You are on page 1of 19

REVIEWS

New treatment paradigms for ADPKD:


moving towards precision medicine
Matthew B. Lanktree1 and Arlene B. Chapman2
Abstract | The natural history of autosomal dominant polycystic kidney disease (ADPKD) is
characterized by a variable rate of cyst development and increase in total kidney volume
(TKV), variable kidney function decline and age of onset of end-stage renal disease (ESRD), and
variable presentation of renal and extrarenal manifestations. Precision medicine is proposed
to improve patient outcomes by tailoring therapy to the specific genetic background,
pathophysiology, disease burden, prognosis and status of each individual. This approach to the
management of patients with ADPKD is nearing clinical implementation owing to advances in
genetics, imaging, biomarker development and therapeutics. In this Review, we discuss
pharmacological and non-pharmacological interventions for the treatment of hypertension
and proteinuria, and for slowing the rate of cyst growth in patients with ADPKD before the
development of ESRD. We provide recommendations for the management of renal
complications, including cyst infection, nephrolithiasis, haematuria and chronic pain.
The early treatment of patients with ADPKD who are largely asymptomatic is associated with
a therapeutic burden but slows cyst growth and delays subsequent loss of kidney function,
which ultimately delays the need for renal replacement therapy and has a positive effect on
the quality of life of patients.

Autosomal dominant polycystic kidney disease The average age at which ESRD is reached in patients
(ADPKD) is a systemic disease that affects 1 in 2,000 with ADPKD has not improved, despite improved over-
individuals and is the most common hereditary kid- all survival owing to a higher prevalence of renal trans-
ney disease1–3. ADPKD is equally represented among plantation and better ESRD care3. Our understanding of
ethnicities and is more common than sickle cell dis- the pathogenesis of cyst formation and growth has also
ease, cystic fibrosis, Down syndrome, haemophilia and increased dramatically, improving our ability to diagnose,
Huntington disease combined1. ADPKD is completely evaluate and prognosticate disease severity in patients
penetrant and is characterized by the development of with ADPKD. Data from randomized controlled trials
renal cysts and increasing total kidney volume (TKV), and observational cohort studies are beginning to affect
leading to urinary concentrating defects, hypertension, treatment decisions in an evidence-based fashion5–8.
1
Nephrology Division, polyuria, nocturia, pain, nephrolithiasis, haematuria, As such, the goal of disease management for patients
Department of Medicine, infections and progressive loss of kidney function1. with ADPKD has shifted towards creating meaningful
McMaster University, St However, symptoms at disease presentation and the changes in clinical outcomes, and the potential to provide
Joseph’s Healthcare Hamilton, disease course are both highly variable. Patients with ­personalized care strategies to delay the onset of ESRD.
50 Charlton Avenue E,
Hamilton, Ontario L8N 4A6,
ADPKD, on average, progress to end-stage renal dis- Precision medicine entails a ‘new taxonomy’ for
Canada. ease (ESRD) by the age of 60 years, with 70% of patients disease that includes traditional signs, symptoms and
2
Department of Medicine, requiring renal replacement therapy (RRT) by the age clinical manifestations, but adds genetic information
Section of Nephrology, of 70 years3. In Europe, 1 in 10 patients with ESRD has in conjunction with the findings of advanced imaging
University of Chicago, 5841 S
ADPKD3, and 1 in 20 patients with ESRD in the USA, techniques and biomarker analyses, and incorporates the
Maryland Avenue, W503,
Chicago, Illinois 60637, USA. Australia and New Zealand has ADPKD2,4. Reducing experience and expectations of the individual patient9.
Correspondence to A.B.C.
the incidence of complications related to cyst burden Using this approach, patients who are unlikely to benefit
achapman1@ (for example, hypertension, pain, haematuria, infection from therapy can be spared an unnecessary therapeutic
medicine.bsd.uchicago.edu and nephrolithiasis) or delaying progressive loss of kid- burden (such as adverse effects, psychological effects and
doi:10.1038/nrneph.2017.127 ney function or the onset of ESRD provide significant financial costs) of ineffective, intolerable or unneeded
Published online 9 Oct 2017 improvements in quality of life. interventions. Owing to advances in our understanding

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 1

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REVIEWS

Key points organisms suggests that metabolic reprogramming takes


place in ADPKD, with increased utilization of aerobic
• Clinicians are now able to identify which patients with autosomal dominant glycolysis and downregulation of oxidative phosphory­
polycystic kidney disease (ADPKD) are at highest risk of progression and most likely lation14. Supporting a central role of arginine vasopressin
to benefit from early therapy (AVP) in disease pathogenesis, the offspring of a cross
• MRI measurement of total kidney volume (TKV) is a valuable method to predict future between Pkhd1‑knockout rats — a model of autosomal
rate of increase in TKV, rate of decline in kidney function and risk of end-stage renal recessive polycystic kidney disease (ARPKD) — and the
disease (ESRD)
Avp-knockout Brattelboro rat that is incapable of pro-
• The availability of genetic testing will continue to increase and can provide a ducing AVP, have normal appearing and functioning
diagnosis in unusual or atypical cases or in young (<30 years of age) patients being
kidneys15. Administration of 1‑desamino-8‑d‑arginine
assessed for kidney donation
vasopressin (DDAVP; also known as desmopressin)
• ADPKD is typically the result of germline PKD1 or PKD2 mutations, with somatic
to these rats recapitulates the phenotype of the paren-
mutations, genetic mosaicism and modifier mutations occasionally contributing to
the ADPKD phenotype
tal Pkhd1‑knockout rat15. Additional insults, including
ischaemia–reperfusion injury, might also contribute to
• Disruption of polycystin trafficking and signalling, environmental exposures and the
compounding ‘snowball’ effects of regional ischaemia, inflammation and tubular
accelerated cystogenesis16. Finally, cyst formation and
obstruction further contribute to disease progression expansion itself is accompanied by inflammation, macro­
phage activation, ischaemia, cytokine production and
• Novel strategies intended to limit cyst burden have provided encouraging results,
whereas treatment of hypertension and proteinuria remain the mainstays of medical renal tubular obstruction, which might further promote
management of ADPKD an environment of increased cyst formation and growth
• Assessment and treatment of ADPKD-related complications, including cyst in what has been referred to as a ‘snowball’ effect17.
haemorrhage, cyst infection, nephrolithiasis and chronic pain, require special
consideration and attention Phenotypic variability of ADPKD
The phenotype of ADPKD can be characterized using
clinical, biochemical, imaging, environmental exposure
of the progression and the genetics of ADPKD and their and biomarker data18,19 (FIG. 2). Cysts vary in size, distri-
contribution to the variability in ADPKD presenta- bution and growth rate between patients, with resulting
tion, the opportunity to tailor therapy to the individual variations in the rate of TKV increase20. The rate of TKV
patient is approaching. increase is the sum of the volumes of newly formed cysts
In this Review, we summarize the evidence for opti- and the growth of existing cysts20. Without intervention,
mizing the management of patients with ADPKD. We the rate of TKV increase remains remarkably constant
focus on the care of patients before kidney transplan- in an individual, yielding an exponential increase in
tation or dialysis, with the ultimate goal of delaying the TKV20,21. In individuals with ADPKD, kidney function
onset of ESRD. We discuss therapies and their targets for is typically in the normal range (estimated glomerular
the management of hypertension and proteinuria, treat- filtration rate (eGFR) >90 ml/min/1.73 m2) throughout
ments aimed at reducing cyst burden, management of the first three decades of life. Decreases in eGFR to ­levels
acute and chronic complications of ADPKD, including of established renal insufficiency or stage 3 chronic kid-
pain, nephro­lithiasis, cyst infection and haemorrhage, ney disease (CKD) occur long after the accumulation
and d ­ ietary recommendations for salt, protein and of a substantial cyst burden (FIG. 3). Importantly, a cli-
water intake. nician might recognize a downward trend in the eGFR
of an individual patient while serum creatinine level
Pathophysiology of ADPKD and eGFR remain in the ‘normal’ range, which should
The manifestations of ADPKD are caused by functional raise concerns22. Diagnosis and initiation of therapies to
deficits in polycystin 1 (PC1; encoded by PKD1) or poly­ prevent progression of ADPKD should be started early,
cystin 2 (PC2; encoded by PKD2). Deficiencies in either when the renal parenchyma is intact and long before
of these proteins results in abnormal primary ciliary clinically significant impairment in kidney function is
function, leading to a cascade of altered cell signalling detected23. By shifting from a simple ADPKD diagnosis
and aberrant cellular proliferation and fluid secretion to the assessment of a continuous network of interrelated
resulting in the formation and growth of cysts10,11 (FIG. 1). quantitative traits, clinical heterogeneity can be quanti-
A number of factors might contribute to the pathogenesis fied, disease status can be characterized and phenotypes
and phenotypic variation of ADPKD: haploinsufficiency, can be measured that reflect the underlying molecular
defined as having only a single functional copy of PKD1 and genetic defects that cause ADPKD18,19.
or PKD2, owing to either heterozygous loss‑of‑function
mutations or hypomorphic (reduced function) alleles; Clinical evaluation of ADPKD. ADPKD can have a wide
somatic mutations; altered gene expression; disruption range of clinical presentations: severe cases are diag-
of the trafficking and post-translational modification of nosed in utero or neonatally, whereas mild cases can go
PC1 or PC2; genetic variation in downstream signalling unrecognized until 60–70 years of age and ESRD might
pathways and provocation by environmental factors10–13. be avoided altogether1. Collection of a detailed family
Alterations in PC1 or PC2 function result in changes history is essential for ADPKD diagnosis and to assess
in intracellular calcium and cAMP levels, and subse- the severity of ADPKD. ADPKD is typically diagnosed
quent alteration in the mechanistic target of rapamycin following an ultrasound scan that reveals numerous
(mTOR) pathway10. Evidence from cell lines and model renal cysts, kidney enlargement and liver cysts, which

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REVIEWS

Somatic mutations Impaired trafficking

Decreased production Altered post-translational


• Haploinsufficiency Deficiency of PC1 or PC2 modification
• Hypomorphic alleles (e.g. glycosylation)

Loss of intracellular inhibitory signalling


↓Ca2+, ↑cAMP, ↑mTOR, ↓AMPK, ↑ERK, ↑JAK–STAT

↑Aerobic glycolysis Cystogenesis ↑AVP

Inflammation, ischaemia, ↑cytokines, tubular obstruction

Figure 1 | Pathogenic mechanisms and pathways in ADPKD. Loss of polycystin 1 (PC1) and/or polycystin 2 (PC2) activity
leads to measurable biochemical changes in several cellular signalling pathways, including elevated cAMP levels and
Nature Reviews | Nephrology
increased activation of mechanistic target of rapamycin (mTOR) complex 1, extracellular signal-regulated kinase (ERK),
and Janus kinase–signal transducer and activator of transcription (JAK–STAT) signalling pathways with reductions in
intracellular calcium levels and 5ʹ‑AMP-activated protein kinase (AMPK) activation. These alterations lead to subsequent
cyst formation, cellular proliferation and metabolic dysregulation, including alterations in the level of arginine vasopressin
(AVP). Each of the above mechanisms has been observed with loss of PC1 or PC2 function, although the relative
contribution of each mechanism probably varies from patient to patient.

are associated with haematuria, abdominal pain and typically have the most severe disease, with the largest
early-onset hypertension. Alternatively, ADPKD can TKV and earliest-onset ESRD, followed by patients with
be diagnosed in the absence of symptoms in individu- PKD1 non-truncating missense mutations, patients
als with a family history of ADPKD, or coincidentally with PKD2 mutations and, finally, patients in whom no
during medical evaluation of unrelated issues. Renal PKD1 or PKD2 mutation is detected (NMD)26. Further
complications, such as hypertension or proteinuria, cyst stratification of disease severity according to muta-
pain, infection or gross haematuria, especially occurring tion strength based upon the predicted effect of mis-
before 35 years of age, are predictors of increased disease sense mutations can provide additional information27.
severity and are the major components of the retrospec- Approximately 15% of patients with ADPKD present
tively developed predicting renal outcomes in ADPKD without a family history of disease, and in such patients
(PROPKD) prognostic model24,25. All patients should be the disease is most often caused by de novo mutations in
asked about symptoms, including discomfort, nocturia, PKD1 or PKD2, whereas somatic PKD1 or PKD2 muta-
polydipsia, dyspareunia or early satiety, as they might not tions are occasionally responsible28,29. Genetic results are
volunteer such details, despite the possibility of a substan- incorporated into the prognostic PROPKD model, which
tial adverse effect on quality of life. Information on num- contains data collected from over 1,000 patients, includ-
ber of pregnancies, complications during pregnancy and ing retrospectively collected data on clinical features,
previous episodes of acute kidney injury (AKI) should albeit without renal imaging or quantification of TKV24.
also be sought25. Environmental exposures, including A high-risk PROPKD score has a positive predictive value
caffeine intake, smoking and low birth weight, are often of 81% for the prediction of ESRD by 60 years of age24 .
difficult to quantify and lack rigorous evidence of a Importantly, substantial heterogeneity in ADPKD pres-
considerable role in ADPKD, but should be clarified25. entation exists, including age of diagnosis, rate of eGFR
Dietary patterns are important in the management of decline, and extrarenal manifestations, even among fam-
patients with ADPKD and intake of sodium, protein and ily members carrying the same mutation, suggesting that
fluids should be evaluated and reviewed with the patient. additional genetic or environmental factors contribute to
disease severity26,30. Mutations in a third gene, GANAB
Genetic testing for ADPKD. Patients with ADPKD can (encoding glucosidase II subunit‑α (GIIα)), have been
have mutations in either PKD1 or PKD2, which account identified in a small subset of patients with ADPKD
for disease in approximately 78% and 13% of all patients, (<2%), who typically have mild renal cyst involvement
respectively10. Significant allelic heterogeneity exists in but moderate-­to‑severe liver cysts12,29,31. GANAB was rec-
both PKD1 and PKD2, with >1,200 and >200 pathogenic ognized as an ideal candidate for a role in cysto­genesis, as
mutations reported, respectively (ADPKD Mutation mutations in PRKCSH (encoding glucosidase II s­ ubunit-β
Database). Patients with truncating PKD1 mutations (GIIβ)) cause polycystic liver disease (PLD)12.

