• No results found

University of Groningen ADPKD Messchendorp, Annemarie Lianne

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen ADPKD Messchendorp, Annemarie Lianne"

Copied!
295
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

ADPKD

Messchendorp, Annemarie Lianne

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from

it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2019

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Messchendorp, A. L. (2019). ADPKD: Risk Prediction for Treatment Selection. Rijksuniversiteit Groningen.

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Risk Prediction for Treatment Selection

(3)

978-94-6375-283-1 (digital version)

Financial support by the University Medical Center Groningen, Research institute GUIDE, University of Groningen for the publication of this thesis is gratefully acknowledged. Financial support for the printing of this thesis was also kindly provided by:

Cover design: Marije Esselink, Studio Neon

Layout and design: Elisa Calamita, persoonlijkproefschrift.nl Printing: Ridderprint BV | www.ridderprint.nl

© A.L. Messchendorp 2019

All rights reserved. No part of this thesis may be reproduced, stored or transmitted in any way or by any means without the prior permission of the author.

(4)

Risk Prediction for Treatment Selection

Proefschrift

ter verkrijging van de graad van doctor aan de Rijksuniversiteit Groningen

op gezag van de

rector magnificus prof. dr. E. Sterken en volgens besluit van het College voor Promoties.

De openbare verdediging zal plaatsvinden op maandag 25 februari 2019 om 14.30 uur

door

Annemarie Lianne Messchendorp geboren op 12 oktober 1988

(5)

Prof. dr. C.A.J.M. Gaillard Beoordelingscommissie Prof. dr. H.J. Lambers Heerspink Prof. dr. J.W. de Fijter

Prof. dr. R. Torra

Paranimfen dr. L.M. Kieneker H.L. Waldus

(6)

1. Introduction 7 2. Estimation of total kidney volume in autosomal dominant polycystic

kidney disease

21 3. T1 vs. T2 weighted magnetic resonance imaging to assess total kidney

volume in patients with autosomal dominant polycystic kidney disease

47 4. Urinary biomarkers to identify ADPKD patients with a high likelihood

of disease progression

69 5. Urinary biomarkers to select patients with rapidly progressive

autosomal dominant polycystic kidney disease

99 6. Kidney Function Reserve Capacity in early and later stage autosomal

dominant polycystic kidney disease

127 7. Association of plasma somatostatin with disease severity and

progression in patients with autosomal dominant polycystic kidney disease

161

8. Somatostatin in renal physiology and the place of somatostatin analogues in autosomal dominant polycystic kidney disease

183 9. Effect of a somatostatin analogue on the vasopressin pathway in

patients with autosomal dominant polycystic kidney disease

207 10. Recommendations for the use of tolvaptan in ADPKD: a proposal to

update the ERA-EDTA decision algorithm

225

11. General discussion and future perspectives 247

Nederlandse samenvatting 265

Dankwoord 277

(7)
(8)

1

Introduction

(9)

Autosomal Dominant Polycystic Kidney Disease (ADPKD) is the most common form of inherited kidney disease. The disease has a prevalence of 3 to 4 per 10,000 in the general population1, 2 and is characterized by bilateral progressive renal cyst formation and growth throughout life.

Pathophysiology of ADPKD

ADPKD is caused by a mutation in the PKD1 gene (85% of cases) or in the PKD2 gene (15% of cases). These genes encode for the proteins Polycystin-1 and Polycystin-2 respectively3. These proteins form the so-called polycystin complex that is localized at the basis of the primary cilium of renal epithelial cells, which acts as a mechanosensor that detects flow in the renal tubules. When this sensor is stimulated, calcium influx occurs from pre-urine into the cytoplasm of renal tubular epithelial cells and from intracellular stores. High intracellular calcium inhibits the enzyme adenylyl cyclase (AC), which is localized at the basolateral side of renal tubular epithelial cells, and that stimulates the conversion of adenosine triphosphate (ATP) into cyclic adenosinemonophosphate (cAMP). In normal physiology, cAMP is involved via a cascade of down-stream intracellular processes with regulation of cell growth and chloride driven fluid transport. In ADPKD, the polycystin complex is dysfunctional because of mutations in PKD1 or PKD2 and consequently, calcium cannot enter the cells, nor can calcium be released from intracellular stores. Low intracellular calcium leads to high activity of AC and high intracellular cAMP levels. In turn, this results in high intracellular cAMP levels, which lead to aberrant renal tubular epithelial cell proliferation and chloride driven fluid excretion in the kidney4. These are the two key components of the process of cyst formation and growth in ADPKD (Figure 1). Variable disease course in ADPKD

Eventually, ADPKD can lead to development of end stage kidney disease (ESKD). However, only 70% of patients reach ESKD and the age at which patient reach ESKD shows large interindividual variability. This is partly explained by the locus and type of mutation, since patients with a PKD1 mutation, especially truncating mutations, generally progress faster to ESKD compared to patients with a PKD2 mutation5. Figure 2 represents this variable disease course in the families of our Groningen ADPKD patient cohort per PKD mutation. This figure shows that even between family members that share the same mutation, a large interindividual variability exists in the age at which family members with ADPKD reach ESKD.

(10)

Figure 1. Schematic representation of the pathophysiological progresses that drive cyst for-mation and growth in renal tubular epithelial cells of the collecting duct in ADPKD and the mechanism of action of vasopressin V2 receptor antagonists and somatostatin analogues (modified from Zittema et al.). In ADPKD the polycystin complex (formed by the proteins PC1 and

PC2 on the apical membrane) is dysfunctional which leads to diminished calcium influx or diminished release of calcium from intracellular stores. Low intracellular calcium levels in turn stimulate acti-vation of adenylate cyclase (AC), which converts adenosine triphosphate (ATP) into cyclic adenosine monophosphate (cAMP). cAMP is an important player in several pathways that could possibly lead to cell proliferation and cell growth, for instance, via activation of the Ras/B-Raf/MEK/ERK-pathway. Furthermore, cAMP activates apical positioned chloride channels (CFTR-channels) leading to fluid secretion, which together results in cyst formation. cAMP production can be inhibited by blocking the vasopressin V2 receptor (V2R), which is coupled to G stimulatory (Gs) proteins that can activate AC. Activation of the somatostatin receptor (SSTR) can inhibit cAMP production in a direct and indirect way. AC can directly be inhibited by the receptor coupled G inhibitory (Gi) proteins. Activation of these Gi proteins can also activate calcium channels and stimulate intracellular release of calcium via phospholipase C (PLC ) which can restore intracellular calcium stores. This leads indirectly to inhibition of cAMP production. Orange and grey lines indicate that the pathway is either activated or inactivated. Abbreviations are: ATP, adenosine triphosphate; cAMP, cyclic

adenosinemonophos-phate; CFTR, cystic fibrosis transmembrane conductance regulator; ER, endoplasmic reticulum; Gi, G inhitibory; Gs, G stimulatory; mTOR, mammalian target of rapamycin; PC, polycystin; PKA, protein kinase A; PLC, phospholipase C; SSTR, somatostatin receptor; TSC, tuberous sclerosis; V2R, vasopressin V2 receptor

Remarkably, this figure does not confirm differences between PKD1 truncating and non-truncating mutations, which may be explained by referral bias, since the UMCG is a tertiary referral center, and especially subjects with rapidly progressive disease are referred for consultation of inclusion in trials. However, this figure does give the impression that patients with a PKD2 mutation develop ESKD at an older age than subjects with a PKD1 mutation.

(11)

Figure 2. Age at last follow-up of patients with ADPKD per family and PKD mutation. Each dot/

square/triangle within a vertical line represents an affected member from the same family. A solid marker indicates the age at which a family member developed ESKD and open markers indicate the age at last follow-up of a family member without ESKD. The horizontal line indicates the mean age at ESKD per mutation type. Data from this figure is collected from family tree questionnaires which were taken from 68 ADPKD patients at the outpatient clinic from the UMCG.

Factors associated with disease progression

Given the variability in age at which ESKD is reached within families, other factors than type of gene mutation are likely involved in determining the rate of disease progression. Intrafamilial variability may, for instance, be caused by other genes that can modify disease progression6. Furthermore, several studies have reported that males in general show a faster rate of disease progression compared to females, suggesting that sex hormones may be of importance7, 8. Finally, environmental factors that may cause a more rapid rate of disease progression are a low birth weight, high caffeine intake, smoking, low water intake and high protein intake9-14.

