±1053 → 2622±1111 (P<0.01) 56±29 → 53±29 (P>0.05)
 Control Cross-over 2461±959 → 2623±1021 (P<0.05) 55±22 → 52±25 (P>0.05)
Hogan et al., 2010 (40)a
 Treatment 28 Octreotide LAR 40 mg every mo×12 mo 1143±827 → 1128±796 (P>0.05) 68±26 → 64±25 (P>0.05)
 Control 14 803±269 → 873±306 (P<0.05) 70±28 → 66±26 (P>0.05)
 Difference P<0.05 P=0.98
Van Keimpema et al., 2009 (41)
 Treatment 27 Lantreotide 120 mg every mo×24 wk Mean TKV −17 (CI, −126 to 93) 83 (8–158) → 80 (8–153) (P>0.05)
 Control 26 Mean TKV 50 (CI −99 to 199) 91 (9–173) → 96 (−12 to 205) (P>0.05)b
 Difference P=0.02
Caroli et al., 2013 (42)a,c
 Treatment 35 Octreotide LAR 40 mg every mo×36 mo Mean ∆TKV 220±49 88±4 → 76±4; annual GFR slope over 0–3 years, −3.85 (−6.2 to −1.92)
 Control 35 Mean ∆TKV 453±80 C: 77±5 → 68±6a; annual GFR; slope over 0–3 years, −4.95 (−7.5 to −1.97)
 Difference P=0.25 P=0.13

Data are presented as the mean±SD or median (interquartile range). Octreotide LAR, octreotide acetate for injectable suspension; CI, confidence interval.

a

GFR by iothalamate clearance.

b

Reported as creatinine μmol/L.

c

Slope of GFR over 3 years between treatment and placebo.

Statins

This issue of CJASN also includes the first published randomized placebo-controlled clinical trial of deliberate statin use to evaluate potential benefit on height-corrected TKV change of ADPKD in children and young adults (43). This trial evaluated either 20 or 40 mg pravastatin combined with an angiotensin-converting enzyme inhibitor in young patients aged 8–22 years with ADPKD. The trial enrolled 110 patients, and 91 patients completed the trial. Those patients randomized to pravastatin demonstrated significantly less increase in height-adjusted TKV over 3 years than placebo-receiving patients. There was no significant change in creatinine clearance or LDL cholesterol or proteinuria between groups. Pravastatin was well tolerated with no apparent adversity. The mechanisms of statin benefit in ADPKD are unclear. This study was well conceived, well executed, and well analyzed. Conclusions of benefit are justified. This study provides a firm foundation for careful and vigilant statin use in ADPKD, particularly if introduced early in the course of ADPKD evolution.

We are on the threshold of effective interventions for ADPKD. The novel therapies under study are reasonable and rational. Some studies were pilots. Duration varied and was largely insufficient in most trials. Stage of disease varied substantially. Power has been small. Genotype of ADPKD was not a required assessment, which may be a variable for natural progression. Thus, except for careful prescription of statins, these trials cannot compel routine use of one of these interventions outside of additional clinical trials. In my opinion, new trials should be conducted over 3–5 years and should have greater power, include biomarkers, and enroll participants of the same PKD genotype and stage of disease.

Disclosures

R.L.M. has served as a coinvestigator in the Otsuka-sponsored TEMPO trial. He does not receive and has not received compensation.

Footnotes

Published online ahead of print. Publication date available at www.cjasn.org.

See related articles, “Low-Dose Rapamycin (Sirolimus) Effects in Autosomal Dominant Polycystic Kidney Disease: An Open-Label Randomized Controlled Pilot Study,” and “Effect of Pravastatin on Total Kidney Volume, Left Ventricular Mass Index, and Microalbuminuria in Pediatric Autosomal Dominant Polycystic Kidney Disease,” on pages 881–888 and pages 889–896 respectively.