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 3

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REVIEWS

Brain MRI imaging


Buccal saliva sample Presence and size of aneurysm
DNA studies including whole-genome, whole- Cerebral or arachnoid cysts
exome or gene panel-targeted resequencing aneurysm

Cardiac MRI and echocardiography


Blood sample Valvular function, left ventricular
Genomic, epigenomic, transcriptomic mass index and ejection fraction
and metabolomic analysis
Heart with LVH or valvulopathy
Arteries
Cystic liver Automated and ambulatory
blood pressure monitoring
Cyst fluid sample
Genomic, transcriptomic, proteomic,
metabolomic and microRNA analysis of Liver MRI and ultrasonography imaging
single cells, the cell-free fraction and Cyst number and distribution, total
exosomes liver volume, total liver cyst volume

Cystic kidney
Kidney MRI and ultrasonography
Urine sample imaging
Genomic, transcriptomic, proteomic, Cyst number and distribution, total kidney
peptidomic, metabolomic and microRNA volume, cyst volume, parenchymal volume
analysis of single cells, the cell-free fraction
and exosomes Bladder

Figure 2 | Sources of data to characterize the ADPKD clinical phenotype. Beyond vital information obtained from
analyses of a patient’s medical and family history, molecular genetic and imaging techniques Nature
can be used to further
Reviews | Nephrology
characterize the current disease status and prognosis of a patient. Other modalities that can provide clinically relevant
information include blood-pressure monitoring and analyses of bodily fluids for the presence of specific biomarkers.
LVH, left ventricular hypertrophy.

Other hereditary renal cystic conditions exist, for experienced clinician, but obtaining a molecular genetic
which mutations in over 100 additional genes have diagnosis can support identification and classification in
been identified (TABLE 1). Autosomal dominant cystic some patients.
diseases for which genetic mutations have been identi- With the cost of next-generation sequencing
fied include: polycystic liver disease (PLD) with or with- (NGS) falling, targeted gene-panel, whole-exome and
out renal cysts (mutations in PRKCSH, SEC63, LRP5, whole-genome sequencing to identify the cause of
GANAB, ALG8 and SEC61B); tuberous sclerosis com- hereditary diseases is becoming increasingly accessible
plex (TSC; mutations in TSC1 and TSC2), which also for clinical diagnostics and will most likely contribute
includes a contiguous deletion syndrome involving both to a precision medicine approach. Targeted candidate
TSC2 and PKD1, which accounts for approximately 2% gene panels enable the sequencing of selected groups
of patients with ADPKD; syndrome of renal cysts and of genes (usually 10–50), which enables increased
diabetes (RCAD; mutations in HNF1B); von Hippel– read depth and reduces the per-patient costs, whereas
Lindau syndrome (mutations in VHL); and medullary whole-exome sequencing involves the entire cod-
cystic disorders associated with autosomal dominant ing region of the genome. Owing to the presence of
tubulointerstitial kidney disease (ADTKD; mutations sequence identity with six pseudogenes, the first half
in UMOD, MUC1 and REN). The remaining cystic gene of PKD1 has been poorly sequenced following cap-
disorders are rare, severe syndromic ciliopathies with ture for gene-panel or exome strategies, but is an area
autosomal recessive patterns of inheritance or spontan­ of active research interest33. Locus-specific long-range
eous occurrence, which account for a small percentage PCR is required to reliably and accurately sequence this
of patients with cystic kidney disease32. The clinical region of PKD1.34 Whole-genome sequencing does not
presentation, or phenotype, of patients with non-AD- require sequence capture and enables the pseudogene
PKD cystic kidney diseases can be recognized by the issue to be overcome using long paired-end reads, but

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REVIEWS

a b c
Rate of TKV increase TKV eGFR
6 4,000 120

Increase in TKV/year (%)

eGFR (ml/min/1.73 m2
5 100
3,000
4 80

TKV (ml)
3 2,000 60
2 40
1,000
1 20
0 0 0
0 20 40 60 80 0 20 40 60 80 0 20 40 60 80
Age (years) Age (years) Age (years)

1% 3% 5%

Figure 3 | Theoretical relationship between total kidney volume (TKV) and kidney function. Rate of TKV increase
Nature Reviews | Nephrology
(part a), measured TKV (part b) and estimated glomerular filtration rate (eGFR; part c) over time in patients with either mild
(blue line, defined as a 1% increase in TKV per year), moderate (red line, defined as a 3% increase in TKV per year) or severe
(green line, defined as 5% TKV growth per year) autosomal dominant polycystic kidney disease (ADPKD). The model
assumes a TKV of 400 ml at 20 years of age, with a baseline eGFR of 120 ml/min/1.73 m2 and a calculated
eGFRt = eGFRt-1 – (TKVt/400). eGFRt is the eGFR at time t in years45.

remains technically challenging35. Detection of dele- ADPKD risk stratification and monitoring with TKV.
tions that can range from a few bases to many exons Ultrasonography is the screening method of choice
can be difficult and often requires multiplex ligation-­ for ADPKD, especially in those with a family history
dependent probe amplification (MLPA) or quantitative of ADPKD and who are >40 years of age42. However,
PCR. However, the detection of deletions will improve TKV (adjusted for height and age), as measured using
with the use of NGS techniques35. NGS will also enable MRI, provides a more precise evaluation of disease sta-
the detection and quantification of somatic mutations tus, prognosis and longitudinal risk assessment21,43,44.
that create mosaicism in blood, urine and pathologi- The average increase in TKV in patients with ADPKD
cal specimens and that are responsible for a small per- is 5–7% per year, but ranges from little or no increase
centage of ADPKD cases29. In addition to a main effect to a >20% increase per year45. Measurement of TKV
PKD1 or PKD2 mutation, modifier effect heterozygous enables the prediction of slope of eGFR decline, and
mutations in additional cystic kidney disease genes, therefore the age of onset of ESRD5,46,47. Increased TKV
as described above (such as HNF1A and LRP5), have is also associated with renal complications including
been identified as potential contributors to the pheno­ hyper­tension, albuminuria, nephrolithiasis, renal cyst
type, representing digenic inheritance36,37. Families infections, haematuria and worsening abdominal pain46.
with compound hetero­zygous or bilineal hypo­morphic Evaluation of the progression of ADPKD via sequen-
PKD1 alleles 38 and uniparental disomy of mutated tial TKV measurements has been utilized as a primary
PKD2 alleles39 have also been identified with severe, outcome in m ­ ultiple randomized controlled trials6,7,48–57.
early-onset phenotypes. As a further example of the Automated segmentation methods that enable cyst
evolving genetic complexity of ADPKD, homo­zygous burden to be calculated using the sum of the kidney area
or compound heterozygous mutations in PKHD1, in each MRI slice have been developed, and favourable
which encodes fibrocystin, cause autosomal recessive comparisons with methods such as MRI-based mid-
polycystic kidney disease (ARPKD), whereas hetero­ slice area and ellipsoid equation volume (ellipsoid vol-
zygous PKHD1 mutation carriers can present with iso- ume = ∏/6 × length × width × depth), ultrasound-based
lated PLD12,40. The modifier effect of common alleles or kidney length and volume, and manual segmentation
rare NGS-identified hypomorphic mutations in other methods support their use for risk stratification, particu-
­cystogenic genes requires further investigation. larly in patients with smaller kidneys, but are not yet
In our opinion, clinical genetic testing for ADPKD widely implemented44,58. Nevertheless, ellipsoid-based
is currently best utilized in patients with ADPKD who measures of TKV using MRI seem to be sufficient and
have atypical presentations, including early-onset dis- clinically applicable for risk assessment owing to their
ease, in the context of variable presentations among fam- ease of use, speed of use and availability59.
ily members, or in those patients with a lack of family The Mayo clinic ADPKD classification scheme, which
history of ADPKD10,41 or, alternatively, young (<30 years was developed to enable the identification of individuals
of age) at‑risk individuals with no evidence of cysts on with ADPKD who have intact kidney function for inclu-
imaging but who require a definite diagnosis, including sion in clinical trials, classifies patients as having typi-
potential kidney donors from known ADPKD families. cal disease (class 1; bilateral and symmetrical disease,
Genetic testing might also be helpful for pre-­conception found in >95% of patients) or atypical disease (class 2;
counselling. With the availability of additional geno- uni­lateral, segmental, or asymmetrical disease, found
type–phenotype information, the implementation of in <5% of patients)21. Patients with class 1 disease are
genetic testing for molecular diagnosis of ADPKD is sub­divided into five risk strata: A (low risk) through
likely to increase. to E (high risk), after adjustment for height and age21.

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 5

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REVIEWS

This schema does not include kidney function, gender stratum (1C) and no reduction in the lowest risk stra-
or ethnicity in its formulae. Both height-adjusted TKV tum (1A) over 10 years of follow‑up. The Mayo risk
and the Mayo ADPKD classification can be simply classification (class 1A to 1E) remained fairly stable over
determined from measurements of kidney sagittal and time for the majority of patients; however, as many as
coronal length, and axial width and depth. In the Mayo 20% of patients shifted up or down one stratum over
and the Consortium for Radiologic Imaging Studies of 10 years of follow‑up21. The rate of TKV increase meas-
Polycystic Kidney Disease (CRISP) cohorts, patients in ured following serial imaging, as opposed to estimation
the highest risk stratum (1E) averaged a decline in eGFR from a single scan, might also yield an improvement in
of nearly 5 ml/min/1.73 m2 per year, compared with a risk stratification and requires further study45. As out-
decline of 2.5 ml/min/1.73 m2 per year in the middle lined in a review published in 201645, the use of TKV

Table 1 | Cystic kidney diseases


Disease AD/ Gene Typical Characteristics Prevalence
AR decade of
diagnosis;
ESRD
Autosomal dominant AD PKD1 Childhood−30s; • Early age of cyst onset, hypertension, ESRD, innumerable • ~78% of ADPKD
polycystic kidney 50s small renal cortical cysts • ~1 in 2,000
disease (ADPKD), type 1

Autosomal dominant AD PKD2, 30s; 70s • Late age of cyst onset, hypertension, ESRD, fewer, larger • ~13% of ADPKD
polycystic kidney GANAB renal cysts and liver cysts very common • ~1 in 7,000
disease, type 2
Autosomal recessive AR PKHD1 Birth; early • Neonatal death in 30% of patients, Potter phenotype, ~1 in 20,000
polycystic kidney childhood oligohydramnios
disease • Renal symptoms include early renal failure and severe
polycystic kidneys
• Biliary symptoms include dysgenesis, portal hypertension
and cholangitis
Renal cysts and AD HNF1B, TCF2 Teens; 15% • Renal cysts, diabetes, hypomagnesaemia, hyperuricaemia Unknown
diabetes syndrome ESRD in teens and genital tract malformations; also known as
mature-onset diabetes of youth 5 (MODY5)
Polycystic liver disease AD PRKCSH, 30s; NA • Massive liver volume expansion with numerous cysts, liver • Isolated: ~1 in
SEC63, LRP5, cysts were traditionally considered entirely isolated from 100,000
GANAB, polycystic kidneys, but phenotypic overlap with ADPKD • Liver cysts in up
ALG8, now recognized to 90% of patients
SEC61B with ADPKD
Tuberous sclerosis AD TSC1, TSC2 Childhood; 40s • Dermatomal symptoms include facial angiofibroma, ~1 in 10,000
shagreen patch and hypomelanotic macules
• Cerebral symptoms include cortical tubers, subependymal
giant cell astrocytoma, seizures and developmental delay
• Renal symptoms include polycystic kidneys and
angiomyolipoma
• Pulmonary symptoms include lymphangioleiomyomatosis
• Ophthalmic symptoms include retinal hamartomas
TSC2–PKD1 contiguous AD TSC2–PKD1 Childhood; 20s • Similar presentation to that of tuberous sclerosis with Rare
deletion syndrome more severe renal phenotype
Von Hippel–Lindau AD VHL 20s; NA • Neuroendocrine tumours of the pancreas, ~1 in 36,000
disease pheochromocytoma, renal cell carcinoma, serous
cystadenomas, cerebellar and spinal haemangioblastoma,
retinal angioma
Autosomal dominant AD UMOD, Teens−20s; • Slowly progressive kidney disease, medullary cysts, Unknown
tubulointerstitial kidney MUC1, REN 40s−60s hyperuricaemia and gout in ADTKD–MUC1; also known as
disease medullary cystic kidney disease
Medullary sponge NA Unknown; 30s; NA • Medullary nephrocalcinosis, nephrolithiasis ~1 in 5,000
kidney familial
clustering
Nephronophthisis AR >90 genes Childhood; • Paediatric-onset tubulointerstitial nephritis, large range Rare
teenage years of rare monogenic multisystem conditions (including
Joubert, Senior–Loken, Bardet–Biedl and Meckel–Gruber
syndromes)
Acquired cystic disease NA Unknown Variable • Associated with chronic renal failure, ESRD Common, up to 50%
• Normal or small-sized kidneys might have 1 cyst
AD, autosomal dominant; AR, autosomal recessive; ESRD, end-stage renal disease; NA, not applicable.