Currently used predictors of disease progression

Since the disease course of ADPKD is highly variable, it is difficult to predict the rate of disease progression in an individual patient. Obviously, the ability to predict the rate of

(12)

disease progression in patients with ADPKD would help patients and caregivers alike in treatment related decisions. Patients with a higher rate of disease progression will probably benefit the most from therapy, since in these patients the benefit to risk ratio of treatment is expected to be better, especially when treatment is started early15. Currently, there are several variables used to predict the rate of disease progression in ADPKD. ADPKD is in general a slowly progressive kidney disease. As ESKD is the endpoint to be prevented, it makes sense to use glomerular fitration rate (GFR), indexed for age, as a predictor for the rate of disease progression. However, GFR indexed for age may be less sensitive in early stages of this disease, as kidney function remains relatively stable in the near-normal range for prolonged periods of time before it starts to decline. It is hypothesized that in early stage ADPKD, GFR remains in the normal range due to compensatory hyperfiltration of remnant nephrons, while cysts are progressively formed and nephrons are lost16 (Figure 3). Therefore, much attention has focused on total kidney volume (TKV) as a predictor of disease progression, because an increase in TKV starts already at a very young age. The CRISP (Consortium for Radiologic Imaging Studies of Polycystic Kidney Disease) study was one of the largest and most important studies to confirm that TKV can indeed be used as a surrogate for disease progression in ADPKD. Thereafter several other studies have corroborated this finding17.

As mentioned above, the rate of disease progression is also partly explained by ADPKD genotype, and genotype is therefore also commonly used to predict disease progression5. However, genotype and TKV are often not available in routine clinical care, because their assessment is laborious and expensive. Furthermore, at an individual patient level their predictive power for the rate of disease progression is limited. Therefore cheap and easy to measure risk markers need to be developed that alone, or in combination with conventional risk markers, can predict the rate of disease progression in ADPKD.

(13)

Figure 3. Representation of the natural disease course of ADPKD.

Promising new risk markers

Urinary damage and inflammation markers

Measurement of urinary damage and inflammation markers are of interest, especially because these markers are relatively inexpensive and easy to measure. Many of these markers have been shown to be associated with disease severity and disease progression in non-ADPKD chronic kidney disease18-30 and some are even approved by the FDA as an official biomarker for kidney damage31. In ADPKD, cross-sectional studies already showed that urinary damage markers are associated with disease severity, assessed as GFR and TKV32, 33. However, little attention has been focused on the possible ability of these markers to predict disease progression in ADPKD34-37. In Table 1 various urinary damage and inflammation markers are shown. This table displays the performance of the assays, and whether previous studies have shown associations of these markers cross-sectionally with disease severity and longitudinally with disease progression in patients with ADPKD.

(14)

Table 1. Urinary inflammation and tubular damage markers representing different segments of the nephron and their association with disease severity and disease progression in patients with ADPKD.

Assay

performance32, 38-40 Cross-sectional32 Longitudinal34-37

Intra-CV

(%) Inter-CV(%) Association with GFR Association with TKV

Association with change in GFR Association with change in TKV General - Albumin 2.2 2.6 + + + NA Glomerular - IgG 8.4 16.4 + + + NA Proximal tubular - β2MG 6.3 8.0 + - - NA - KIM-1 7.4 14.5 - + + NA - NAG 3.1 13.7 + + - NA Distal tubular - H-FABP 9.3 17.6 + - - NA Inflammatory - MCP-1 8.3 12.7 + + + NA - MIF 5.2 9.2 - - - NA - NGAL 6.8 19.6 + - -

-Abbreviations are: IgG, immunoglobulin G; β2MG, β2-microglobulin; KIM-1, kidney injury

molecule 1; NAG, N-actyl-β-D-glucosaminidase; HFABP, heart-type fatty acid binding protein; MCP-1, monocyte chemotactic protein 1; MIF, macrophage migration inhibitory factor; NGAL, neutrophil gelatinase-associated lipocalin; GFR, glomerular filtration rate; TKV, total kidney volume; NA, not applicable.

Hyperfiltration: the earliest marker of disease severity and possible future disease progression?

As it is hypothesized that kidney function stays stable in the early stages of the disease by compensatory hyperfiltration of remnant nephrons16, the extent to which a patient is hyperfiltrating may be an early marker of disease severity and predictor of future disease progression. Unfortunately, renal hyperfiltration cannot be directly measured in humans, and therefore surrogate measures are used. Hyperfiltration is sometimes defined as an unstimulated increased kidney function, but this does not seem applicable for patients with ADPKD since it is speculated that patients hyperfilter in ADPKD to compensate for GFR loss. In these cases, glomerular hyperfiltration is sometimes defined as an increased filtration fraction, determined by GFR divided by the effective kidney plasma flow41. Indeed increased filtration fraction indicates that

(15)

the kidney is attempting to maintain kidney function to a certain level despite lower effective kidney plasma flow. However, measurement of effective kidney plasma flow by infusion of exogenous tracers such as para-aminohippuric acid (PAH) may be inaccurate, and lead to overestimation of filtration fraction, especially when tubular function is compromised as in ADPKD42. Glomerular hyperfiltration is therefore more commonly defined as the loss of kidney function reserve capacity, i.e. the impairment of the kidney to increase GFR in response to stimuli such as dopamine or amino acids43, 44. If patients with ADPKD hyperfilter in early stages of the disease, a loss of kidney function reserve capacity is expected to occur prior to a decline in GFR.

Endogenous substances involved in ADPKD disease progression

One of the pivotal detrimental factors in the pathophysiology of ADPKD are elevated levels of cAMP in renal tubular cells. These levels will increase further by stimulation of the vasopressin V2 receptor by vasopressin (Figure 1). Thus, high levels of vasopressin are expected to be associatied with a worse disease course and might therefore serve as a risk marker for future rapid disease progression4. Indeed, studies have shown that copeptin, a surrogate marker for vasopressin, is associated with a faster rate of kidney function decline in patients with ADPKD45 and blocking the vasopressin V2 receptor, attenuates ADPKD disease progression46, 47. Another hormone that may interfere in this pathway is somatostatin. Somatostatin is a hormone that is involved in many cell processes, and that can induce a broad spectrum of biological effects. Importantly, somatostatin has the ability to directly and indirectly inhibit tubular cAMP production48-50 (Figure 1). Therefore, it may be that patients with low levels of endogenous somatostatin have a worse disease course compared to patients with high levels. In line, the administration of somatostatin analogues are of interest as a possible therapeutic option in ADPKD.

Somatostatin analogues in ADPKD

Somatostatin analogues indeed have te ability to inhibit cAMP production by the inhibition of AC activity, which resembles the working mechanism of tolvaptan. Nine distinct membrane-bound AC isoforms (AC1-9) and one soluble AC (sAC) have been identified. Except for AC8, all of these isoforms are expressed in the kidney. Specific AC isoforms can exert unique effects in various cell types of the kidney, potentially relevant for channel activation and thus cystogenesis51. It is currently unknown if there are specific AC isoforms associated with the vasopressin V2 or somatostatin receptor. It may therefore well be that both receptors interact with the same AC isoform and that there is a pharmacological interaction between somatostatin analogues and

(16)

tolvaptan. Interestingly, some studies have suggested involvement of somatostatin in renal water handling52-55, which suggests that there indeed may be an interaction between the somatostatin and vasopressin pathways.

AIMS OF THE THESIS

The general aim of this thesis is to study if current risk markers that predict disease progression in ADPKD can be improved, and to search for new markers that may predict disease progression beyond the currently used risk markers.

OUTLINE OF THE THESIS

The assessment of TKV by the gold standard manual tracing method is very laborious and therefore not generally applicable in routine clinical care. In chapter 2 we investigated if TKV could be assessed by easy and less laborious estimation methods instead. Historically, gadolinium enhanced T1 weighted images were used for the measurement of TKV because of the short scanning time, low variations in image quality and high contrast of the renal structures against the surrounding tissues56. Gadolinium, however, is currently not routinely used in patients with impaired kidney function, because exposure to gadolinium has been found to be associated with a higher incidence of nephrogenic systemic fibrosis57. When not using gadolinium contrast, T2 weighted images might be preferred over T1 weighted images for the measurement of the TKV, because this technique shows high kidney tissue-contrast and hyperintense renal cysts, that may help to better delineate the kidney boundaries against background tissue58. Moreover, the single-shot T2 weighted techniques have evolved over the last years with for instance shorter examination time and fewer motion artifacts. This makes T2 weighted imaging potentially preferable over T1 weighted imaging for TKV measurement. In chapter 3 we therefore compared the performance of using T2 and T1 weighted MR images for measurement of TKV and growth in TKV in patients with ADPKD.