References

  • 1.Torres VE, Harris PC, Pirson Y: Autosomal dominant polycystic kidney disease. Lancet 369: 1287–1301, 2007 [DOI] [PubMed] [Google Scholar]
  • 2.Grantham JJ: Clinical practice. Autosomal dominant polycystic kidney disease. N Engl J Med 359: 1477–1485, 2008 [DOI] [PubMed] [Google Scholar]
  • 3.Torres VE: Treatment strategies and clinical trial design in ADPKD. Adv Chronic Kidney Dis 17: 190–204, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Wang X, Wu Y, Ward CJ, Harris PC, Torres VE: Vasopressin directly regulates cyst growth in polycystic kidney disease. J Am Soc Nephrol 19: 102–108, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Pei Y: Molecular genetics of autosomal dominant polycystic kidney disease. Clin Invest Med 26: 252–258, 2003 [PubMed] [Google Scholar]
  • 6.Higashihara E, Horie S, Muto S, Mochizuki T, Nishio S, Nutahara K: Renal disease progression in autosomal dominant polycystic kidney disease. Clin Exp Nephrol 16: 622–628, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Gattone VH, 2nd, Wang X, Harris PC, Torres VE: Inhibition of renal cystic disease development and progression by a vasopressin V2 receptor antagonist. Nat Med 9: 1323–1326, 2003 [DOI] [PubMed] [Google Scholar]
  • 8.Torres VE, Wang X, Qian Q, Somlo S, Harris PC, Gattone VH, 2nd: Effective treatment of an orthologous model of autosomal dominant polycystic kidney disease. Nat Med 10: 363–364, 2004 [DOI] [PubMed] [Google Scholar]
  • 9.Wang X, Gattone VH, 2nd, Harris PC, Torres VE: Effectiveness of vasopressin V2 receptor antagonists OPC-31260 and OPC-41061 on polycystic kidney disease development in the PCK rat. J Am Soc Nephrol 16: 846–851, 2005 [DOI] [PubMed] [Google Scholar]
  • 10.Nagao S, Nishii K, Katsuyama M, Kurahashi H, Marunouchi T, Takahashi H, Wallace DP: Increased water intake decreases progression of polycystic kidney disease in the PCK rat. J Am Soc Nephrol 17: 2220–2227, 2006 [DOI] [PubMed] [Google Scholar]
  • 11.Torres VE, Bankir L, Grantham JJ: A case for water in the treatment of polycystic kidney disease. Clin J Am Soc Nephrol 4: 1140–1150, 2009 [DOI] [PubMed] [Google Scholar]
  • 12.Grantham JJ: Therapy for polycystic kidney disease? It’s water, stupid! J Am Soc Nephrol 19: 1–7, 2008 [DOI] [PubMed] [Google Scholar]
  • 13.Wahl PR, Serra AL, Le Hir M, Molle KD, Hall MN, Wüthrich RP: Inhibition of mTOR with sirolimus slows disease progression in Han:SPRD rats with autosomal dominant polycystic kidney disease (ADPKD). Nephrol Dial Transplant 21: 598–604, 2006 [DOI] [PubMed] [Google Scholar]
  • 14.Shillingford JM, Murcia NS, Larson CH, Low SH, Hedgepeth R, Brown N, Flask CA, Novick AC, Goldfarb DA, Kramer-Zucker A, Walz G, Piontek KB, Germino GG, Weimbs T: The mTOR pathway is regulated by polycystin-1, and its inhibition reverses renal cystogenesis in polycystic kidney disease. Proc Natl Acad Sci U S A 103: 5466–5471, 2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Wang X, Ward CJ, Harris PC, Torres VE: Cyclic nucleotide signaling in polycystic kidney disease. Kidney Int 77: 129–140, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Devuyst O, Torres VE: Osmoregulation, vasopressin, and cAMP signaling in autosomal dominant polycystic kidney disease. Curr Opin Nephrol Hypertens 22: 459–470, 2013 [DOI] [PubMed] [Google Scholar]
  • 17.Torres VE, Harris PC: Strategies targeting cAMP signaling in the treatment of polycystic kidney disease. J Am Soc Nephrol 25: 18–32, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.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. Gastroenterology 132: 1104–1116, 2007 [DOI] [PubMed] [Google Scholar]