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REVIEWS

as a surrogate outcome to evaluate the effectiveness of might also yield new insights into disease patho­genesis,
a therapy or to monitor disease progression has been a but they require further evaluation and ­validation before
source of controversy. TKV is the strongest prognostic application in clinical practice.
term in predictive modelling of eGFR decline and pro-
gression to ESRD, and has been approved by both the Cumulative risk stratification. As mentioned above,
FDA and the European Medicines Agency (EMA) for use the PROPKD score24 was developed from retrospective
as a biomarker for clinical trial enrichment, but TKV has clinical data to predict the onset of ESRD at <60 years of
not been approved as a primary end point for use in ran- age, and includes information on the main mutated gene
domized controlled trials60. Nevertheless, in our opinion, (PKD1, PKD2 or NMD), as well as other clinical factors
TKV is a useful prognostic tool for predicting the future (sex, history of hypertension and/or urological adverse
rate of decline in kidney function, is a sufficient surrogate events, such as haematuria, cyst infection or nephrolithi­
outcome for evaluating the effectiveness of interventions asis, before 35 years of age). However, this score does
in randomized controlled trials and is a useful method to not take into account the age, serum creatinine level or
evaluate the extent of longitudinal disease progression. the TKV of patients24. The Mayo Clinic ADPKD classi-
fication24 was developed for risk stratification in patients
Serum and urinary biomarkers, metabolomics, with ADPKD to optimize enrolment in randomized con-
­ roteomics and exosomes. Predictive biomarkers enable
p trolled trials, and includes TKV, with adjustments for age
the quantitative identification of patients with greatest and height, but does not include other aspects such as
disease activity and the highest risk of disease progres- genotype, serum creatinine level or other clinical vari­
sion, and the potential differentiation of patients who ables. Neither the PROPKD nor the Mayo classification
might respond to therapy from those who would not. include current eGFR or the trajectory of previous eGFR
Ideally, biomarkers should not only associate with eGFR loss, which has been incorporated into the European
or TKV cross-sectionally, but should also be predictive Renal Association–European Dialysis and Transplant
of longitudinal changes in TKV and/or decline in eGFR. Association (ERA–EDTA) recommendations for identi­
Evaluation of a biomarker in interventional trials can fying patients with high-risk disease for initiation of tol-
implicate a causal role of a biomarker in disease patho- vaptan treatment22. Ultimately, selection of treatments
genesis. The clinical value of over a dozen biomarkers for the management of ADPKD will be personalized and
as prognostic indicators for the severity and progression will likely incorporate clinical, biochemical and genomic
of ADPKD has been evaluated (TABLE 2). The severity of factors in the context of current disease status, increase
ADPKD is associated with an increasingly impaired in TKV to date and prediction of eGFR decline and onset
urine-concentrating ability and lower urinary osmo- of ESRD (FIG. 4).
lality; therefore, maximum urine-­concentrating ability
after water deprivation has been used to assess disease Management of hypertension
severity61–63. Copeptin, which is the stable portion of In patients with ADPKD, hypertension has an aver-
the precursor of AVP, is an easily measured surrogate age age of onset of 31 years, before the emergence of a
indicator of AVP production that can be measured in measurable reduction in eGFR in 60% of patients with
urine or serum. Higher AVP levels are associated with ADPKD5,49,74,75. The presence, age of onset, and severity
a worse renal prognosis in patients with ADPKD61,63–68. of hypertension is associated with increased TKV5,21,74,
Mass spectrometry analysis of urinary peptides impli- and is a risk factor for younger age at onset of ESRD24,76.
cates proteins that are known to reside in cystic fluid For every 100 ml increase in TKV, the relative risk of
(such as apolipoprotein A1, antithrombin III, fibrinogen development of hypertension increases by 1.4‑fold77.
and α1 antitrypsin), as well as novel peptides that require Additionally, patients with ADPKD are at an increased
further study in ADPKD (such as collagen degradation risk of both left ventricular hypertrophy and adverse
products, cathepsin and matrix metalloproteinases)69. ­cardiovascular events78–80.
Exosomes, which are vesicles that are released from the An increase in cyst burden or TKV is thought to
plasma membrane of cells and are present in all bodily result in bilateral intrarenal ischaemia and activation
fluids, contain cellular proteins, RNA and metabolites, of the renin–angiotensin–aldosterone system (RAAS),
and are thought to have a role in cell‑to‑cell communi- resulting in vasoconstriction and sodium retention,
cation and transport70. The contents of urinary exosomes worsening hypertension and potential acceleration of
can be identified, thus providing information about cyst growth74. Patients with ADPKD and hypertension
the cells from which the exosomes originated71. Using have elevated levels of plasma renin activity and serum
this approach, reduced levels of PC1 and PC2 have aldosterone concentrations compared with individu-
been detected in urinary exosomes from patients with als with essential hypertension81. Importantly, ciliary
ADPKD compared with those without ADPKD72, and an defects in endothelial cells and vascular smooth muscle
overabundance of desmosome components (periplakin cells lead to altered intracellular calcium concentrations,
and envoplakin), villin 1 and complement proteins have elevated cAMP levels and increased cellular prolifera-
been identified in the urinary exosomes of patients tion, thus creating a primary vasculopathy82. Additional
with ADPKD70. Using a machine-learning classifier, mechanisms that contribute to the hypertensive state
the effects of multiple biomarkers were combined into a include endothelial dysfunction, reduced nitric oxide
single, more-powerful test69,73. Biomarkers could poten- levels, increased levels of reactive oxygen species and
tially have an important role in risk stratification, and activation of the sympathetic nervous system74,83.

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 7

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REVIEWS

Table 2 | Emerging serum and urinary biomarkers in ADPKD not yet in clinical practice
Biomarker Source Features Effect in ADPKD
Loss of polycystin
PC1, PC272 Urinary Reduced expression of PC1 and PC2 Expression reduced by 50% in exosomes
exosomes
Water handling
Osmolality61,63,100 Urine, serum Quantify loss of urine concentrating ability Maximum urine concentration reduced by 15%
Copeptin 61,63–68
Serum, urine Portion of AVP precursor peptide, a Higher copeptin concentration associates with higher
surrogate marker of AVP TKV and decline in eGFR
Apelin177,178 Serum Inverse relationship with AVP, also 39% lower level in non-progressors
associated with fibrosis
Metabolic
HDL‑C, apolipoprotein Serum, urine Anti-atherogenic, reverse cholesterol Lower HDL‑C associates with greater TKV and
A169,100 peptide transport, anti-inflammatory increased eGFR decline
Uric acid179,180 Serum, urine Reduced filtration, increased tubular Hyperuricaemia is associated with eGFR decline
reabsorption, endothelial dysfunction,
hypertension
Klotho, FGF23181–183 Serum Low Klotho appears to prevent phosphouric Higher FGF23, lower Klotho levels early in ADPKD
effect of high FGF23 progression
Inflammatory
MCP1184,185 Urine, blood Chemokine that regulates monocyte Higher MCP1 associated with greater TKV and decline
activity in eGFR
Complement (C3, C5, C4B, Urine Activation of complement cascade >2‑fold higher in urinary exosomes of patients with
C9, CfB, C1q)70 exosomes ADPKD than in patients with CKD
IL‑18185 Urine Pro-inflammatory cytokine Increased level in patients with ADPKD, but not
associated with TKV or eGFR
Proliferation
HB-EGF186 Blood, urine Growth, migration and proliferation of >2‑fold higher in patients with ADPKD than in
tubular cells patients with CKD, also associated with increased TKV
and lower eGFR
sFRP4187 Blood, urine Antagonist of canonical WNT signalling Predictive of eGFR decline, limited association with
TKV
Tubular injury
KIM1184 Urine Ectodomain of KIM1 is shed after tubular Increased, also associated with increased TKV and
injury eGFR decline
NGAL185 Urine Part of innate immunity, elevated in acute Increased, but not associated with TKV or eGFR
kidney injury decline
NAG184 Urine Proximal tubular lysosomal enzyme, marker Increased, also associated with increased TKV and
of tubular injury eGFR decline
H-FABP184,186 Urine Distal tubular cytosolic protein released Increased, also associated with eGFR decline, and a
with injury loss of renal blood flow
Cellular morphology
Villin 170 Urine Actin-modifying protein that affects >2‑fold higher in patients with ADPKD than in
exosomes morphology, motility patients with CKD
Periplakin, envoplakin70 Urine Part of desmosome needed for gap >2‑fold higher level in patients with ADPKD than in
exosomes junctions and cell polarity patients with CKD
Unknown
Peptides derived from Urine Pathway analysis-derived hypothesized Part of machine learning peptidomic classifier
collagen, antithrombin III, α1 peptides functions
antitrypsin and cathepsin69,73
ADPKD, autosomal dominant polycystic kidney disease; AVP, arginine vasopressin; CKD, chronic kidney disease; eGFR, estimated glomerular filtration rate; FGF23,
fibroblast growth factor 23; HB‑EGF, heparin-binding epidermal growth factor; HDL‑C, high density lipoprotein cholesterol; H-FABP, heart-type fatty acid-binding
protein; IL‑18, interleukin 18; KIM1, kidney injury molecule 1; MCP1, monocyte chemoattractant protein 1; NAG, N‑acetyl-β‑D glucosaminidase; NGAL, neutrophil
gelatinase-associated lipocalin; PC1, polycystin 1; sFRP4, secreted frizzle-related protein 4; TKV, total kidney volume.

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REVIEWS

Treatment of hypertension in patients with ADPKD


should include nonpharmacological interventions, such
ESRD@ 50 years old

(% increase in TKV/year)
as weight reduction, exercise and dietary salt restriction. 5

Rate of progression
Exclusion of additional secondary causes of hypertension, ESRD@ 70
such as primary hyperaldosteronism, pheochromo­cytoma years old
3
or obstructive sleep apnoea, should all be considered with
clinical suspicion of their existence84,85. Inhibition of the
RAAS using angiotensin-converting enzyme (ACE) inhib- 1
itors is favoured, as well as the use of angiotensin-­receptor
blockers (ARBs) and β‑blockers83. The findings of early
studies with small cohorts of patients demonstrated that
Patient 1 Patient 2 Patient 3 Patient 4
ACE inhibitors reduce the severity of proteinuria and
left ventricular mass compared to diuretics and calcium
channel blockers86, whereas ARBs resulted in a greater Main genetic effect: Somatic genetic
reduction in proteinuria than calcium channel blockers87, mutation of PKD1 vs changes, epigenetic
PKD2; missense vs modifications
and data from two small-cohort studies suggest equipoise truncating mutations
between β‑blockers and ACE inhibitors with respect to Environmental effects:
Other genetic effects hypertension,
blood-pressure control, proteinuria, left ventricular mass (common or rare): nephrolithiasis, acute
index and decline in eGFR88,89. Given RAAS activation, genes involved in kidney injury, drugs,
stimulation of vasopressin release with dehydration, and cystic, signalling and trauma, vasopressin,
the potential untoward effects of AKI, concern regard- trafficking pathways; tubular obstruction,
ing the use of diuretics given concomitantly with RAAS genes altering capsular pressure,
metabolic risk factors blood flow
blockers, particularly ACE inhibitors, should be consid-
ered, though not supported by direct evidence83,90. The
findings of the HALT–PKD studies showed that most Figure 4 | The theoretical contribution of genetic and
Nature Reviews
environmental factors to polycystin protein| Nephrology
dosage
patients can safely achieve rigorous blood pressure control
(<110/75 mmHg) with an ACE inhibitor as mono­therapy explains the rate of cyst progression in patients with
and d ­ iuretics as second-line agent, without evidence autosomal dominant polycystic kidney disease
(ADPKD). Bars represent the relative loss of functional
of worsening kidney function, AKI or a major risk of
polycystin protein dosage and contribution of genetic
adverse events6,51. and environmental factors to subsequent cyst growth and
The most recent guidelines from Kidney Disease: increase in total kidney volume (TKV). The upper
Improving Global Outcomes (KDIGO) on the control of horizontal orange line indicates a growth rate consistent
blood pressure in patients with CKD suggest a target with reaching end-stage renal disease (ESRD) at 50 years
of ≤140/90 mmHg for patients without albuminuria and of age, whereas the lower horizontal red line indicates a
≤130/80 mmHg for those with albuminuria of ≥30 mg growth rate consistent with ESRD at 70 years of age.
per day91. The randomized, controlled, HALT–PKD Patient 1 has a missense mutation in PKD2 (encoding
trial included 558 patients with ADPKD <50 years of polycystin 2), which has less severe consequences for
disease progression than other mutations, but has had
age with an eGFR >60 ml/min/1.73 m2, and revealed a
environmental exposures that increase the rate of cyst
reduced rate of increase in TKV (5.6% versus 6.6% per
growth. Patients 2 and 3 have similar main genetic effects,
year; P = 0.006), a decrease in urinary albumin excretion a missense mutation in PKD1, but patient 2 has also had a
(−3.77% versus 2.43%; P <0.001), and a reduced left-­ ‘second-hit’ somatic mutation, whereas patient 3 also has
ventricular mass index (−1.17 g/m2 versus −0.57 g/m2 other genetic modifiers. Patient 4 has a nonsense mutation
per year; P <0.001), but no overall difference in the rate in PKD1, resulting in early-onset ESRD.
of eGFR decline between aggressive (<110/75 mmHg)
and standard (<130/80 mmHg) blood pressure targets
over 5 years of follow‑up6. As expected from haemo­ The HALT–PKD study B randomized controlled trial
dynamic effects, patients with aggressive blood pres- included 486 patients with eGFR of 25–60 ml/min/1.73 m2
sure-reduction targets had an initial decline in eGFR in who were randomly assigned to receive either ACE
the first 4 months of study compared to those on stand- inhibitors or ACE inhibitors plus ARBs, but did not
ard blood pressure control (−3.1 ml/min/1.73 m2 versus evaluate blood pressure targets directly, as a target of
0.5 ml/min/1.73  m2; P <0.001), but less decline in eGFR <130/80 mmHg was set for both groups; a similar decline in
over the remainder of the study (−2.7 ml/min/1.73 m2 eGFR (−3.91 ml/min/1.73 m2 versus −3.87 ml/min/1.73 m2
per year versus −3.1 ml/min/1.73 m2 per year; P = 0.05), per year; P = 0.86) was observed in the two groups51. An
resulting in no significant overall difference in eGFR over earlier study, the two-year Modification of Diet in Renal
5 years of follow‑up (−2.9 ml/min/1.73 m2 per year versus Disease (MDRD) study involving patients with moder-
−3.0 ml/min/1.73 m2 per year; P = 0.55)6. At the time the ate to advanced CKD (eGFR 25–55 ml/min/1.73 m2),
HALT–PKD study was designed, the Mayo Clinic classi- which included 220 patients with ADPKD, revealed
fication of risk stratification had not been developed. In a no significant difference in eGFR (inulin clearance;
post hoc analysis of the HALT–PKD study, patients with −6.0 ml/min/1.73 m 2 versus −5.7 ml/min/1.73 m 2
a higher baseline TKV (class 1C, 1D or 1E) had the greatest per year, P = not significant) between use of aggres-
level of benefit from blood pressure reduction (P = 0.001)6. sive (mean arterial pressure (MAP) ≤92 mmHg) and