Since ADPKD is a tubular disease with an inflammatory component, measurement of urinary tubular damage and inflammation markers is of interest to predict the rate of disease progression, especially because these markers are relatively inexpensive and easy to measure. In chapter 4 we therefore investigated if urinary markers were

(17)

associated with ADPKD disease progression. In chapter 5 we investigated whether the markers that were identified in chapter 4 were associated with disease progression in an independent cohort of ADPKD patients.

Although it is never formally been tested, it is assumed that patients with ADPKD hyperfilter prior to a decline in GFR. If this can be confirmed, then the extent to which a patient is hyperfiltrating may possibly be the earliest marker of disease severity and predictor of disease progression. In chapter 6 we are the first to formally investigate if patients hyperfilter prior to a decline in GFR by using a generally accepted definition of hyperfiltration in chronic kidney disease, i.e. loss of kidney function reserve capacity. Elevated cAMP levels are one of the pivotal detrimental factors in the pathophysiology of ADPKD. As somatostatin has te ability to inhibit intracellular cAMP production, we hypothesized that endogenous somatostatin levels may be associated with ADPKD disease progression. In chapter 7 we therefore investigated if endogenous plasma somatostatin has potential to serve as a prognostic biomarker. Extending this line of reasoning, administration of somatostatin, in the form of somatostatin analogues, is a possible therapeutic option in ADPKD. In chapter 8 a review is given of the complex physiology of somatostatin, in particular in renal physiology and its potential therapeutic role in ADPKD. In addition, the results of studies with somatostatin analogues in ADPKD are discussed. As somatostatin analogues can inhibit cAMP production in a similar way as tolvaptan, through inhibition of AC activity, there may be an interaction between both pathways. In line, some studies have suggested that somatostatin is involved in renal water handling. In chapter 9 we investigated therefore if there are differences in diuresis and free water clearance in ADPKD patients using the somatostatin analogue lanreotide compared to patients using standard care. Furthermore, we investigated if differences were dependent on patient characteristics.

In 2016, tolvaptan, the first disease modifying drug has become available to treat patients with ADPKD. Initially, patients with signs of rapid disease progression and a relatively preserved kidney function were eligible for treatment59. Now clinical experience has accumulated and with the results of an additional recent clinical trial with tolvaptan47, an update was needed of the recommendations for the use of tolvaptan in ADPKD. In chapter 10 we therefore propose novel recommendations how to select patients with rapidly progressive disease for treatment with tolvaptan and demonstrate how these recommendations work in clinical practice.

(18)

REFERENCES

1. Patch C, Charlton J, Roderick PJ, Gulliford MC. Use of antihypertensive medications and mortality of patients with autosomal dominant polycystic kidney disease: A population-based study. Am J Kidney Dis. 2011;57(6):856-862.

2. Neumann HP, Jilg C, Bacher J, et al. Epidemiology of autosomal-dominant polycystic kidney disease: An in-depth clinical study for south-western germany. Nephrol Dial Transplant. 2013;28(6):1472-1487.

3. Tan YC, Blumenfeld JD, Anghel R, et al. Novel method for genomic analysis of PKD1 and PKD2 mutations in autosomal dominant polycystic kidney disease. Hum Mutat. 2009;30(2):264-273. 4. Torres VE, Harris PC. Strategies targeting cAMP signaling in the treatment of polycystic

kidney disease. J Am Soc Nephrol. 2014;25(1):18-32.

5. Cornec-Le Gall E, Audrezet MP, Chen JM, et al. Type of PKD1 mutation influences renal outcome in ADPKD. J Am Soc Nephrol. 2013;24(6):1006-1013.

6. Fain PR, McFann KK, Taylor MR, et al. Modifier genes play a significant role in the phenotypic expression of PKD1. Kidney Int. 2005;67(4):1256-1267.

7. Gabow PA, Johnson AM, Kaehny WD, et al. Factors affecting the progression of renal disease in autosomal-dominant polycystic kidney disease. Kidney Int. 1992;41(5):1311-1319. 8. Johnson AM, Gabow PA. Identification of patients with autosomal dominant polycystic kidney

disease at highest risk for end-stage renal disease. J Am Soc Nephrol. 1997;8(10):1560-1567. 9. Orskov B, Christensen KB, Feldt-Rasmussen B, Strandgaard S. Low birth weight is associated

with earlier onset of end-stage renal disease in danish patients with autosomal dominant polycystic kidney disease. Kidney Int. 2012;81(9):919-924.

10. Belibi FA, Wallace DP, Yamaguchi T, Christensen M, Reif G, Grantham JJ. The effect of caffeine on renal epithelial cells from patients with autosomal dominant polycystic kidney disease.

J Am Soc Nephrol. 2002;13(11):2723-2729.

11. Orth SR, Stockmann A, Conradt C, et al. Smoking as a risk factor for end-stage renal failure in men with primary renal disease. Kidney Int. 1998;54(3):926-931.

12. Wang CJ, Creed C, Winklhofer FT, Grantham JJ. Water prescription in autosomal dominant polycystic kidney disease: A pilot study. Clin J Am Soc Nephrol. 2011;6(1):192-197.

13. Wang CJ, Grantham JJ, Wetmore JB. The medicinal use of water in renal disease. Kidney Int. 2013;84(1):45-53.

14. Klahr S, Breyer JA, Beck GJ, et al. Dietary protein restriction, blood pressure control, and the progression of polycystic kidney disease. modification of diet in renal disease study group.

J Am Soc Nephrol. 1995;5(12):2037-2047.

15. Chapman AB, Devuyst O, Eckardt KU, et al. Autosomal-dominant polycystic kidney disease (ADPKD): Executive summary from a kidney disease: Improving global outcomes (KDIGO) controversies conference. Kidney Int. 2015;88(1):17-27.

16. Grantham JJ, Torres VE, Chapman AB, et al. Volume progression in polycystic kidney disease.

N Engl J Med. 2006;354(20):2122-2130.

17. Chapman AB, Guay-Woodford LM, Grantham JJ, et al. Renal structure in early autosomal-dominant polycystic kidney disease (ADPKD): The consortium for radiologic imaging studies of polycystic kidney disease (CRISP) cohort. Kidney Int. 2003;64(3):1035-1045.

18. Astor BC, Matsushita K, Gansevoort RT, et al. Lower estimated glomerular filtration rate and higher albuminuria are associated with mortality and end-stage renal disease. A collaborative meta-analysis of kidney disease population cohorts. Kidney Int. 2011;79(12):1331-1340.

(19)

19. Gansevoort RT, Matsushita K, van der Velde M, et al. Lower estimated GFR and higher albuminuria are associated with adverse kidney outcomes. A collaborative meta-analysis of general and high-risk population cohorts. Kidney Int. 2011;80(1):93-104.

20. Branten AJ, du Buf-Vereijken PW, Klasen IS, et al. Urinary excretion of beta2-microglobulin and IgG predict prognosis in idiopathic membranous nephropathy: A validation study. J Am

Soc Nephrol. 2005;16(1):169-174.

21. Tofik R, Aziz R, Reda A, Rippe B, Bakoush O. The value of IgG-uria in predicting renal failure in idiopathic glomerular diseases. A long-term follow-up study. Scand J Clin Lab Invest. 2011;71(2):123-128.

22. Groop L, Makipernaa A, Stenman S, DeFronzo RA, Teppo AM. Urinary excretion of kappa light chains in patients with diabetes mellitus. Kidney Int. 1990;37(4):1120-1125.

