• 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!
15
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)

1

Introduction

(3)

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.

(4)

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.

(5)

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

(6)

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.

(7)

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.

(8)

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

(9)

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

(10)

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

(11)

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.

(12)

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.

(13)

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.

(14)

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.

(15)

Referenties

GERELATEERDE DOCUMENTEN

Since we observed differences in kidney function reserve capacity between age groups, we tested whether there were determinants, other than disease severity (i.e. measured GFR

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