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 9

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REVIEWS

standard (MAP ≤107 mmHg) blood p ­ ressure targets, and CRISP cohort provided longitudinal evidence for associ­
potential harm of aggressive blood pressure targets in ations between elevated albumin-to‑creatinine ratio
patients with eGFR <24 ml/min/1.73 m2 (eGFR decline (ACR), increased TKV and decreased eGFR100. Baseline
−4.9 ml/min/1.73 m2 per year versus −3.9 ml/min/1.73 m2 characteristics in the HALT–PKD and TEMPO 3:4 trials
per year, P = 0.03)92. Importantly, these rates of decline are demonstrated statistically significant independent associ-
significantly greater than those observed in the similar ations between urinary albumin excretion, hypertension,
HALT–PKD study B trial, and following the conclusion of higher TKV and lower eGFR97,101. In the early HALT–
the MDRD study, long-term follow‑up of the participants PKD (study A) trial, aggressive blood pressure lowering
revealed a potential benefit of aggressive blood pressure (<110/75 mmHg) was associated with a 3.77% reduction
reduction in those with protein-to‑­creatinine ratios >0.22 in ACR compared with a 2.43% increase in the standard
and a reduced risk of mortality once ESRD was reached93,94. blood pressure target group (<130/80 mmHg; P <0.001)6.
The SPRINT trial has led to a re‑­evaluation of blood-­ In the TEMPO 3:4 trial, baseline ACR was also associ-
pressure targets in patients >50 years of age with a high ated with a more rapid eGFR reduction over the course
level of cardiovascular risk, including those with CKD but of the study (P <0.0001), and tolvaptan treatment was
without diabetes, although this trial specifically excluded associated with a 0.4 mg/mmol reduction in ACR com-
patients with ADPKD95. The findings of the SPRINT trial pared with a 0.23 mg/mmol increase in the placebo group
revealed fewer cardiovascular adverse events and reduced (P <0.001), independent of changes in blood pressure or
­levels of mortality, with no difference in renal outcomes concomitant use of ACE inhibitors97. Direct evidence that
in patients with CKD, when targeting a systolic blood reducing albuminuria in ADPKD affects progression to
pressure of <120 mmHg compared to <140 mmHg95,96. ESRD is not currently available, but we believe that an
Overall, these data suggest, either directly (HALT–PKD elevated ACR is a risk factor for progressive disease and
study A) or through inference (MDRD and SPRINT), that should be included in the decision to target lower blood
a lower, rather than a moderate blood pressure reduction pressures and increase the dose of RAAS inhibitors.
target is advantageous in patients with ADPKD who
have hypertension. Slowing the increase in cyst burden
We encourage home blood pressure monitoring and Ten distinct types of medication have been or are cur-
utilization of an automated digital blood pressure monitor rently being tested in randomized controlled trials, with
for both improved patient ease and satisfaction. A sched- the goal of reducing the rate of increase in TKV or cyst
ule of at least 10 measurements over a 3 month period burden over time and maintaining eGFR (TABLE 3).
during drug trough periods, including both weekday
and weekend measurements, is considered an inform- Vasopressin V2 receptor antagonists. Tolvaptan is a
ative approach to managing patients that also engages highly selective vasopressin V2 receptor antagonist
them in self-care. For clinic-based blood pressure meas- that was assessed for ADPKD in the TEMPO 3:4 trial7.
urements, we recommend the use of three automated In a total of 1,445 patients with estimated creatinine
measurements after a 5 minute rest in a seated position95. clearance >60 ml/min and TKV >750 ml, tolvaptan
Stringent control of dietary sodium intake is advised for decreased the rate of TKV increase by almost 40%
the management of hypertension, as well as for reduction (2.8% versus 5.5% per year; P <0.001), and slowed the
of cyst growth, as outlined below. In our opinion, blood rate of decline in eGFR (2.72 ml/min/1.73 m2 versus
pressure targets need to be tailored to the needs of each 3.70 ml/min/1.73 m2 per year; P <0.001) compared with
patient based on their age, extent of ADPKD progression, placebo after 3 years of follow‑up. Patients who received
prognosis, current eGFR, comorbid­ities and tolerance of tolvaptan had fewer complications, including clinically
treatment. Given the greater lifetime cardiovascular risk significant renal pain requiring hospitalization or nar-
of patients with ADPKD and the potential benefit of cotic intervention (27% versus 35%), urinary tract infec-
low blood pressure targets in slowing the progression tions (8.3% versus 12.6%) and episodes of haematuria
of renal disease that was demonstrated in the HALT– (7.8% versus 14.1%), than patients who received placebo.
PKD (study A) trial, we target blood pressures below However, increased thirst (55.3% versus 20.5%), poly­
110/75 mmHg in patients <50 years of age with preserved uria (38.3% versus 17.2%) and polydipsia (10.4% versus
kidney function. The remaining patients are treated in 3.5%) were more frequent in patients in the tolvaptan
accordance with the KDIGO guidelines91. group than in those in the placebo group. An alanine
aminotransferase level >2.5‑fold above the upper normal
Management of proteinuria levels was also more frequently observed in patients who
The KDIGO guidelines recommend the annual assess- received tolvaptan than in patients who received placebo
ment of albuminuria and proteinuria in all patients with (4.9% versus 1.2%), which resolved following drug dis-
CKD in order to evaluate disease severity, progression continuation. Post-hoc analyses demonstrated that tol-
and prognosis91. Among patients with ADPKD with a vaptan had simi—lar beneficial effects in patients with
creatinine clearance >60 ml/min that were enrolled in stage 1–3 CKD102, that patients with the greatest decline
the TEMPO3:4 trial, >50% had moderately increased in urine osmolality displayed the greatest therapeutic
albuminuria (3–30 mg/mmol or 30–300 mg/g)7,91,97. benefit103, that tolvaptan treatment resulted in a reduc-
Glomerular-range proteinuria (>1 mg per day) is tion in ACR independent of RAAS blockade and blood
rarely seen in patients with ADPKD and an alternative pressure97 and that tolvaptan reduced acute kidney pain
glomerular-­based diagnosis should be sought98,99. The events in patients with CKD of any stage104.

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REVIEWS

TEMPO 4:4 was a 2‑year open-label observational pleiotropic beneficial effects in patients with ADPKD,
extension study involving patients from the TEMPO 3:4 albeit via mechanisms that are currently poorly under-
cohort. This study compared the outcomes of an ‘early-­ stood, including enhanced renal blood flow, reduced
treatment’ group comprising patients from the tolvaptan inflammation and increased vascular and glomerular
group of TEMPO 3:4 with a ‘delayed-treatment’ group nitric oxide production48. Data from small short-term
comprising patients from the placebo group of TEMPO studies examining surrogate outcomes suggest that
3:4 who started on tolvaptan treatment following the treatment with statins early in the course of ADPKD
conclusion of the original trial8. The primary outcome could potentially lead to improvements in inflamma-
was negative (29.9% increase in TKV in the patients in tory biomarkers, renal plasma flow and endothelial
the early-treated group versus 31.6% among patients cell dysfunction106–108. The findings of a 3‑year ran-
in the delayed-treatment group from TEMPO 3:4 base- domized controlled trial involving 110 children and
line to TEMPO 4:4 closure; P = 0.38). After excluding young adults with ADPKD (mean age of 16 years),
Japanese participants, who were undertaking a sepa- indicate that pravastatin significantly reduces the rate
rate follow‑up extension trial, and those who dropped of TKV increase, after correction for age, gender and
out of TEMPO 3:4, 91.9% of patients who were eligible blood pressure, compared with placebo (23% ± 3%
for TEMPO 4:4 enrolled in this study (60.3% of total versus 31% ± 3% over 3 years; P = 0.02)48. By contrast,
TEMPO 3:4 enrolment). Interpretation of the study a 2‑year open-label trial involving 49 patients with
results is complicated by prognostic imbalance result- ADPKD (mean eGFR 54 ml/min/1.73 m2) identified
ing from loss of the ran­domization, with the delayed-­ no difference in rate of eGFR decline or proteinuria
treatment group including more men, and more patients with pravastatin treatment compared with placebo109.
with higher TKV and ACR at the start of TEMPO 4:4. Furthermore, a post hoc propensity score-based analysis
Owing to the onset of new aquaretic adverse effects in of the effectiveness of statins as a co‑intervention in par-
TEMPO 4:4 when switching from placebo to tolvaptan, ticipants in the HALT–PKD trial found no evidence for
the delayed-treatment group had twice the dropout benefit of pravastatin treatment in early stage (study A)
rate of the early-treatment group (early-treated n = 50 or late stage (study B) ADPKD110. These observations
(9.0%) versus delayed-treatment n = 58 (18.5%)), which suggest that early intervention with statin therapy might
is consistent with the expected incidence of first-time be bene­ficial in patients with ADPKD, but additional
aquaretic adverse effects when starting tolvaptan treat- evidence is required before widespread prescription of
ment. Nevertheless, the results of TEMPO 4:4 showed statins to patients with ADPKD. However, given the low
sustained beneficial effects of tolvaptan on eGFR decline level of risk and the likelihood of beneficial vascular
(a difference of 3.15 ml/min/1.73 m2 between patients effects, we have a low threshold for initiation of statin
in the early-treated versus delayed-treatment groups; therapy in patients with ADPKD who have either a high
P <0.001) and after correction for covariates, beneficial risk of disease progression or an elevated cardiovascu-
effects on TKV (−4.15% between the early versus delayed lar risk, after appropriate counselling, especially in the
treatment groups; P = 0.04). Moreover, no differences in context of dyslipidaemia.
safety profile were observed, and patients with the great-
est level of risk (Mayo class 1C−1E, or those with PKD1 Somatostatin analogues, mTOR inhibitors and others.
truncating mutations) had the greatest level of benefit in Somatostatin analogues, including octreotide, lanreotide
TEMPO 4:4, similar to the findings of TEMPO 3:4.7,102 and pasireotide, have demonstrated promise as treat-
Tolvaptan is now approved for the treatment of ments for ADPKD, especially in patients with associated
ADPKD in Japan, Canada, South Korea, Switzerland PLD. A systematic review of three small randomized
and Europe, and the ERA–EDTA and Canadian guide- controlled trials investigating the efficacy of somato­
lines for the use of this agent have been published22,41. statin analogues in patients with ADPKD or PLD showed
Current recommendations for use are based upon the a 9% reduction in TKV increase over 3 years (95% CI
inclusion criteria of TEMPO 3:4, and on identifying −10.33 to −7.58; P <0.001; total n = 136), with no change
patients with high-risk disease (18–50 years of age, in eGFR52,54,111,112. Total liver volumes also decreased by
eGFR >45 ml/min/1.73 m2, with rapid decline in eGFR 5% per year54,112,113. Concerns regarding somatostatin
or Mayo class 1C−1E)22,41. FDA approval of tolvaptan analogue-associated hepatic cyst infections currently
for patients with ADPKD is currently awaiting results exist, as an interim analysis of the ongoing DIPAK trial
of the upcoming REPRISE clinical trial105. A precision reported eight hepatic cyst infections in seven patients
medicine strategy might help to identify patients who over 342 patient-years of lanreotide use, compared with
are appropriate for therapy by enabling the selection of no cyst infections in patients receiving placebo114,115. Until
patients with maintained renal function but who are the results of further studies are available, somatostatin
at risk of disease progression, as demonstrated by risk analogues are not recommended for use in patients with
­evaluation and stratification outlined above. ADPKD without substantial hepatic involvement as the
primary indication.
Statins. Statins have a role in reducing cardiovascular The efficacy of inhibitors of mTOR complex 1
risk in patients with CKD who are not on dialysis and (mTORC1), including sirolimus and everolimus, has
are either >50 years of age or have a level of 10‑year been tested in five randomized controlled trials involv-
cardiovascular risk exceeding 10%91. Supported by ing patients with ADPKD. Despite a strong mechanistic
animal studies, statins have been proposed to have foundation and impressive results in model organisms,