23. Shin JR, Kim SM, Yoo JS, et al. Urinary excretion of beta2-microglobulin as a prognostic marker in immunoglobulin A nephropathy. Korean J Intern Med. 2014;29(3):334-340. 24. van den Brand JA, Hofstra JM, Wetzels JF. Low-molecular-weight proteins as prognostic markers

in idiopathic membranous nephropathy. Clin J Am Soc Nephrol. 2011;6(12):2846-2853. 25. van Timmeren MM, Vaidya VS, van Ree RM, et al. High urinary excretion of kidney injury

molecule-1 is an independent predictor of graft loss in renal transplant recipients.

Transplantation. 2007;84(12):1625-1630.

26. Nauta FL, Boertien WE, Bakker SJ, et al. Glomerular and tubular damage markers are elevated in patients with diabetes. Diabetes Care. 2011;34(4):975-981.

27. Hofstra JM, Deegens JK, Steenbergen EJ, Wetzels JF. Urinary excretion of fatty acid-binding proteins in idiopathic membranous nephropathy. Nephrol Dial Transplant. 2008;23(10):3160-3165. 28. Tam FW, Riser BL, Meeran K, Rambow J, Pusey CD, Frankel AH. Urinary monocyte

chemoattractant protein-1 (MCP-1) and connective tissue growth factor (CCN2) as prognostic markers for progression of diabetic nephropathy. Cytokine. 2009;47(1):37-42.

29. Bruchfeld A, Carrero JJ, Qureshi AR, et al. Elevated serum macrophage migration inhibitory factor (MIF) concentrations in chronic kidney disease (CKD) are associated with markers of oxidative stress and endothelial activation. Mol Med. 2009;15(3-4):70-75.

30. Zhou LT, Lv LL, Pan MM, et al. Are urinary tubular injury markers useful in chronic kidney disease? A systematic review and meta analysis. PLoS One. 2016;11(12):e0167334.

31. Blank M, De Felice A, Goodsaid F, et al. Review of qualification data for biomarkers of nephrotoxicity submitted by the predictive safety testing consortium. . 2009.

32. Meijer E, Boertien WE, Nauta FL, et al. Association of urinary biomarkers with disease severity in patients with autosomal dominant polycystic kidney disease: A cross-sectional analysis. Am J Kidney Dis. 2010;56(5):883-895.

33. Petzold K, Poster D, Krauer F, et al. Urinary biomarkers at early ADPKD disease stage. PLoS

One. 2015;10(4):e0123555.

34. Parikh CR, Dahl NK, Chapman AB, et al. Evaluation of urine biomarkers of kidney injury in polycystic kidney disease. Kidney Int. 2012;81(8):784-790.

35. Boertien WE, Meijer E, Gansevoort RT. Urinary biomarkers in autosomal dominant polycystic kidney disease: Is there no prognostic value? Kidney Int. 2012;82(3):361.

36. Park HC, Hwang JH, Kang AY, et al. Urinary N-acetyl-beta-D glucosaminidase as a surrogate marker for renal function in autosomal dominant polycystic kidney disease: 1 year prospective cohort study. BMC Nephrol. 2012;13:93-2369-13-93.

37. Chapman AB, Bost JE, Torres VE, et al. Kidney volume and functional outcomes in autosomal dominant polycystic kidney disease. Clin J Am Soc Nephrol. 2012;7(3):479-486.

(20)

38. Nauta Ferdau L FL. Glomerular and tubular damage markers in individuals with progressive albuminuria. Clinical Journal of the American Society of Nephrology. 2013-7;8(7):1106-14. 39. RnD systems. Human MIF immunoassay. . 2016.

40. Nauta Ferdau L FL. Effect of frozen storage on urinary concentration of kidney damage markers. American Journal of Kidney Diseases. 2012-4;59(4):586-9.

41. Helal I, Fick-Brosnahan GM, Reed-Gitomer B, Schrier RW. Glomerular hyperfiltration: Definitions, mechanisms and clinical implications. Nat Rev Nephrol. 2012;8(5):293-300. 42. Battilana C, Zhang HP, Olshen RA, Wexler L, Myers BD. PAH extraction and estimation of

plasma flow in diseased human kidneys. Am J Physiol. 1991;261(4 Pt 2):F726-33.

43. ter Wee PM, Geerlings W, Rosman JB, Sluiter WJ, van der Geest S, Donker AJ. Testing renal reserve filtration capacity with an amino acid solution. Nephron. 1985;41(2):193-199. 44. ter Wee PM, Rosman JB, van der Geest S, Sluiter WJ, Donker AJ. Renal hemodynamics during

separate and combined infusion of amino acids and dopamine. Kidney Int. 1986;29(4):870-874. 45. Boertien WE, Meijer E, Li J, et al. Relationship of copeptin, a surrogate marker for arginine

vasopressin, with change in total kidney volume and GFR decline in autosomal dominant polycystic kidney disease: Results from the CRISP cohort. Am J Kidney Dis. 2013;61(3):420-429. 46. Torres VE, Chapman AB, Devuyst O, et al. Tolvaptan in patients with autosomal dominant

polycystic kidney disease. N Engl J Med. 2012;367(25):2407-2418.

47. Torres VE, Chapman AB, Devuyst O, et al. Tolvaptan in later-stage autosomal dominant polycystic kidney disease. N Engl J Med. 2017;377(20):1930-1942.

48. Winkler SN, Torikai S, Levine BS, Kurokawa K. Effect of somatostatin on vasopressin-induced antidiuresis and renal cyclic AMP of rats. Miner Electrolyte Metab. 1982;7(1):8-14.

49. Ishikawa S, Saito T, Kuzuya T. Reversal of somatostatin inhibition of AVP-induced cAMP by pertussis toxin. Kidney Int. 1988;33(2):536-542.

50. Patel YC. Somatostatin and its receptor family. Front Neuroendocrinol. 1999;20(3):157-198. 51. Rieg T, Kohan DE. Regulation of nephron water and electrolyte transport by adenylyl cyclases.

Am J Physiol Renal Physiol. 2014;306(7):F701-9.

52. Brautbar N, Levine BS, Coburn JW, Kleeman CR. Interaction of somatostatin with PTH and AVP: Renal effects. Am J Physiol. 1979;237(5):E428-36.

53. Walker BJ, Evans PA, Forsling ML, Nelstrop GA. Somatostatin and water excretion in man: An intrarenal action. Clin Endocrinol (Oxf). 1985;23(2):169-174.

54. Vora JP, Owens DR, Ryder R, Atiea J, Luzio S, Hayes TM. Effect of somatostatin on renal function. Br Med J (Clin Res Ed). 1986;292(6537):1701-1702.

55. Ray C, Carney S, Morgan T, Gillies A. Somatostatin as a modulator of distal nephron water permeability. Clin Sci (Lond). 1993;84(4):455-460.

56. Bae KT, Tao C, Zhu F, et al. MRI-based kidney volume measurements in ADPKD: Reliability and effect of gadolinium enhancement. Clin J Am Soc Nephrol. 2009;4(4):719-725.

57. Thomsen HS, European Society of Urogenital Radiology (ESUR). ESUR guideline: Gadolinium-based contrast media and nephrogenic systemic fibrosis. Eur Radiol. 2007;17(10):2692-2696. 58. Bae KT, Grantham JJ. Imaging for the prognosis of autosomal dominant polycystic kidney

disease. Nat Rev Nephrol. 2010;6(2):96-106.

59. Gansevoort RT, Arici M, Benzing T, et al. Recommendations for the use of tolvaptan in autosomal dominant polycystic kidney disease: A position statement on behalf of the ERA-EDTA working groups on inherited kidney disorders and european renal best practice.

Nephrol Dial Transplant. 2016;31(3):337-348.

(21)
(22)

2

Estimation of total kidney volume in autosomal

dominant polycystic kidney disease

Edwin M. Spithoven Maatje D.A. van Gastel*

A. Lianne Messchendorp*

Niek F. Casteleijn Joost P.H. Drenth Carlo A.J.M. Gaillard Johan W. de Fijter Esther Meijer Dorien J.M. Peters Peter Kappert Remco J. Renken Folkert W. Visser Jack Wetzels Robert Zietse Ron T. Gansevoort on behalf of the DIPAK Consortium

* both authors contributed equally

(23)

ABSTRACT

Background

In autosomal dominant polycystic kidney disease (ADPKD), obtaining measured total kidney volume (mTKV) by magnetic resonance (MR) imaging and manual tracing is time consuming. Two alternative MR imaging methods have recently been proposed to estimate TKV (eTKVellipsoid and eTKVPANK), which require less time. We investigated

if eTKVellipsoid and eTKVPANK could be measured as reliable and reproducible as mTKV in patients with ADPKD.