  • 19.Winkler SN, Torikai S, Levine BS, Kurokawa K: Effect of somatostatin on vasopressin-induced antidiuresis and renal cyclic AMP of rats. Miner Electrolyte Metab 7: 8–14, 1982 [PubMed] [Google Scholar]
  • 20.Elzinga LW, Barry JM, Torres VE, Zincke H, Wahner HW, Swan S, Bennett WM: Cyst decompression surgery for autosomal dominant polycystic kidney disease. J Am Soc Nephrol 2: 1219–1226, 1992 [DOI] [PubMed] [Google Scholar]
  • 21.Chapman AB, Johnson A, Gabow PA, Schrier RW: The renin-angiotensin-aldosterone system and autosomal dominant polycystic kidney disease. N Engl J Med 323: 1091–1096, 1990 [DOI] [PubMed] [Google Scholar]
  • 22.Tkachenko O, Helal I, Shchekochikhin D, Schrier RW: Renin-angiotensin-aldosterone system in autosomal dominant polycystic kidney disease. Curr Hypertens Rep 9: 12–20, 2013 [DOI] [PubMed] [Google Scholar]
  • 23.Chapman AB, Stepniakowski K, Rahbari-Oskoui F: Hypertension in autosomal dominant polycystic kidney disease. Adv Chronic Kidney Dis 17: 153–163, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Schrier RW: Renal volume, renin-angiotensin-aldosterone system, hypertension, and left ventricular hypertrophy in patients with autosomal dominant polycystic kidney disease. J Am Soc Nephrol 20: 1888–1893, 2009 [DOI] [PubMed] [Google Scholar]
  • 25.Cadnapaphornchai MA, McFann K, Strain JD, Masoumi A, Schrier RW: Prospective change in renal volume and function in children with ADPKD. Clin J Am Soc Nephrol 4: 820–829, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Chapman AB, Torres VE, Perrone RD, Steinman TI, Bae KT, Miller JP, Miskulin DC, Rahbari Oskoui F, Masoumi A, Hogan MC, Winklhofer FT, Braun W, Thompson PA, Meyers CM, Kelleher C, Schrier RW: The HALT polycystic kidney disease trials: Design and implementation. Clin J Am Soc Nephrol 5: 102–109, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Chapman AB, Guay-Woodford LM, Grantham JJ, Torres VE, Bae KT, Baumgarten DA, Kenney PJ, King BF, Jr, Glockner JF, Wetzel LH, Brummer ME, O’Neill WC, Robbin ML, Bennett WM, Klahr S, Hirschman GH, Kimmel PL, Thompson PA, Miller JP; Consortium for Radiologic Imaging Studies of Polycystic Kidney Disease cohort: Renal structure in early autosomal-dominant polycystic kidney disease (ADPKD): The Consortium for Radiologic Imaging Studies of Polycystic Kidney Disease (CRISP) cohort. Kidney Int 64: 1035–1045, 2003 [DOI] [PubMed] [Google Scholar]
  • 28.Grantham JJ, Chapman AB, Torres VE: Volume progression in autosomal dominant polycystic kidney disease: The major factor determining clinical outcomes. Clin J Am Soc Nephrol 1: 148–157, 2006 [DOI] [PubMed] [Google Scholar]
  • 29.Torres VE, Chapman AB, Devuyst O, Gansevoort RT, Grantham JJ, Higashihara E, Perrone RD, Krasa HB, Ouyang J, Czerwiec FS; TEMPO 3:4 Trial Investigators: Tolvaptan in patients with autosomal dominant polycystic kidney disease. N Engl J Med 367: 2407–2418, 2012. 23121377 [Google Scholar]
  • 30.US Food and Drug Administration: Samsca (tolvaptan): Drug warning—potential risk of liver injury, 2013. Available at: http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlerts forHumanMedicalProducts/ ucm336669.htm. Accessed March 4, 2014
  • 31.Erickson KF, Chertow GM, Goldhaber-Fiebert JD: Cost-effectiveness of tolvaptan in autosomal dominant polycystic kidney disease. Ann Intern Med 159: 382–389, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Soliman AR, Ismail E, Zamil S, Lotfy A: Sirolimus therapy for patients with adult polycystic kidney disease: A pilot study. Transplant Proc 41: 3639–3641, 2009 [DOI] [PubMed] [Google Scholar]