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 11

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REVIEWS

Table 3 | Randomized controlled trials testing therapies for reducing kidney cyst burden
Therapy Trial name, lead Sample Effect on eGFR or TKV*
author or trial no. size (n)
Vasopressin V2 receptor antagonists
Tolvaptan TEMPO7 1,445 • eGFR: −2.61 versus −3.81 mg/ml per year (P <0.001)
• TKV growth: 2.8% versus 5.5% per year (P <0.001)
REPRISE105 1,337 • Not yet reported
ACE inhibitors and/or ARBs
Lisinopril +  HALT-PKD6,51 Early: 558 • eGFR: −2.9 versus −3.0 ml/min/1.73 m2 per year (P = 0.55) with
telmesartan versus low BP target or combination of ACE inhibitor and ARB
lisinopril alone • TKV increase: 5.6% per year in low BP target versus 6.6% per
year in standard BP target (P = 0.006). No differences with
combination of ACE inhibitor and ARB
Late: 486 • No difference in TKV or eGFR
Somatostatin analogues
Octreotide Hogan et al.54,113 34 • iGFR: −5.1 versus −7.2 ml/min in 1 year (P = 0.6)
• TKV increase: 0.3% versus 8.6% per year (P = 0.05)
Ruggenenti et al.57 12 • eGFR: −2.5 versus −3.2 ml/min/1.73 m2 (P = NS)
• TKV increase: 2.2% versus 5.9% in 6 months (P <0.05)
ALADIN52 75 • eGFR: −3.9 versus −5.0 ml/min/1.73 m2 in 3 years (P = NS)
• TKV increase: 46.2 ml versus 143.7 ml per year (P = 0.03)
Lanreotide van Keimpema 54 • Creatinine: −2 versus 4 μmol/l (P = NS)
et al.112 • TKV increase: –1.5% versus 3.4% per 24 weeks (P = 0.02)
DIPAK114,115 309 • Phase III, ongoing, not yet reported
Pasireotide NCT01670110 48 • Phase II, ongoing
mTORC1 inhibitors
Everolimus Walz et al.50 433 • eGFR: −8.9 versus −7.7 ml/min/1.73 m2 at 2 years (P = 0.15)
• TKV increase at 1 year: 102 ml versus 157 ml (P = 0.02)
• TKV increase at 2 years: 230 ml versus 301 ml (P = 0.06)
Sirolimus RAPYD53 55 • Ramipril versus ramipril + high dose versus ramipril + low dose
• GFR: −2.8 versus 4.5 versus 0.8 at 2 years (P = NS)
SUISSE49 100 • eGFR: 0.2 versus −3.5 ml/min/1.73 m2 (P = 0.07)
• TKV increase: 36 versus 15 versus 14 ml at 2 years (P = NS)
Braun et al.55 30 • Low-dose versus high-dose versus standard care in 1 year
• iGFR: 7.7 versus 1.6 versus −11.2 ml/min/1.73 m2 (P <0.01)
• TKV increase: 99 cm3 versus 97 cm3 at 18 months (P = 0.26)
SIRENA-II188 40 • TKV increase: 9.3% versus 4.8% versus 7.8% (P = 0.29)
• Stopped owing to safety concerns
Statin
Pravastatin Cadnapaphornchai 110 • Creatinine clearance: 126 versus 126 ml/min (P = 0.98)
et al.48 • >20% height-corrected TKV increase: 46% versus 68%
(P = 0.03)
Fassett et al.109 49 • eGFR: −1.34 versus −0.31 ml/min/1.73 m2 at 2 years (P = 0.78)
• TKV: not assessed
Tyrosine kinase inhibitors
Bosutinib NCT01233869 172 • Ongoing, phase II
Tesevatinib NCT03096080 18 • Ongoing, phase I
(KD019)
Other approaches
Niacinamide NCT02140814 10 • Completed, not yet reported
(Vitamin B3)
NCT02558595 36 • Ongoing, phase II
Metformin NCT02656017 96 • Ongoing, phase II
NCT02903511 50 • Ongoing, phase II

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REVIEWS

Table 3 (cont.) | Randomized controlled trials testing therapies for reducing kidney cyst burden
Therapy Trial name, lead Sample Effect on eGFR or TKV*
author or trial no. size (n)
Other approaches (cont.)
PPARγ agonist: NCT02697617 28 • Ongoing, phase II
pioglitazone
Aldosterone NCT01853553 60 • Completed, not yet reported
antagonist:
spironolactone
Water intake NCT00784030 13 • Completed, not yet reported
NCT02776241 32 • Recruiting
NCT03102632 60 • Recruiting
*Comparisons are treatment versus placebo unless otherwise noted. ACE, angiotensin-converting enzyme; ARB, angiotensin 2
receptor blocker; BP, blood pressure; eGFR, estimated glomerular filtration rate; iGFR, 125I-iothalamate GFR; mTORC1, mechanistic
target of rapamycin complex 1; NS, not significant; PPARγ, peroxisome proliferator-activated receptor‑γ; TKV, total kidney volume.

the findings of a systematic review demonstrated no haemorrhage, infection, nephrolithiasis or an under-


beneficial effects of mTORC1 inhibition on the rate lying malignancy. Cyst haemorrhage can be asymp-
of change of TKV or the rate of decline in eGFR111. tomatic and occur with or without gross haematuria,
Adverse effect profiles, including stomatitis, podo­cyto­ fever and/or pain120,121. Haemorrhage might be the
pathy, protein­uria and renal toxicity, as well as wide var- result of trauma, and is a common cause of presenta-
iations in doses used in different studies, which either tion in affected children and adolescents following
were subtherapeutic or resulted in high levels of adverse contact sports or falls. Isolated haematuria is usually
effects, make full interpretation of the potential benefits self-­limiting and resolves within a week in the absence
of mTORC1 inhibition difficult. The lack of efficacy of of specific treatment122. Unenhanced CT should reveal
these inhibitors has been hypothesized to reflect lower increased density (>25 Hounsfield units) and an irregu-
cyst-specific drug concentrations in patients, compared lar haematoma and wall thickening within the haemor-
to the concentrations that are effective in preventing rhagic cyst123. Administration of ACE inhibitors should
ADPKD progression in mouse models116. Nonetheless, be delayed until bleeding resolves, owing to a risk
mTORC1 inhibitors are not currently recommended for of AKI124. The risks and benefits of delaying the use of
the treatment of patients with ADPKD. anticoagulant or antiplatelet therapies should be deter-
Several randomized controlled trials involving inhib- mined based upon the specific indication. Patients with
itors that specifically target the proliferative nature of non-resolving haemorrhages might develop substantial
ADPKD, based on growing insights into cyst biology subcapsular or retroperitoneal haematomas or require
from cellular and animal studies, are currently o ­ ngoing. blood transfusions, haemostatic agents, interventional
These agents include the tyrosine-kinase inhib­itors embolization, or surgical procedures123.
bosutinib and tesevatinib, as well as metformin, piogli-
tazone, spironolactone and niacinamide. Finally, oligo- Cyst infections. The presence of numerous cysts can
nucleotides targeting microRNAs are a new therapeutic create areas of non-laminar tubular flow, and pockets
strategy that could be employed in the treatment of of urinary stasis and obstruction can serve as a nidus
ADPKD. An oligonucleotide directed against miR‑17 for infection, or act as an infectious reservoir. Cysts
is currently moving through preclinical studies and an can become disconnected from tubules and thus, when
oligo­nucleotide directed against miR‑21, which was infected, effectively become an abscess. Both ascending
tested in a phase I trial for Alport syndrome, could be urinary and haematogenous spread have been identified
helpful in the treatment of ADPKD117,118. as sources of kidney cyst infections120. In addition, liver
cysts can also become infected. Diagnosing kidney and
Managing renal complications in ADPKD liver cyst infections can be difficult and should be sus-
Patients with ADPKD can be fairly asymptomatic, especi­ pected in any patient with ADPKD who has fever and/or
ally early in the course of disease, but chronic vague abdominal pain, especially those with elevated serum
abdominal discomfort, early satiety, polyuria and poly- white blood cell counts and C‑reactive protein levels,
dipsia remain common. Acute adverse events, such as and in the absence of an alternative diagnosis120,125. The
gross haematuria, infections, acute pain syndromes and identification of infected cyst aspirate remains the gold
nephrolithiasis, often require medical attention and lead standard for diagnosis, but aspiration is uncommon in
to increased medical resource utilization and ­hospital practice owing to the invasive nature of the procedure,
visits compared with that of the general population119. the risk of complications and the difficulty in identifying
and accessing the infected cyst126. Urine and blood cul-
Haematuria and cyst haemorrhage. The initial manage- tures should be obtained, as they might guide the selec-
ment of haematuria is reliant on identifying the under- tion of antibiotics, but their usefulness is limited as >50%
lying aetiology, which can include cyst rupture and/or of such cultures are sterile, including those from patients

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 13

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REVIEWS

with positive cyst-aspirate cultures126. Ultrasonography abnormalities that predispose to urinary crystal formation
and non-contrast CT are of limited utility, and although include low urinary pH, hypocitraturia, hypomagnesuria,
contrast-enhanced CT might be helpful, its use is limi­ hyperuricaemia and hyperuricosuria, hyperoxaluria
ted by concerns about the use of contrast in those with and, occasionally, hypercalciuria and secondary hyper-
impaired kidney function126. Advanced imaging studies, parathyroidism132,133. Compared with patients with
including MRI and fluorodeoxyglucose positron emis- ADPKD with no previous nephrolithi­asis, patients with
sion tomography/CT (FDG–PET/CT), seem better stone-forming ADPKD have a greater number of cysts
approaches for the diagnosis of cyst infections, but are and an increased cyst size and TKV, and lower eGFR and
limited by costs and availability126–129. 24 h urine volumes132,134. Phenotypic overlap or coexist-
The frequency of kidney and liver cyst infections in ence with medullary sponge kidney can occur in patients
patients with ADPKD is difficult to estimate, but such with ADPKD133. Uric acid stones are the most commonly
infections are not a common event. In a 10‑year (2005– reported stone composition in patients with ADPKD,
2015) single-centre retrospective analysis including 173 although calcium-containing and struvite stones both
patients with ADPKD, 16 patients had a total of 23 cyst also occur frequently133. Ultrasonography might enable
infections120. In a meta-analysis, investigators collected the detection of larger stones, although altered anatomy
a published case series describing a total of 85 patients and the presence of parenchymal and cyst wall calcifi-
with ADPKD that had renal cyst infections130. Cyst cations can reduce the sensitivity and specificity of this
infections appear more frequently in older patients with imaging technique, which makes non-contrast CT the
later stage disease (mean age of 51 years; 79% of patients preferred modality for diagnosis132.
had eGFR <60 ml/min/1.73 m2, and 40% of patients had Urological treatments of patients with ADPKD and
ESRD)130. Persistent infections that are resistant to symptomatic stones might include percutaneous nephro­
antibiotic eradication were reported in 61% of patients lithotomy135, extracorporeal shockwave lithotripsy or
following initial treatment, and were more common uretero­scopy with laser lithotripsy136. Increased fluid
in patients with large (>5 cm in diameter) cysts, when intake is the mainstay of stone prevention, which is help-
urolithiasis and atypical pathogens (organisms other ful for patients with ADPKD, regardless of the presence
than Escherichia coli, such as Staphylococcus aureus, of stones. Screening for concomitant hypercalcaemia and
Enterococcus faecium, Clostridium perfringens and hyperparathyroidism, and standard dietary counselling
Candida krusei, among others) were present, and when for stone prevention, including high fluid intake, high
shorter (<2‑week) durations of empiric treatment were fruit and vegetable intake, and a low-salt, protein and
used130. Positive cyst aspirates were obtained in half of all oxalate containing diet, is advisable137. Correcting meta­
patients and E. coli was the most common organism in bolic abnormalities by prescribing potassium citrate,
cyst aspirates (in 51% of patients)130. Fluoroquinolones sodium bicarbonate or vitamin D, where appropriate
and trimethoprim-sulfamethoxazole are generally the is also a reasonable approach. Thiazide diuretics might
preferred first-line treatments of cyst infections owing be useful for recurrent nephrolithiasis in the context
to their lipid solubility and therefore increased likeli- of hypercalciuria138.
hood of cyst penetration131. In the meta-analysis, the
average duration of antibiotic treatment was 14 days130, Pain. Acute abdominal pain secondary to cyst rupture,
and given the abscessed nature of cyst infections, treat- cyst infection or the presence of kidney stones should be
ment without drainage required a longer duration of treated with analgesics, including acetaminophen and
antibiotic therapy131. Without ­invasive cyst fluid col- opioids. The judicious use of short courses of NSAIDs
lection, the selection of an appropriate antimicrobial might also be considered in patients with an eGFR in the
agent remains empirical, and the risk of antimicrobial normal range139. Patients with ADPKD might also have
resistance must be considered when patients do not chronic pain, which has been attributed to the compres-
recuperate on an appropriate trajectory. Evidence to sion of adjacent structures, capsular distension, traction
guide the timing of advanced imaging studies is scant, of the renal pedicle and aberrant activation of the sen-
but, in our opinion, such studies should be sought in sory or autonomic nervous system121. Pain sensations
patients with refractory symptoms that do not improve in these patients can be either sharp and stabbing or
after 1 week of empirically selected cyst-penetrating dull and aching, and fluctuating in intensity, and might
treatment. Even when antimicrobial therapy is under- affect a wide dermatomal distribution121. The high level
taken, invasive procedures are typically required when of variability in symptoms at presentation makes a diag-
cyst diameters exceed 5 cm, and were required in a sig- nosis difficult, especially as patients with ADPKD are
nificant proportion of patients included in the above- not precluded from having chronic pain of other aetiol-
mentioned meta-analysis (percutaneous cyst drainage ogies, including fibromyalgia, gynaecological pain and
(31%); surgical drainage (8%); and partial (1%) and inflammatory or irritable bowel disease, among others.
total (33%) nephrectomy)130. Pain is more common in the later stages of ADPKD, but
is not proportional to TKV at early disease stages (eGFR
Nephrolithiasis. Metabolic and structural abnormal- >60 ml/min/1.73 m2)104,140.
ities predispose patients with ADPKD to nephrolithi­ The management of chronic pain begins with use
asis, and reports suggest that up to a quarter of patients of nonpharmacological interventions, including phys-
with ADPKD will have a kidney stone at some point ical interventions (for example, massage, physical
in their disease course132. ADPKD-related metabolic therapy, ice and heating pads) and psycho-educational