Methods

For this study we included patients with ADPKD with a wide range of kidney function and an approved T2-weighted MR image. First, we investigated the reproducibility of mTKV and eTKV in a test-set of ADPKD patients. Second, we assessed bias, precision and accuracy of eTKV cross-sectionally in a cohort of ADPKD patients. Third, in a subgroup, we determined the association between change in mTKV and change in eTKV over time longitudinally.

Results

In the test set, intra- and intercoefficients of variation for mTKV, eTKVellipsoid, and eTKVPANK were 1.8% and 2.3%, 3.9% and 6.3%, and 3.0% and 3.4%, respectively. In

cross-sectional analysis, baseline mTKV, eTKVellipsoid, and eTKVPANK were 1.96 (IQR, 1.28-2.82), 1.93 (IQR, 1.25-2.82), and 1.81 (IQR, 1.17-2.62) L, respectively. Bias was 0.02%±3.2%, 1.4%±9.2%, and 4.6%±7.6% for repeat mTKV, eTKVellipsoid, and eTKVPANK, respectively. In longitudinal analysis, no significant differences were observed between percentage change in mTKV (16.7%±17.1%) and percentage change in eTKVellipsoid (19.3%±16.1%) and eTKVPANK (17.8%±16.1%) over 3 years.

Conclusions

Both methods for eTKV perform relatively well compared to mTKV and can detect change in TKV over time. Because eTKVellipsoid requires less time than eTKVPANK, we

(24)

INTRODUCTION

Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the formation and growth of numerous cysts in both kidneys, leading to an increase

in kidney volume. These cysts compress healthy kidney tissue, causing progressive kidney function decline and, in most patients, ultimately a need for renal replacement therapy. In patients with ADPKD, total kidney volume (TKV) has been shown to be an early marker of disease severity and predictor of kidney function decline1.

Measurement of TKV is therefore used to assess prognosis in clinical care and for selection of patients for randomized controlled trials2. In these trials that investigate

potential treatments for patients with ADPKD, assessment of TKV is often used as

the primary or secondary study end point3-5.

The true gold-standard method to assess TKV is the manual tracing method. Computer tomogram or magnetic resonance (MR) images are used, and in each slice, the kidney boundaries are traced manually using dedicated software. Measured TKV (mTKV)

is calculated from a set of contiguous images by summing the products of the area

measurements within the kidney boundaries and slice thickness6. This method is

laborious, which limits its use in trial settings, but especially in clinical care.

If kidney volume could be estimated with sufficient accuracy and reliability, it would

alleviate the time-consuming process of kidney volume measurement. Recently, 2

kidney volume estimation methods have been developed: the midslice method7 by

the Consortium for Radiologic Imaging Studies of ADPKD (CRISP) and the ellipsoid

method2 by the Mayo Clinic. For both methods, measured and estimated kidney

volumes appeared to be well correlated, but other groups have not yet validated

these methods. In addition, the midslice method was developed in a cohort that

included only patients with creatinine clearance 70 mL/min. In general, such patients

have relatively small kidneys, making manual tracing measurement of TKV relatively

easy, which may have influenced the results that were obtained. This method should

therefore also be validated in patients with lower kidney function. Estimation methods

to assess TKV may also be used in clinical trials, but only when they can accurately

and reliably detect changes in TKV over time. To our knowledge, these issues have

not been investigated to date.

(25)

Given these considerations, the objective of the present study was to investigate

cross-sectionally these methods to estimate TKV in a patient group with a wide range

of kidney function. Furthermore, we investigated in a longitudinal study whether

these estimation methods can accurately detect changes in TKV.

METHODS

Patients and study design

For this study, all MR images of patients with ADPKD that were available from 2007 through 2014 were used. These patients participated in 1 of 3 studies that were performed by the departments of nephrology at the University Medical Centers of Groningen, Leiden, Nijmegen, and Rotterdam (all in the Netherlands). Details of the study protocols have been published elsewhere4,8,9; see Figure S1 for a flow diagram

showing the assembly of the cohort. All patients were included if an MR image was available. ADPKD was diagnosed based on the modified Ravine criteria10. The

Medical Ethics Committee of the University Medical Center Groningen approved the protocols of the 3 studies that were conducted in accordance with the International Conference of Harmonization Good Clinical Practice Guidelines and in adherence to the ethics principles that have their origin in the Declaration of Helsinki. All patients gave written informed consent.

Measurement and collections

All participants collected a 24-hour urine sample the day preceding the MR imaging (MRI), in which urinary albumin concentration was measured. At the outpatient clinic on the day of MRI, blood pressure was assessed at rest in a supine position with an automatic device (Dinamap; GE Medical Systems) for 15 minutes and weight and height were determined. Blood samples were drawn for determination of creatinine level with an enzymatic assay (isotope-dilution mass spectrometry traceable; Modular; Roche Diagnostics), which was used to estimate glomerular filtration rate (GFR) using the CKD-EPI (Chronic Kidney Disease Epidemiology Collaboration) equation11.

MR imaging

All participants underwent a standardized abdominal MRI protocol without the use of intravenous contrast. For the specific MRI protocol, see Item S1.

(26)

Gold-standard method: mTKV

Kidney and liver volumes were measured on the coronal fat saturated T2-single shot fast spin-echo sequence if possible. If the T2-weighted images showed too low quality, the MR image was excluded. Kidney and liver volumes were measured using the manual tracing method. Kidney and liver boundaries were manually traced using the commercially available software Analyze Direct 11.0 (Analyze Direct Inc). Kidney and liver volumes were calculated from the set of contiguous images by summing the products of the area measurements within the kidney or liver boundaries and slice thickness6. Nonrenal parenchyma (e.g. the renal hilus) was excluded from measurement.

Estimation methods: estimated TKV

The 2 formulas used to estimate kidney volume were derived from the literature2,7.

We first used the midslice method to estimate TKV (eTKVPANK)7. The midslices of the

coronal MR images were selected for each kidney separately. The midslice was defined as the slice for which the slice number corresponds to half the sum of the numbers of the first and last slice that contained the kidney. If the sum was odd, the midslice number was rounded up. eTKVPANK was calculated in milliliters, with midslice area

and slice thickness in millimeters squared and millimeters, respectively. eTKVPANK was calculated as the sum of the left eKVPANK (i.e., 0.624 * midslice area * number of slices

covering the left kidney * slice thickness/1000) and right eKVPANK (i.e., 0.637 * midslice area * number of slices covering the right kidney * slice thickness/1000).

Second, we used the ellipsoid method to estimate TKV (eTKVellipsoid)2. For each kidney,

length was measured as the average maximal longitudinal diameter measured in the coronal and sagittal plane. Width was obtained from the transversal image at maximum transversal diameter, and depth was measured from the same image perpendicular to the width measurement. eTKVellipsoid was calculated in milliliters, with length, width,

and depth all in millimeters. eTKVellipsoid was calculated as the sum of the left KVellipsoid and right KVellipsoid, both derived by the equation π/6 * (lengthcoronal + lengthsagittal)/2 *

width * depth/1000. Of note, to assess eTKVellipsoid, no specific software is necessary, in contrast to assessment of mTKV and eTKVPANK.

Statistical analyses

All analyses were performed with SPSS, version 22.0 (SPSS Inc). Normality of data was assessed by drawing Q-Q plots. Normally distributed variables are expressed as mean ± standard deviation, whereas non-normally distributed variables are given as

(27)

median with interquartile range (IQR). Baseline characteristics of the study population are given overall (Table 1) and stratified for estimated GFR (eGFR) <60 and ≥60 ml/ min/1.73m2 (Table S1). Differences between groups were tested using a 2-sample t

test for normally distributed and Mann-Whitney U test for non-normally distributed data. For paired analyses, a paired t test was used for normally distributed and a Wilcoxon signed rank test was used for non-normally distributed data. McNemar test was used for paired nominal data. A 2-sided p<0.05 was considered to indicate statistical significance. In a test set of 10 patients stratified for kidney volume and MRI scanner, kidney volumes were measured and estimated twice by 4 reviewers (MDAvG, JvM, BvS, JvE). All reviewers were blinded to their previous results. Reproducibility was evaluated by assessing intra- and intercoefficient of variation (CV) for mTKV,

eTKVellipsoid, and eTKVPANK. The inter-CV was calculated for each of the 10 MR images

as the standard deviation of TKV values assessed by all 4 assessors divided by the mean TKV of that image multiplied by 100%. The inter-CV given in this study is the mean of the inter-CVs of these 10 MR images. Intra-CV was calculated per MR image for each of the 4 assessors as the standard deviation of TKV values divided by the mean TKV multiplied by 100%. Per assessor, an average intra-CV was calculated. The intra-CV given in this study is the mean intra-CV (plus standard deviation) of these 4 assessors. We used paired t test to compare CVs between mTKV and eTKV.