  • 33.Perico N, Antiga L, Caroli A, Ruggenenti P, Fasolini G, Cafaro M, Ondei P, Rubis N, Diadei O, Gherardi G, Prandini S, Panozo A, Bravo RF, Carminati S, De Leon FR, Gaspari F, Cortinovis M, Motterlini N, Ene-Iordache B, Remuzzi A, Remuzzi G: Sirolimus therapy to halt the progression of ADPKD. J Am Soc Nephrol 21: 1031–1040, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Serra AL, Poster D, Kistler AD, Krauer F, Raina S, Young J, Rentsch KM, Spanaus KS, Senn O, Kristanto P, Scheffel H, Weishaupt D, Wüthrich RP: Sirolimus and kidney growth in autosomal dominant polycystic kidney disease. N Engl J Med 363: 820–829, 2010 [DOI] [PubMed] [Google Scholar]
  • 35.Stallone G, Infante B, Grandaliano G, Bristogiannis C, Macarini L, Mezzopane D, Bruno F, Montemurno E, Schirinzi A, Sabbatini M, Pisani A, Tataranni T, Schena FP, Gesualdo L: Rapamycin for treatment of type I autosomal dominant polycystic kidney disease (RAPYD-study): A randomized, controlled study. Nephrol Dial Transplant 27: 3560–3567, 2012 [DOI] [PubMed] [Google Scholar]
  • 36.Braun WE, Schold JD, Stephany BR, Spirko RA, Herts BR: Low dose rapamycin (sirolimus) effects in autosomal dominant polycystic kidney disease: An open-label randomized control pilot study. Clin J Am Soc Nephrol 9: 881–888, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Walz G, Budde K, Mannaa M, Nürnberger J, Wanner C, Sommerer C, Kunzendorf U, Banas B, Hörl WH, Obermüller N, Arns W, Pavenstädt H, Gaedeke J, Büchert M, May C, Gschaidmeier H, Kramer S, Eckardt KU: Everolimus in patients with autosomal dominant polycystic kidney disease. N Engl J Med 363: 830–840, 2010 [DOI] [PubMed] [Google Scholar]
  • 38.Granthum JJ, Bennett WM, Perrone RD: mTOR inhibitors and autosomal dominant polycystic kidney disease. N Engl J Med 364: 286–287, 2011 [DOI] [PubMed] [Google Scholar]
  • 39.Ruggenenti P, Remuzzi A, Ondei P, Fasolini G, Antiga L, Ene-Iordache B, Remuzzi G, Epstein FH: Safety and efficacy of long-acting somatostatin treatment in autosomal-dominant polycystic kidney disease. Kidney Int 68: 206–216, 2005 [DOI] [PubMed] [Google Scholar]
  • 40.Hogan MC, Masyuk TV, Page LJ, Kubly VJ, Bergstralh EJ, Li X, Kim B, King BF, Glockner J, Holmes DR, 3rd, Rossetti S, Harris PC, LaRusso NF, Torres VE: Randomized clinical trial of long-acting somatostatin for autosomal dominant polycystic kidney and liver disease. J Am Soc Nephrol 21: 1052–1061, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.van Keimpema L, Nevens F, Vanslembrouck R, van Oijen MG, Hoffmann AL, Dekker HM, de Man RA, Drenth JP: Lanreotide reduces the volume of polycystic liver: A randomized, double-blind, placebo-controlled trial. Gastroenterology 137: 1661–1668, e1–e2, 2009 [DOI] [PubMed] [Google Scholar]
  • 42.Caroli A, Perico N, Perna A, Antiga L, Brambilla P, Pisani A, Visciano B, Imbriaco M, Messa P, Cerutti R, Dugo M, Cancian L, Buongiorno E, De Pascalis A, Gaspari F, Carrara F, Rubis N, Prandini S, Remuzzi A, Remuzzi G, Ruggenenti P; ALADIN study group: Effect of longacting somatostatin analogue on kidney and cyst growth in autosomal dominant polycystic kidney disease (ALADIN): A randomised, placebo-controlled, multicentre trial. Lancet 382: 1485–1495, 2013 [DOI] [PubMed] [Google Scholar]
  • 43.Cadnapaphornchai M, George D, Wang W, Reed-Gitomer B, McFann K, Strain J, Schrier R: Effect of pravastatin on total kidney volume, left ventricular mass index, and microalbuminuria in pediatric autosomal dominant polycystic kidney disease. Clin J Am Soc Nephrol 9: 889–896, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Clinical Journal of the American Society of Nephrology : CJASN are provided here courtesy of American Society of Nephrology

RESOURCES