14 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REVIEWS

interventions, although the effectiveness of these inter- and can be considered following a failure of minimally
ventions has not been specifically studied in patients invasive strategies; however, such interventions do not
with ADPKD141. Further interventions can be divided improve blood pressure control and might be associated
into pharmacological and procedural modalities, and with worsening renal function144,152,153. Partial, or total
are best delivered by a multidisciplinary team including nephrectomy should be considered as an absolute last
nephrologists, urologists, interventional radiologists and resort, in order to avoid the loss of any residual renal
pain physicians. function. No gold-standard modality for pain relief
Pharmacological analgesia should be prescribed exists in patients with ADPKD; therefore, the selection
according to the WHO three-step analgesic ladder, of treatment options is at least partially dictated by local
which was originally developed for cancer-related expertise and practice patterns.
pain142. The steps of the ladder include: firstly, aceta-
minophen; secondly, NSAIDs, with the caveat of lim- Dietary counselling
iting their use in patients with impaired renal function, Patients with ADPKD are often motivated to comply
or mild opioids (such as tramadol); and thirdly, strong with therapeutic recommendations, and questions
opioids (such as morphine or hydromorphone)142. regarding dietary recommendations are frequently
Patients must be counselled about the possible substan- among the first asked by patients. Modifications to diet
tial adverse effects of opioids, which include constipa- are difficult to adopt and maintain — in consequence,
tion, nausea, sedation and a risk of dependence and/or most patients regress to their baseline dietary pattern,
overdose142. Impaired clearance of opioids in patients driven by intrinsic taste, thirst and habit100. However,
with renal dysfunction is a problem. Furthermore, con- data from clinical trials have demonstrated that major,
cerns have been raised about the excessive use of opioids sustainable dietary alterations are possible with repeated
for chronic non-cancer-related pain, and the presence use of counselling interventions154–156.
of unrecognized harms that can occur with their pre-
scription143. Adjuvant analgesics that are intended for Salt restriction. Dietary sodium intake is best evaluated
patients with neuropathic pain, including pregabalin using 24 h urine collection157. Despite heated contro-
or gabapentin, and antidepressants, including amitrip- versy, and the possible presence of a J‑shaped relationship
tyline, nortriptyline or duloxetine, have been proposed between dietary sodium intake and both CKD and cardio­
for the treatment of pain in patients with ADPKD, vascular disease risk, most clinicians agree that patients
but have not been studied in this context121,141,144. Patients should avoid a high-sodium diet (>5 g per day)158–163. The
have anecdotally reported beneficial effects of using current KDIGO recommendation is a daily intake tar-
medicinal marijuana145, but its use has not been studied get of <2 g of sodium (corresponding to <5 g of sodium
or reported in the formal literature. In the TEMPO 3:4 chlor­ide or table salt) in all patients with CKD without
trial, patients receiving tolvaptan had fewer kidney pain contraindications, especially in those with hypertension
events than those in the placebo group (10.1% versus or proteinuria91. The results of two randomized controlled
16.7%; P <0.001). The number needed to treat in order trials involving patients with diabetic and non-diabetic
to prevent any pain event was 35, and to prevent a severe CKD suggest that increased sodium intake reduces the
event requiring clinician-prescribed intervention with effectiveness of RAAS blockade164,165. Salt avoidance is
narcotics or hospitalization was 94.104 important in patients with ADPKD owing to a high preva­
Following the failure of conservative and pharma- lence of salt-sensitive hypertension and a requirement for
cological strategies, the stepwise use of minimally inva- optimal RAAS blockade. The CRISP trial provided lon-
sive, followed by more-invasive procedures has been gitudinal evidence of an association between higher uri-
proposed141,146, and has been evaluated in a 44‑patient nary sodium excretion, which is a surrogate indicator of
prospective cohort study139. The underlying theory is dietary sodium intake, and both an increased rate of TKV
that pain originating from pressure on adjacent organs increase and a fall in GFR100.
is transmitted via the coeliac plexus and the major The best evidence for the efficacy of reducing sodium
splanchnic nerves, whereas pain from renal capsule intake in ADPKD comes from a post-hoc analysis of
pressure is transmitted via the aorticorenal plexus and data from the early (study A) and late (study B) HALT–
the minor and least splanchnic nerves139,141,147. A diagnos- PKD trials6,51,155. Study A revealed a significant associ-
tic coeliac or splanchnic nerve block with short-­acting ation between each 18 mEq (414 mg) urinary sodium
local anaesthetic can be used to indicate the source of excretion per 24 h increase and increased TKV (0.43%
pain141. Renal denervation, either surgically or using per year, P <0.001), and a nonsignificant fall in eGFR
percutaneous transluminal catheter-based radioablation, (−0.07 ml/min/1.73 m2; P = 0.094), whereas in Study B,
might help to relieve renal-capsule-related pain148–150. each 18 mEq (414 mg) urinary sodium excretion per 24 h
Sympathetic nerve ablation is the same procedure as increase was associated with more-rapid eGFR decline
that used in the negative SYMPLICITY-HTN trials for (−0.09 ml/min/1.73 m2; P <0.001) and an increased risk
refractory hypertension151. Minimally invasive liver of the composite outcome (a 50% reduction in eGFR, and
or kidney cyst aspiration, with sclerotherapy or fenes- ESRD or death, HR 1.08; P = 0.01)155. The lack of a sig-
tration, especially for large cysts (>5 cm in diameter), nificant association between sodium intake and eGFR in
is also a potential treatment option141. Laparoscopic study A was attributed to the slow decline in eGFR
or surgical interventions aimed at cyst decortication or in patients with early stage ADPKD and the inclusion of
deroofing have been reported to improve pain outcomes patients with mild disease who are unlikely to experience

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 15

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REVIEWS

a decline in eGFR during the study period155. At enrol- can result in failed free water excretion and subsequent
ment, study participants were counselled to reduce their hyponatraemia62. Additionally, individuals with severely
sodium intake below 100 mEq per day (2.3 g per day), reduced salt and protein intake, or those using diuretics
and modest reductions in intake were achieved over the might be at an increased risk of hyponatraemia62.
course of the study (baseline mean intake was 178 mEq The findings of observational studies including a total
per day (4.1 g of sodium per day), mean reduction was of 18 patients with ADPKD demonstrated the safety and
12 mEq per day (276 mg per day) in study A and 26 mEq tolerability of a water prescription, as well as a reduction
per day (598 mg per day) in study B)155. in urine osmolality and decreased circulating AVP con-
Studies of the effect of dietary protein restriction in centrations174,175. A 1-year observational study failed to
patients with ADPKD are lacking, but the KDIGO guide- reveal any differences in TKV or eGFR slopes between
lines recommend reducing protein intake to 0.8 g/kg per those with high (2.6 l per day) and those with free (1.4 l
day for adults with an eGFR <30 ml/min/1.73 m2 and per day) patterns of water intake in 34 patients with
avoiding high protein intake (>1.3 g/kg per day) in all ADPKD176. Other randomized controlled trials investi-
patients with CKD who are at risk of disease progres- gating the efficacy of water intake as a prescription in
sion91. Dietary protein intake can be estimated from patients with ADPKD are currently underway (TABLE 2).
measurements of 24 h urinary urea nitrate excretion166. In patients with eGFR >30 ml/min/1.73 m2 who are
Excessive protein intake is thought to result in glomer- not using diuretics, do not have a condition or take a
ular hyperfiltration and the accumulation of nitrogen­ drug that alters AVP (that is, a possible contributor to
ous wastes, especially in the context of reduced GFR167. the syndrome of inappropriate antidiuretic hormone),
In  North America, high-protein, low-­carbohydrate and have no urological abnormalities, we prescribe an
diets are common168, especially with the popularity of increase in free water intake targeting a urinary osmo-
diets such as the ‘South Beach diet’ and the ‘Atkins diet’169. lality of <250 mosm/kg H20. This prescription typically
Despite equivocal results of the MDRD study, which only consists of a daily water intake of >3 l per day173,175.
included patients with ADPKD170, a meta-analysis of
data from 10 randomized controlled trials investigating Conclusions
restriction of protein intake in patients with non-diabetic ADPKD can be a devastating disease that leads to consid-
CKD, about a quarter of whom had ADPKD, supports erable morbidity and mortality in affected families. Owing
the restriction of dietary protein171. The number needed to the marked variability in the severity and progression
to treat for 1 year to prevent one renal death ranged from of ADPKD, a detailed personalized evaluation can assist
as few as two, and up to 56.171 in differentiating between patients who can be reassured
We employ 24 h urine collections in conjunction with regarding their mild disease and those who require closer
multidisciplinary education, food records and motiva- monitoring and early therapeutic intervention. Genetic
tional interviewing to encourage patients to reduce their testing has an important role in diagnosis and precon-
salt and protein intake in accordance with the KDIGO ception counselling in patients with ADPKD, especially
guidelines. We advocate a balanced diet of complex car- in complex or unusual cases of ADPKD or involving
bohydrates, fruits and vegetables, with a low sodium young (<30 years old) patients. MRI-based measurement
(<2.3 mg per day) and modest protein (<0.8 g/kg per of TKV is a powerful imaging biomarker that enables
day) content. accurate ADPKD-related risk assessment. Risk stratifi-
cation is required before the prescription of treatments
Water intake. Humans have basal levels of AVP, which targeting cyst reduction to ensure that the therapeutic
assist in urine concentration and avoid the need for burden of costs and adverse effects is outweighed by
excess fluid intake62. The renal concentrating defects in the benefits of treatment. Data from randomized con-
patients with ADPKD can lead to mild hypovolaemia, trolled trials provide evidence to direct the management
and a higher concentration of circulating AVP. Increased of hypertension and proteinuria, based upon risk strata.
AVP levels, and levels of its surrogate copeptin, are Dietary interventions, including increasing water intake
associated with progression of ADPKD and increased and limiting salt and protein intake, are likely to be bene­
TKV64,65,67,68. Fluid intake reduces the concentration of ficial and empower patients — supporting evidence for
circulating AVP via a negative feedback loop, whereas these recommendations come from post hoc analyses of
AVP concentrations increase in response to V2 recep- randomized controlled trials involving other therapeutics.
tor antagonism62,172. Given the role of AVP in the patho- The establishment of centres of excellence will enable the
genesis of ADPKD, suppression of AVP concentrations development of expertise for accurate risk assessment,
by excess water ingestion seems a reasonable treatment and the diagnosis and treatment of the complications
approach173. In individuals without kidney disease, low of ADPKD. Ultimately, continued patient participation
levels of urinary osmolality are strongly correlated with in studies is needed to generate sufficient evidence for a
low AVP activity64,65. However, owing to impaired uri- full precision medicine approach. Currently, strong evi-
nary concentrating deficits, patients with ADPKD might dence exists suggesting that early assessment and treat-
have low levels of urinary osmolality despite the pres- ment of patients with high-risk ADPKD improves the
ence of AVP, especially in those with advanced-stage outcomes of these patients. This approach is associated
disease61. Patients in whom the use of water prescription with a therapeutic burden, but seems worth the effort to
is limited include those with impaired GFR and with a slow the deteri­oration of kidney function and cyst growth,
relative excess or inappropriate AVP secretion, which ­ultimately delaying the onset of ESRD.