To investigate whether eTKV correlated with mTKV, orthogonal regression analysis was performed, and Lins’ concordance correlation coefficient was calculated using all MRI scans of our cohort12. Orthogonal regression uses the least square data modeling

technique in which observational errors in both dependent and independent variables are taken into account. Agreement between eTKV and mTKV was evaluated by Bland-Altman analyses, with calculation of agreement limits (95% confidence interval). We used manual tracing as the gold standard for TKV measurement on the x-axis. Performance of the estimation methods compared with mTKV was assessed using bias, precision, and accuracy. For cross-sectional analyses, bias is expressed as mean percentage difference ([mTKV - eTKV]/mTKV * 100%), with positive values indicating underestimation of mTKV. Precision was defined as 1 standard deviation of bias. Accuracy was calculated as the percentage of eTKV values within 10%, 15%, and 20% of mTKV [P10, P15, and P20 respectively]). To investigate whether bias is dependent on patient or MR image characteristics, we performed regression analyses between bias and various variables; that is, age, length, body mass index, liver volume, and T1/T2-weighted images in univariate analyses. Differences in bias among the various

(28)

scanners that were used were tested with analysis of variance. As standard quality control, ~10% of all MRI scans were measured twice for mTKV, and this is referred to as

mTKVrepeat. This was done to ensure that the observers maintained low interobserver

variability. These scans were used to assess the precision and bias of mTKV.

To investigate whether the estimation methods can accurately detect changes in TKV, data for patients who had follow-up MR images available were used. For these longitudinal analyses, bias is expressed as the percent change in mTKV less the percent change in eTKV. Importantly, all follow-up scans were performed at the same MRI scanner as at baseline, and TKV was measured and estimated using the same series of images as at baseline, by reviewers blinded for baseline results.

To assess the consequences of using eTKV instead of mTKV, 2 analyses were performed. First, the effect on classification based on disease prognosis was assessed. To assess prognosis for clinical care, a classification system is used that categorizes patients into 5 classes based on thresholds for height-corrected TKV at a given age (A through E, with A indicating the best and E indicating the worst prognosis with respect to future kidney function decline)2. In addition, there is a classification indicating whether a

patient is suitable for inclusion in clinical trials. This classification contains 3 classes: patients who should not be included in clinical trials [I], patients whose suitability should be re-evaluated at yearly intervals [II], and patients who are optimal candidates for clinical trials [III])2. To assess reclassification, we created 5 * 5 and 3 * 3

cross-tabulations using height-corrected TKV limits for their specific age2. In these tables,

the proportion of reclassified participants was calculated when using height corrected eTKV instead of height corrected mTKV. For this analysis, only the “typical cases” were used, as advised for this classification system, defined as MR images with cysts with bilateral and diffuse distribution, in which all cysts contribute similarly to TKV2.

Second, we assessed what the consequences were for sample size calculation for clinical trials using change in eTKV instead of change in mTKV. Sample size calculations were based on the literature13 and used data from all patients who had longitudinal

follow-up data available with respect to change in mTKV and eTKV. The number of patients needed per group was calculated assuming a power of 80% and 2-sided α of 0.05 to detect a percentage difference in TKV growth between treatment groups.

(29)

RESULTS

Study participants

The study population consisted of 220 patients with ADPKD; their characteristics are listed in Table 1. We excluded 44 patients because no T2-weighted images were available to perform both estimation methods. The patients were relatively young, with a mean age of 47.0 ± 8.6 years, and already showed clear signs of disease. Most patients used antihypertensive medication. eGFRs were decreased (56.8 ± 20.3 [range, 17.0-129.2] ml/min/1.73m2). Urinary albumin excretion (46.7 [IQR, 21.2-88.2] mg/24

hour) and TKV (1.96 [IQR, 1.28-2.82] L) were increased.

Table 1. Participants’ characteristics.

Whole study group (n=220) Patients with follow-up (n=48) Test set (n=10) Age (y) 47.0 ± 8.6 39.2 ± 7.4 44.3 ± 10.2 Male (% (n)) 51.8 (114) 70.8 (34) 3 (30)

Body mass index (kg/m2) 26.9 ± 4.3 26.3 ± 3.4 27.1 ± 7.2

Body surface area (m2) 2.0 ± 0.2 2.1 ± 0.2 1.96 ± 0.2

Diastolic blood pressure (mmHg) 82.2 ± 9.5 82.6 ± 8.8 85.4 ± 11.0 Systolic blood pressure (mmHg) 132.7 ± 13.0 132.9 ± 11.6 134.1 ± 18.0 Antihypertensive medication (% (n)) 86.4 (190) 81.3 (39) 9 (90) Plasma creatinine (mmol/L) 125.5 ± 39.7 102.1 ± 31.7 127. ± 6 20.4 eGFR (mL/min/1.73m2) 56.8 ± 20.3 79.7 ± 22.6 49.6 ± 10.2

24h Urine volume (L) 2.36 ± 0.77 2.48 ± 0.87 2.60 ± 0.80 Albuminuria (mg/24h) 46.7 (21.2-88.2) 46.2 (19.0-181.0) 67.9 (17.0-95.4) Total kidney volume (L) 1.96 (1.28-2.82) 1.79 (1.36-2.56) 1.78 (1.37-2.86) - Left kidney volume (L) 1.00 (0.67-1.52) 0.99 (0.73-1.39) 0.92 (0.70-1.62) - Right kidney volume (L) 0.92 (0.60-1.38) 0.80 (0.57-1.17) 0.91 (0.67-1.24) Liver volume (L) 2.74 (1.73-3.07) NA 1.76 (1.62-3.64)

Values for categorical variables are given as number (percentage); values for continuous variables, as mean ± standard deviation or median (interquartile range).

Abbreviations are: BP, blood pressure; eGFR, estimated glomerular filtration rate; NA, not available.

Reproducibility of mTKV and eTKV

Table 2 shows a test set for assessing reproducibility. Average intraobserver CVs were 1.8% for mTKV and 2.6% for total liver volume, whereas interobserver CVs were 2.3% and 3.5%, respectively. Variability for eTKVellipsoid was significantly higher than for mTKV, whereas for eTKVPANK, no significant differences were found when compared

(30)

65 minutes for total liver volume, with higher analysis times in case of larger organs. Average time needed per MR image to estimate TKV using the midslice method was 15 minutes; using the ellipsoid method, 5 minutes.

Table 2. Test set for assessing reproducibility.

Both

kidneys kidneyLeft kidneyRight

mTKV - Intra-observer CV (%) 1.8 2.3 1.9 - Inter-observer CV (%) 2.3 2.6 2.9 eTKVellipsoid - Intra-observer CV (%) 3.9* 4.9* 4.3* - Inter-observer CV (%) 6.3* 6.0* 8.5* eTKVPANK - Intra-observer CV (%) 3.0 3.8 3.1 - Inter-observer CV (%) 3.4 4.2 3.1

All CVs were calculated based on 10 patients.

Abbreviations are: CV, coefficient of variation; eKVellipsoid, estimated kidney volume using ellipsoid method;

eKVPANK, estimated kidney volume using midslice method; mKV, measured kidney volume.