16 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REVIEWS

1. Gabow, P. A. Autosomal dominant polycystic kidney dominant polycystic kidney disease. J. Am. Soc. dominant polycystic kidney disease. Clin. J. Am. Soc.
disease. N. Engl. J. Med. 329, 332–342 (1993). Nephrol. 27, 942–951 (2016). Nephrol. 9, 889–896 (2014).
2. Reule, S. et al. ESRD from autosomal dominant 25. Schrier, R. W. et al. Predictors of autosomal dominant 49. Serra, A. L. et al. Sirolimus and kidney growth in
polycystic kidney disease in the United States, polycystic kidney disease progression. J. Am. Soc. autosomal dominant polycystic kidney disease.
2001–2010. Am. J. Kidney Dis. 64, 592–599 Nephrol. 25, 2399–2418 (2014). N. Engl. J. Med. 363, 820–829 (2010).
(2014). 26. Hwang, Y. H. et al. Refining genotype-phenotype 50. Walz, G. et al. Everolimus in patients with autosomal
3. Spithoven, E. M. et al. Renal replacement therapy correlation in autosomal dominant polycystic kidney dominant polycystic kidney disease. N. Engl. J. Med.
for autosomal dominant polycystic kidney disease disease. J. Am. Soc. Nephrol. 27, 1861–1868 (2016). 363, 830–840 (2010).
(ADPKD) in Europe: prevalence and survival — 27. Heyer, C. M. et al. Predicted mutation strength of 51. Torres, V. E. et al. Angiotensin blockade in late
an analysis of data from the ERA-EDTA Registry. nontruncating PKD1 mutations aids genotype- autosomal dominant polycystic kidney disease.
Nephrol. Dial. Transplant. 29, iv15–iv25 (2014). phenotype correlations in autosomal dominant N. Engl. J. Med. 371, 2267–2276 (2014).
4. Fernando, M. R., Dent, H., McDonald, S. P. & polycystic kidney disease. J. Am. Soc. Nephrol. 27, 52. Caroli, A. et al. Effect of longacting somatostatin
Rangan, G. K. Incidence and survival of end-stage 2872–2884 (2016). analogue on kidney and cyst growth in autosomal
kidney disease due to polycystic kidney disease in 28. Cornec‑Le Gall, E. et al. Type of PKD1 mutation dominant polycystic kidney disease (ALADIN):
Australia and New Zealand. Popul. Health Metr. 15, influences renal outcome in ADPKD. J. Am. Soc. a randomised, placebo-controlled, multicentre trial.
7 (2017). Nephrol. 24, 1006–1013 (2013). Lancet 382, 1485–1495 (2013).
5. Chapman, A. B. et al. Renal structure in early 29. Iliuta, I. A. et al. Polycystic kidney disease without 53. Stallone, G. et al. Rapamycin for treatment of type I
autosomal-dominant polycystic kidney disease an apparent family history. J. Am. Soc. Nephrol. 28, autosomal dominant polycystic kidney disease (RAPYD-
(ADPKD): The Consortium for Radiologic Imaging 2768–2776 (2017). study): a randomized, controlled study. Nephrol. Dial.
Studies of Polycystic Kidney Disease (CRISP) cohort. 30. Barua, M. et al. Family history of renal disease Transplant. 27, 3560–3567 (2012).
Kidney Int. 64, 1035–1045 (2003). severity predicts the mutated gene in ADPKD. 54. Hogan, M. C. et al. Randomized clinical trial of long-
6. Schrier, R. W. et al. Blood pressure in early autosomal J. Am. Soc. Nephrol. 20, 1833–1838 (2009). acting somatostatin for autosomal dominant polycystic
dominant polycystic kidney disease. N. Engl. J. Med. 31. Porath, B. et al. Mutations in GANAB, encoding the kidney and liver disease. J. Am. Soc. Nephrol. 21,
371, 2255–2266 (2014). glucosidase IIα subunit, cause autosomal-dominant 1052–1061 (2010).
7. Torres, V. E. et al. Tolvaptan in patients with autosomal polycystic kidney and liver disease. Am. J. Hum. Genet. 55. Braun, W. E., Schold, J. D., Stephany, B. R.,
dominant polycystic kidney disease. N. Engl. J. Med. 98, 1193–1207 (2016). Spirko, R. A. & Herts, B. R. Low-dose rapamycin
367, 2407–2418 (2012). 32. Braun, D. A. & Hildebrandt, F. Ciliopathies. (sirolimus) effects in autosomal dominant polycystic
8. Torres, V. E. et al. Multicenter, open-label, extension Cold Spring Harb. Perspect. Biol. 9, a028191 (2017). kidney disease: an open-label randomized controlled
trial to evaluate the long-term efficacy and safety of 33. Song, X., Haghighi, A., Iliuta, I. A. & Pei, Y. Molecular pilot study. Clin. J. Am. Soc. Nephrol. 9, 881–888
early versus delayed treatment with tolvaptan in diagnosis of autosomal dominant polycystic kidney (2014).
autosomal dominant polycystic kidney disease: the disease. Expert Rev. Mol. Diagn. 13, 1–11 (2017). 56. Perico, N. et al. Sirolimus therapy to halt the
TEMPO 4:4 Trial. Nephrol. Dial. Transplant. 34. Kinoshita, M. et al. Technical evaluation: identification progression of ADPKD. J. Am. Soc. Nephrol. 21,
http://dx.doi.org/10.1093/ndt/gfx043 (2017). of pathogenic mutations in PKD1 and PKD2 in 1031–1040 (2010).
9. Committee on A Framework for Developing a New patients with autosomal dominant polycystic kidney 57. Ruggenenti, P. et al. Safety and efficacy of long-acting
Taxonomy of Disease. Toward Precision Medicine: disease by next-generation sequencing and use of a somatostatin treatment in autosomal-dominant
Building a Knowledge Network for Biomedical comprehensive new classification system. PLoS ONE polycystic kidney disease. Kidney Int. 68, 206–216
Research and a New Taxonomy of Disease. 11, e0166288 (2016). (2005).
(The National Academies Press, 2011). 35. Mallawaarachchi, A. C. et al. Whole-genome 58. Kim, Y. et al. Automated segmentation of kidneys from
10. Harris, P. C. & Torres, V. E. Genetic mechanisms and sequencing overcomes pseudogene homology to MR images in patients with autosomal dominant
signaling pathways in autosomal dominant polycystic diagnose autosomal dominant polycystic kidney polycystic kidney disease. Clin. J. Am. Soc. Nephrol. 11,
kidney disease. J. Clin. Invest. 124, 2315–2324 disease. Eur. J. Hum. Genet. 24, 1584–1590 (2016). 576–584 (2016).
(2014). 36. Bergmann, C. et al. Mutations in multiple PKD genes 59. Spithoven, E. M. et al. Estimation of total kidney
11. Cornec‑Le Gall, E., Audrezet, M. P., Le Meur, Y., may explain early and severe polycystic kidney volume in autosomal dominant polycystic kidney
Chen, J. M. & Ferec, C. Genetics and pathogenesis disease. J. Am. Soc. Nephrol. 22, 2047–2056 (2011). disease. Am. J. Kidney Dis. 66, 792–801 (2015).
of autosomal dominant polycystic kidney disease: 37. Cnossen, W. R. et al. LRP5 variants may contribute to 60. Perrone, R. D. et al. Total kidney volume is a prognostic
20 years on. Hum. Mutat. 35, 1393–1406 (2014). ADPKD. Eur. J. Hum. Genet. 24, 237–242 (2016). biomarker of renal function decline and progression to
12. Besse, W. et al. Isolated polycystic liver disease genes 38. Rossetti, S. et al. Incompletely penetrant PKD1 alleles end-stage renal disease in patients with autosomal
define effectors of polycystin‑1 function. J. Clin. Invest. suggest a role for gene dosage in cyst initiation in dominant polycystic kidney disease. Kidney Int.Rep. 2,
127,1772–1785 (2017). polycystic kidney disease. Kidney Int. 75, 848–855 442–450 (2017).
13. Lantinga-van Leeuwen, I. S. et al. Lowering of Pkd1 (2009). 61. Casteleijn, N. F. et al. Urine and plasma osmolality in
expression is sufficient to cause polycystic kidney 39. Losekoot, M. et al. Neonatal onset autosomal patients with autosomal dominant polycystic kidney
disease. Hum. Mol. Genet. 13, 3069–3077 (2004). dominant polycystic kidney disease (ADPKD) in a disease: reliable indicators of vasopressin activity and
14. Rowe, I. et al. Defective glucose metabolism in patient homozygous for a PKD2 missense mutation disease prognosis? Am. J. Nephrol. 41, 248–256
polycystic kidney disease identifies a new therapeutic due to uniparental disomy. J. Med. Genet. 49, 37–40 (2015).
strategy. Nat. Med. 19, 488–493 (2013). (2012). 62. Torres, V. E., Bankir, L. & Grantham, J. J. A case for
15. Wang, X., Wu, Y., Ward, C. J., Harris, P. C. & 40. Ward, C. J. et al. The gene mutated in autosomal water in the treatment of polycystic kidney disease.
Torres, V. E. Vasopressin directly regulates cyst growth recessive polycystic kidney disease encodes a large, Clin. J. Am. Soc. Nephrol. 4, 1140–1150 (2009).
in polycystic kidney disease. J. Am. Soc. Nephrol. 19, receptor-like protein. Nat. Genet. 30, 259–269 63. Zittema, D. et al. Kidney function and plasma copeptin
102–108 (2008). (2002). levels in healthy kidney donors and autosomal
16. Takakura, A. et al. Renal injury is a third hit promoting 41. Soroka, S. et al. Assessing risk of disease progression dominant polycystic kidney disease patients.
rapid development of adult polycystic kidney disease. and pharmacological management of autosomal Clin. J. Am. Soc. Nephrol. 9, 1553–1562 (2014).
Hum. Mol. Genet. 18, 2523–2531 (2009). dominant polycystic kidney disease: a canadian expert 64. Boertien, W. E. et al. Relationship of copeptin, a
17. Leonhard, W. N. et al. Scattered deletion of PKD1 consensus. Can. J. Kidney Health Dis. 4, surrogate marker for arginine vasopressin, with change
in kidneys causes a cystic snowball effect and 2054358117695784 (2017). in total kidney volume and GFR decline in autosomal
recapitulates polycystic kidney disease. J. Am. Soc. 42. Pei, Y. et al. Unified criteria for ultrasonographic dominant polycystic kidney disease: results from the
Nephrol. 26, 1322–1333 (2015). diagnosis of ADPKD. J. Am. Soc. Nephrol. 20, CRISP cohort. Am. J. Kidney Dis. 61, 420–429 (2013).
18. Houle, D., Govindaraju, D. R. & Omholt, S. Phenomics: 205–212 (2009). 65. Boertien, W. E. et al. Copeptin, a surrogate marker for
the next challenge. Nat. Rev. Genet. 11, 855–866 43. Tangri, N. et al. Total kidney volume as a biomarker of vasopressin, is associated with kidney function decline
(2010). disease progression in autosomal dominant polycystic in subjects with autosomal dominant polycystic kidney
19. Lanktree, M. B., Hassell, R. G., Lahiry, P. & kidney disease. Can. J. Kidney Health Dis. 4, disease. Nephrol. Dial. Transplant. 27, 4131–4137
Hegele, R. A. Phenomics: expanding the role of clinical 2054358117693355 (2017). (2012).
evaluation in genomic studies. J. Investig. Med. 58, 44. Turco, D., Busutti, M., Mignani, R., Magistroni, R. 66. Corradi, V. et al. Copeptin levels and kidney function
700–706 (2010). & Corsi, C. Comparison of total kidney volume in ADPKD: case-control study. Clin. Nephrol. 86,
20. Grantham, J. J. et al. Determinants of renal volume quantification methods in autosomal dominant 147–153 (2016).
in autosomal-dominant polycystic kidney disease. polycystic disease for a comprehensive disease 67. Meijer, E. et al. Copeptin, a surrogate marker of
Kidney Int. 73, 108–116 (2008). assessment. Am. J. Nephrol. 45, 373–379 (2017). vasopressin, is associated with disease severity in
21. Irazabal, M. V. et al. Imaging classification of 45. Grantham, J. J. & Torres, V. E. The importance of total autosomal dominant polycystic kidney disease.
autosomal dominant polycystic kidney disease: kidney volume in evaluating progression of polycystic Clin. J. Am. Soc. Nephrol. 6, 361–368 (2011).
a simple model for selecting patients for clinical trials. kidney disease. Nat. Rev. Nephrol. 12, 667–677 68. Nakajima, A., Lu, Y., Kawano, H., Horie, S. & Muto, S.
J. Am. Soc. Nephrol. 26, 160–172 (2015). (2016). Association of arginine vasopressin surrogate marker
22. Gansevoort, R. T. et al. Recommendations for the use 46. Grantham, J. J., Chapman, A. B. & Torres, V. E. Volume urinary copeptin with severity of autosomal dominant
of tolvaptan in autosomal dominant polycystic kidney progression in autosomal dominant polycystic kidney polycystic kidney disease (ADPKD). Clin. Exp. Nephrol.
disease: a position statement on behalf of the ERA- disease: the major factor determining clinical 19, 1199–1205 (2015).
EDTA Working Groups on Inherited Kidney Disorders outcomes. Clin. J. Am. Soc. Nephrol. 1, 148–157 69. Pejchinovski, M. et al. Urine peptidome analysis
and European Renal Best Practice. Nephrol. Dial. (2006). predicts risk of end-stage renal disease and reveals
Transplant. 31, 337–348 (2016). 47. Grantham, J. J. et al. Volume progression in polycystic proteolytic pathways involved in autosomal dominant
23. Grantham, J. J. Rationale for early treatment of kidney disease. N. Engl. J. Med. 354, 2122–2130 polycystic kidney disease progression. Nephrol. Dial.
polycystic kidney disease. Pediatr. Nephrol. 30, (2006). Transplant. 32, 487–497 (2017).
1053–1062 (2015). 48. Cadnapaphornchai, M. A. et al. Effect of pravastatin 70. Salih, M. et al. Proteomics of urinary vesicles links
24. Cornec‑Le Gall, E. et al. The PROPKD score: a new on total kidney volume, left ventricular mass index, plakins and complement to polycystic kidney disease.
algorithm to predict renal survival in autosomal and microalbuminuria in pediatric autosomal J. Am. Soc. Nephrol. 27, 3079–3092 (2016).