*p-value <0.05 for difference in intra- or inter-observer CV eTKV versus corresponding value of mTKV

Performance of the TKV estimation methods

In the cohort for cross-sectional analyses, correlations of mTKV versus mTKVrepeat,

eTKVellipsoid, and eTKVPANK are shown in Figure 1. Figures S2 and S3 show these correlations

for left and right kidneys, separately. High correlations were observed for all 3 methods (mTKVrepeat: R= 0.998, p<0.001; eTKVellipsoid: R=0.989, p<0.001; and eTKVPANK: R=0.990, p<0.001). Figure 1 also shows Bland-Altman plots of mTKV versus the percentage difference between mTKV and mTKVrepeat and both eTKV methods. mTKVrepeat showed low bias (mean, 0.02% ± 3.2%). eTKV also did not systematically over- or underestimate mTKV (bias of 1.4% ± 9.2% and 4.6% ± 7.6% for eTKVellipsoid and eTKVPANK, respectively; Table 3). Bias for eTKVPANK was significantly higher than for mTKVrepeat (p=0.005), whereas

bias for eTKVellipsoid did not significantly differ from that for mTKVrepeat (p=0.4). Given the lower standard deviation, mTKVrepeat had better precision and therefore better

performance compared with eTKVellipsoid and eTKVPANK.

(31)

Table 3. Cohort for cross-sectional analyses: Performance of ellipsoid and midslice methods

for eKV

P for mKVrepeat vs

eTKVellipsoid

(n=220) eTKV(n=220)PANK mTKV(n=28)repeat eTKVellipsoid eTKVPANK Left kidney volume (L) 1.03 (0.65 – 1.48) 0.95 (0.63 – 1.45) 1.03 (0.75 – 1.78) 0.3 <0.001 - Bias (%) -0.7 5.6 0.1 0.9 0.003 - Precision (%) 11.8 9.7 3.6

Right kidney volume (L) 0.90 (0.57 – 1.37) 0.88 (0.54 – 1.33) 0.98 (0.67 – 1.51) 0.003 <0.001 - Bias (%) 2.0 3.2 0.4 0.048 0.10 - Precision (%) 12.4 11.1 3.9

Total kidney volume (L) 1.93 (1.25 – 2.82) 1.81 (1.17 – 2.62) 1.92 (1.51 – 3.18) 0.004 <0.001 - Bias (%) 1.4 4.6 0.2 0.4 0.005 - Precision (%) 9.2 7.6 3.2 - Accuracy P10 78.1 82.1 100 <0.001 <0.001 P15 92.7 93.6 100 <0.001 <0.001 P20 97.7 96.4 100 <0.001 <0.001 - CCC 0.988 0.987 0.998

Values are given as percentage or median (interquartile range). P values are calculated by paired t-test when normally distributed, Wilcoxon signed rank test when non-normally distributed for continuous variables, and McNemar test for nominal variables.

Abbreviations and definitions: accuracy, percentage of eKV values within 10% (P10), 15% (P15), and 20%

(P20) of their corresponding mKV value; bias, mean percentage difference between mKV and eKV; CCC, concordance correlation coefficient; eKVellipsoid, estimated kidney volume using ellipsoid method; eTVPANK,

estimated kidney volume using midslice method; mTVrepeat, repeated measured kidney volume; precision, 1 standard deviation of bias.

In addition, when these analyses were repeated with patients with ADPKD stratified for eGFR, we observed no significant difference in bias for eTKVellipsoid and mTKVrepeat

in patients with eGFRs <60 ml/min/1.73m2 and eGFRs ≥60 ml/min/1.73m2 (p=0.2 and

p=0.3, respectively). Between eTKVPANK and mTKVrepeat, we also observed no significant

difference in patients with eGFR <60 ml/min/1.73m2 (p=0.2) and those with eGFR ≥60

ml/mn/1.73m2 (p=0.9). Table S2 shows bias and accuracy for eTKV stratified by eGFR.

When investigating factors associated with bias, it appeared that liver volume was associated with bias in eTKVPANK (p=0.04), but not with eTKVellipsoid (p=0.1). Bias was not associated with age (p=0.5 and p=0.6), height (p=0.8 and p=0.1), or strength of magnetic field (p=0.8 and p=0.7), respectively, for eTKVellipsoid and eTKVPANK.

(32)

Figure 1. Cohort for cross-sectional analyses: associations between measured total kidney

volume (mTKV) and repeated mTKV (mTKVREPEAT) (upper panels), estimated TKV using the

ellip-soid method (eTKVellipsoid) (middle pannels) and the mid-slice method (eTKVPANK) (lower panels).

Left panel shows scatter plots (solid line representing the line of identity and the dotted line the actual regression line), whereas the right panel shows Bland-Altman plots (solid line indicating no difference and dotted lines representing mean difference [i.e. bias] with 95% confidence interval).

(33)

Ability to detect changes in TKV when using estimation methods

Follow-up data for TKV were available for 48 patients. Baseline characteristics for the longitudinal cohort are given in Table 1. These patients were younger, showed fewer signs of disease, and had higher eGFRs (79.7±22.6 mL/min/1.73 m2) but similar

urinary albumin excretion (46.2 [IQR, 19.0-181.0] mg/24 hour). During a follow-up of 3.0 years, mTKV increased from 1.79 (IQR, 1.36-2.56) to 2.18 (IQR, 1.55-2.73) L (p<0.001). Median differences during follow-up were 0.25 (IQR, 0.04-0.54), 0.30 (IQR, 0.08-0.86), and 0.28 (IQR, 0.08-0.54) L for mTKV, eTKVellipsoid, and eTKVPANK, respectively (Table 4). Change in eTKV compared to change in mTKV was not significantly different for both estimation methods (p=0.2 and p=0.5 for eTKVellipsoid and eTKVPANK, respectively). Figure 2 plots percentage change in mTKV versus percentage change in eTKV. High concordance correlations were observed for eTKVellipsoid (R=0.798, p<0.001) and eTKVPANK (R=0.866, p<0.001). Percentage change in eTKV did not show systematic under- or overestimation, with bias and precision (percent change mTKV - percent change eTKV) of 22.2% ± 10.3% and 21.8% ± 8.3% for eTKVellipsoid and eTKVPANK, respectively (Figure

2). In most patients, bias for change in eTKV was between -10% and 10% (72.3% and 74.5% of patients for eTKVellipsoid and eTKVPANK, respectively).

Table 4. Cohort for longitudinal analyses.

Baseline (L) Follow-up (L) Change (L) Change (%)

Both kidneys mTKV 1.79 (1.36-2.56) 2.18 (1.55-2.73) 0.25 (0.04-0.54) 16.7 ± 17.1 eTKVellipsoid 1.86 (1.32-2.75) 2.39 (1.50-2.80) 0.30 (0.08-0.86) 19.3 ± 16.1 eTKVPANK 1.79 (1.12-2.43) 2.03 (1.49-2.63) 0.28 (0.08-0.54) 17.8 ± 16.1 Left kidney mTKV 0.99 (0.74-1.39) 1.23 (0.83-1.56) 0.13 (0.01-0.29) 15.0 ± 18.7 eTKVellipsoid 1.03 (0.70-1.44) 1.26 (0.85-1.58) 0.10 (0.04-0.37) 17.7 ± 18.1 eTKVPANK 0.92 (0.68-1.24) 1.10 (0.78-1.44) 0.17 (0.04-0.36)* 19.7 ± 19.0* Right kidney mTKV 0.80 (0.57-1.17) 0.99 (0.68-1.29) 0.13 (0.06-0.25) 19.4 ± 18.6 eTKVellipsoid 0.81 (0.58-1.10) 1.04 (0.65-1.39) 0.14 (0.04-0.29) 23.1 ± 22.8 eTKVPANK 0.78 (0.60-1.14) 0.90 (0.65-1.24) 0.13 (0.04-0.24) 17.0 ± 19.6

Baseline and follow-up (T)KV data for 48 patients with autosomal dominant polycystic kidney disease with follow-up data available. Values are given as mean ± standard deviation or median (interquartile range). No significant differences between change in e(T)KV versus change in m(T)KV were noted, except for change in left eKVPANK (as indicated with *).

Abbreviations are: e(T)KVellipsoid, estimated (total) kidney volume using ellipsoid method; e(T)KVPANK, estimated

(total) kidney volume using midslice method; mTKV, measured total kidney volume. * p-value <0.05.

(34)

Figure 2. Cohort for longitudinal analyses: associations between percentage change in

mea-sured total kidney volume (mTKV) and percentage change in estimated total kidney volume (eTKV) using the ellipsoid method and the mid-slice method in 48 ADPKD patients who had fol-low-up data available. Left panel shows scatter plots (solid line representing the line of identity and dotted line the actual regression line), whereas the right panel shows Bland-Altman plots (solid horizontal line indicating no difference, and dotted lines representing mean difference [i.e. bias] with 95% confidence interval).

Consequences of using eTKV instead of mTKV

When using eTKV methods instead of mTKV for risk classification with respect to prognosis for rapid kidney function decline, we excluded the radiologically atypical ADPKD cases (n=27), as advised for this classification system. There were 93.3% (eTKVellipsoid) and 90.2% (eTKVPANK) of patients reclassified to their original risk categories

(Table 5), whereas for both estimation methods, <1.6% of patients were reclassified

(35)

to a higher risk category, and <8.5%, to a lower risk category. For classification for selection of patients for clinical trials, we observed that 97.4% (eTKVellipsoid) and 95.9% (eTKVPANK) of patients were reclassified to their original categories. No patients were

reclassified to a higher risk category when using eTKVellipsoid, and only 1 patient, when using eTKVPANK (Table 5).

Table 5. Reclassification for staging into risk categories for rapid kidney function decline. Risk category classification

eTKVellipsoid eTKVPANK

A B C D E A B C D E mTKV A 5 A 4 1 B 28 B 1 27 C 5 66 2 C 6 65 2 D 4 47 1 D 6 45 E 1 35 E 3 33

Patient selection for trials

eTKVellipsoid eTKVPANK

I II III I II III

mTKV I 5 I 4 1

II 28 II 1 27

III 5 155 III 6 150

Based on Irazabal et al2. Reclassification for staging into risk categories for rapid kidney function decline for

clinical care (A-E) and for selection of patients for clinical trials based on thresholds for height-corrected TKV at a given age (I-III) using ellipsoid method (eTKVellipsoid) and using midslice method (eTKVPANK) instead of mTKV.

Abbreviations are: eTKVellipsoid, estimated total kidney volume using ellipsoid method; eTKVPANK, estimated total kidney volume using midslice method; mTKV, measured total kidney volume.

The consequences of using percentage change in eTKV instead of percentage change in mTKV as the end point for sample size calculation for randomized controlled trials were assessed using data from the 48 patients with ADPKD for whom follow-up data were available. We calculated the number of study participants per treatment group needed to be enrolled to demonstrate a certain percentage decrease in rate of growth in TKV. Results are shown in Table S3. To detect, for instance, a 30% decrease in rate of growth in mTKV over 3 years, 186 patients are needed per treatment group, whereas for eTKVellipsoid and eTKVPANK these numbers are 122 and 143, respectively.

(36)

DISCUSSION

This study was conducted to investigate whether TKV can be estimated accurately using the midslice (PANK) and ellipsoid methods in a group of patients with ADPKD with a wide range of kidney function. In a test set of 10 patients with ADPKD, we found that both estimation methods were highly reproducible. In our study cohort of 220 patients with ADPKD, both methods showed low bias, high precision, and high accuracy when compared to mTKV. This held for the overall cohort, as well as for patients with higher and lower eGFRs. In the 48 patients who had follow-up MR images available, change in eTKV was not different from change in mTKV for both methods.

Assessment of TKV using the gold-standard method of manual tracing is time consuming and needs specific software, which limits its applicability for clinical care. Methods have therefore been sought to estimate TKV in a more feasible way. Two methods have been published recently2,7; however, they have not been validated to date. This

formed the rationale to perform the present study. For determination of whether these estimation methods can be used to assess TKV, it is important to answer the following 5 questions.

First, it is important to investigate what the reliability of the gold-standard method is. In our study, we found that the variability in volumetric assessment by manual tracing was very low. In general, T1- instead of T2-weighted images are used for volumetry in ADPKD because researchers want to align with the original CRISP methodology. However, when the CRISP Study started, gadolinium-enhanced T1-weighted MR images were used. Because of the potential adverse effects of gadolinium, use of this contrast agent has since been discouraged. Bae et al14 showed in 2009 that unenhanced

T1-weighted volumes were significantly lower than contrast enhanced T1-weighted volumes. These differences were more pronounced in smaller kidneys because in such cases, the ratio of kidney boundaries area to kidney volume is higher. Bae et al14

mentioned that one should therefore contemplate using T2 MRI for quantification of TKV because the high kidney tissue contrast and hyperintense renal cysts in T2 images aid in delineating kidney boundaries against background tissues when compared to T1-weighted images. At that time, T2-weighted imaging required longer scanning time and was subjected to increased variation in image quality because of motion artefacts and was therefore not feasible. Nowadays, T2-weighted scanning time is shorter and respiratory triggering to avoid motion artefacts has become available. In

(37)

our experience, this sequence has the best quality in visualizing polycystic kidneys. We therefore chose T2-weighted images instead of T1-weighted images for our study.

Second, do these estimation methods show low variability? Variability in mTKV versus eTKVPANK was not significantly different and satisfactorily low. Variability in eTKVellipsoid

was significantly higher compared to mTKV, meaning that this method is slightly more operator dependent than the midslice method, but still low. In line with this, reclassification to another risk category for rapid kidney function decline for clinical care (Irazabal classes A-E2) happened infrequently when using eTKV

PANK, as well as

eTKVellipsoid (Table 5). Given these results and because eTKVellipsoid is more convenient

(shorter duration per MR image and assessment possible using standard MRI software), we advise that eTKVellipsoid be used rather than eTKVPANK for risk assessment in clinical care.

Third, does the estimation method show good agreement with the gold-standard method? We found for both estimation methods that eTKV correlated strongly with mTKV. Although bias and precision again showed better values for mTKVrepeat (0.02%

and 3.2%, respectively), results for eTKVellipsoid and eTKVPANK were good. Bias was low for eTKVellipsoid and eTKVPANK (1.4% and 4.6%, respectively), although for eTKVPANK, it

was slightly (but significantly) higher than for mTKVrepeat. In addition, precision was reasonable, now with slightly better results for eTKVellipsoid (eTKVellipsoid and eTKVPANK:

9.2% and 7.6%, respectively; Table 3). Consequently, we found good accuracy for both estimation methods (P20 for eTKVPANK and eTKVellipsoid of 96.4% and 97.7%, respectively).

Our findings with respect to accuracy are consistent with values obtained in the cohort in which the ellipsoid method was developed (P10 of 70.3% vs. 78.1% in the present

study)2. When stratified for kidney function, our results with respect to bias suggest

that the midslice method may be less accurate in patients with ADPKD with lower kidney function, who generally have larger kidneys. Besides these statistical data, consequences for clinical care should be investigated when answering the question of whether estimation methods show good agreement with the gold-standard method. Irazabal et al2 proposed a classification system for patients with ADPKD to assess their

risk for rapid kidney function decline and to guide selection of patients for clinical trials. This classification system uses thresholds defined by age- and height-corrected TKV. We investigated the percentage of patients who are reclassified when using eTKV instead of mTKV. In the classification system for risk assessment, we observed that only a limited percentage of patients were reclassified, and these patients were most

Referenties

GERELATEERDE DOCUMENTEN

Given these findings, we hypothesized that endogenous, systemic SST levels are involved in the pathophysiologic cascade of ADPKD and therefore is associated with urinary cAMP,

Several preclinical and clinical studies have been conducted that studied the efficacy of SST analogues to inhibit cAMP production, hepatic and kidney cyst growth, and renal

Overall, there were no differences in change in 24-hour urine volume, FWC, FFWC or copeptin levels in patients with ADPKD receiving 12 weeks of standard care or the somatostatin

In Figure 6 patients are plotted according to eGFR by age with the theoretical threshold line and marked as slowly or rapidly progressive disease predicted by the Genetic Risk

The association of combined total kidney and liver volume with pain and gastrointestinal symptoms in patients with later stage autosomal dominant polycystic kidney

Tubulaire schade- en inflammatiemarkers in de urine zijn daarom veelbelovend om in de klinische zorg gebruikt te worden om ADPKD patiënten met een snelle ziekteprogressie

Daarnaast wil ik de rest van mijn collega’s van het triade/de brug bedanken; Alicia, Antonio (die altijd in is voor een feestje), Arno (die al om 10:30 vraagt of we die avond

The association of combined total kidney and liver volume with pain and gastrointestinal symptoms in patients with later stage autosomal dominant polycystic kidney disease.