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 17

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REVIEWS

71. Street, J. M., Koritzinsky, E. H., Glispie, D. M. & 95. Group, S. R. et al. A randomized trial of intensive 116. Novalic, Z. et al. Dose-dependent effects of sirolimus
Yuen, P. S. T. Urine exosome isolation and versus standard blood-pressure control. N. Engl. on mTOR signaling and polycystic kidney disease.
characterization. Methods Mol. Biol. 1641, 413–423 J. Med. 373, 2103–2116 (2015). J. Am. Soc. Nephrol. 23, 842–853 (2012).
(2017). 96. Ruzicka, M., Burns, K. D. & Hiremath, S. Precision 117. Hajarnis, S. et al. microRNA‑17 family promotes
72. Hogan, M. C. et al. Identification of biomarkers for medicine for hypertension management in chronic polycystic kidney disease progression through
PKD1 using urinary exosomes. J. Am. Soc. Nephrol. kidney disease: relevance of SPRINT for therapeutic modulation of mitochondrial metabolism.
26, 1661–1670 (2015). targets in nondiabetic renal disease. Can. J. Cardiol. Nat. Commun. 8, 14395 (2017).
73. Kistler, A. D. et al. Urinary proteomic biomarkers for 33, 611–618 (2017). 118. Yheskel, M. & Patel, V. Therapeutic microRNAs
diagnosis and risk stratification of autosomal dominant 97. Gansevoort, R. T. et al. Albuminuria and tolvaptan in in polycystic kidney disease. Curr. Opin. Nephrol.
polycystic kidney disease: a multicentric study. autosomal-dominant polycystic kidney disease: results Hypertens. 26, 282–289 (2017).
PLoS ONE 8, e53016 (2013). of the TEMPO 3:4 Trial. Nephrol. Dial.Transplant. 31, 119. Knight, T. et al. Medical resource utilization and costs
74. Chapman, A. B., Stepniakowski, K. & Rahbari-Oskoui, F. 1887–1894 (2016). associated with autosomal dominant polycystic kidney
Hypertension in autosomal dominant polycystic kidney 98. Akinbodewa, A. A., Adejumo, O. A., disease in the USA: a retrospective matched cohort
disease. Adv. Chron. Kidney Dis. 17, 153–163 (2010). Ogunsemoyin, A. O., Osasan, S. A. & Adefolalu, O. A. analysis of private insurer data. Clinicoecon.
75. Chapman, A. B. & Gabow, P. A. Hypertension in Co‑existing autosomal dominant polycystic kidney Outcomes Res. 7, 123–132 (2015).
autosomal dominant polycystic kidney disease. disease and nephrotic syndrome in a Nigerian patient 120. Neuville, M., Hustinx, R., Jacques, J., Krzesinski, J. M.
Kidney Int. Suppl. 61, S71–S73 (1997). with lupus nephritis. Ann. Afr. Med. 15, 83–86 & Jouret, F. Diagnostic algorithm in the management
76. Johnson, A. M. & Gabow, P. A. Identification of (2016). of acute febrile abdomen in patients with autosomal
patients with autosomal dominant polycystic kidney 99. Yenigun, E. C. et al. Coexistence of autosomal dominant polycystic kidney disease. PLoS ONE 11,
disease at highest risk for end-stage renal disease. dominant polycystic kidney disease and amyloidosis e0161277 (2016).
J. Am. Soc. Nephrol. 8, 1560–1567 (1997). in a patient with nephrotic-range proteinuria. 121. Hogan, M. C. & Norby, S. M. Evaluation and
77. Yu, A. S. et al. Trajectory of the GFR in autosomal Iran. J. Kidney Dis. 8, 243–245 (2014). management of pain in autosomal dominant polycystic
dominant polycystic kidney disease. J. Am. Soc. 100. Torres, V. E. et al. Potentially modifiable factors kidney disease. Adv. Chron. Kidney Dis. 17, e1–e16
Nephrol. 27, abstr., FR‑OR006 (2016). affecting the progression of autosomal dominant (2010).
78. Schrier, R. W. Hypertension and autosomal dominant polycystic kidney disease. Clin. J. Am. Soc. Nephrol. 6, 122. Chapman, A. B. et al. Autosomal-dominant polycystic
polycystic kidney disease. Am. J. Kidney Dis. 57, 640–647 (2011). kidney disease (ADPKD): executive summary from a
811–813 (2011). 101. Torres, V. E. et al. Analysis of baseline parameters in Kidney Disease: Improving Global Outcomes (KDIGO)
79. Ecder, T. Cardiovascular complications in autosomal the HALT polycystic kidney disease trials. Kidney Int. Controversies Conference. Kidney Int. 88, 17–27
dominant polycystic kidney disease. Curr. Hypertens. 81, 577–585 (2012). (2015).
Rev. 9, 2–11 (2013). 102. Torres, V. E. et al. Effect of tolvaptan in autosomal 123. Suwabe, T. et al. Clinical features of cyst infection and
80. Chapman, A. B. et al. Left ventricular hypertrophy in dominant polycystic kidney disease by CKD stage: hemorrhage in ADPKD: new diagnostic criteria.
autosomal dominant polycystic kidney disease. results from the TEMPO 3:4 Trial. Clin. J. Am. Soc. Clin. Exp. Nephrol. 16, 892–902 (2012).
J. Am. Soc. Nephrol. 8, 1292–1297 (1997). Nephrol. 11, 803–811 (2016). 124. Chapman, A. B., Gabow, P. A. & Schrier, R. W.
81. Chapman, A. B., Johnson, A., Gabow, P. A. & 103. Devuyst, O. et al. Urine osmolality, response to Reversible renal failure associated with angiotensin-
Schrier, R. W. The renin-angiotensin-aldosterone tolvaptan, and outcome in autosomal dominant converting enzyme inhibitors in polycystic kidney
system and autosomal dominant polycystic kidney polycystic kidney disease: results from the TEMPO 3:4 disease. Ann. Intern. Med. 115, 769–773 (1991).
disease. N. Engl. J. Med. 323, 1091–1096 (1990). Trial. J. Am. Soc. Nephrol. 28, 1592–1602 (2017). 125. Lantinga, M. A. et al. International multi-specialty
82. Kip, S. N. et al. [Ca2+]i reduction increases cellular 104. Casteleijn, N. F. et al. Tolvaptan and kidney pain in delphi survey: identification of diagnostic criteria for
proliferation and apoptosis in vascular smooth muscle patients with autosomal dominant polycystic kidney hepatic and renal cyst infection. Nephron 134,
cells: relevance to the ADPKD phenotype. Circ. Res. disease: secondary analysis from a randomized 205–214 (2016).
96, 873–880 (2005). controlled trial. Am. J. Kidney Dis. 69, 210–219 126. Lantinga, M. A., Drenth, J. P. & Gevers, T. J.
83. Rahbari-Oskoui, F., Williams, O. & Chapman, A. (2017). Diagnostic criteria in renal and hepatic cyst infection.
Mechanisms and management of hypertension in 105. Torres, V. E. et al. Rationale and design of a clinical Nephrol. Dial. Transplant. 30, 744–751 (2015).
autosomal dominant polycystic kidney disease. trial investigating tolvaptan safety and efficacy in 127. Lantinga, M. A., de Sevaux, R. G. & Drenth, J. P.
Nephrol. Dial. Transplant. 29, 2194–2201 (2014). autosomal dominant polycystic kidney disease. 18F‑FDG PET/CT during diagnosis and follow‑up of
84. Hessheimer, A. J., Vidal, O., Valentini, M. & Garcia- Am. J. Nephrol. 45, 257–266 (2017). recurrent hepatic cyst infection in autosomal dominant
Valdecasas, J. C. Pheochromocytoma as a rare cause 106. Zand, L. et al. Renal hemodynamic effects of the polycystic kidney disease. Clin. Nephrol. 84, 61–64
of arterial hypertension in a patient with autosomal HMG-CoA reductase inhibitors in autosomal dominant (2015).
dominant polycystic kidney disease: a diagnostic and polycystic kidney disease. Nephrol. Dial. Transplant. 128. Suwabe, T. et al. Intracystic magnetic resonance
therapeutic dilemma. Int. J. Surg. Case Rep. 14, 31, 1290–1295 (2016). imaging in patients with autosomal dominant
85–88 (2015). 107. Namli, S. et al. Improvement of endothelial polycystic kidney disease: features of severe cyst
85. Hoorn, E. J. et al. A case of primary aldosteronism dysfunction with simvastatin in patients with infection in a case-control study. BMC Nephrol. 17,
revealed after renal transplantation. Nat. Rev. Nephrol. autosomal dominant polycystic kidney disease. 170 (2016).
7, 55–60 (2011). Ren Fail 29, 55–59 (2007). 129. Piccoli, G. B. et al. Positron emission tomography in the
86. Schrier, R. W. ACE inhibitors, left ventricular mass and 108. van Dijk, M. A., Kamper, A. M., van Veen, S., diagnostic pathway for intracystic infection in adpkd
renal cyst growth in ADPKD. Pharmacol Res. 114, Souverijn, J. H. & Blauw, G. J. Effect of simvastatin and “cystic” kidneys. a case series. BMC Nephrol. 12,
166–168 (2016). on renal function in autosomal dominant polycystic 48 (2011).
87. Nutahara, K. et al. Calcium channel blocker versus kidney disease. Nephrol. Dial. Transplant. 16, 130. Lantinga, M. A. et al. Management of renal cyst
angiotensin II receptor blocker in autosomal dominant 2152–2157 (2001). infection in patients with autosomal dominant
polycystic kidney disease. Nephron Clin. Pract. 99, 109. Fassett, R. G., Coombes, J. S., Packham, D., polycystic kidney disease: a systematic review.
c18–c23 (2005). Fairley, K. F. & Kincaid-Smith, P. Effect of pravastatin Nephrol. Dial. Transplant. 32, 144–150 (2017).
88. Zeltner, R., Poliak, R., Stiasny, B., Schmieder, R. E. on kidney function and urinary protein excretion in 131. Suwabe, T. et al. Cyst infection in autosomal dominant
& Schulze, B. D. Renal and cardiac effects of autosomal dominant polycystic kidney disease. polycystic kidney disease: causative microorganisms
antihypertensive treatment with ramipril versus Scand. J. Urol. Nephrol. 44, 56–61 (2010). and susceptibility to lipid-soluble antibiotics.
metoprolol in autosomal dominant polycystic kidney 110. Brosnahan, G. et al. Effect of statin therapy on the Eur. J. Clin. Microbiol. Infect. Dis. 34, 1369–1379
disease. Nephrol. Dial. Transplant. 23, 573–579 progression of autosomal dominant polycystic kidney (2015).
(2008). disease. A secondary analysis of the HALT PKD trials. 132. Nishiura, J. L. et al. Evaluation of nephrolithiasis in
89. van Dijk, M. A., Breuning, M. H., Duiser, R., Curr. Hypertens. Rev. http://dx.doi.org/10.2174/1573 autosomal dominant polycystic kidney disease
van Es, L. A. & Westendorp, R. G. No effect of enalapril 402113666170427142815 (2017). patients. Clin. J. Am. Soc. Nephrol. 4, 838–844
on progression in autosomal dominant polycystic 111. Myint, T. M., Rangan, G. K. & Webster, A. C. (2009).
kidney disease. Nephrol. Dial. Transplant. 18, Treatments to slow progression of autosomal 133. Torres, V. E., Wilson, D. M., Hattery, R. R. &
2314–2320 (2003). dominant polycystic kidney disease: systematic review Segura, J. W. Renal stone disease in autosomal
90. Ecder, T. et al. Diuretics versus angiotensin-converting and meta-analysis of randomized trials. Nephrology dominant polycystic kidney disease. Am. J. Kidney Dis.
enzyme inhibitors in autosomal dominant polycystic 19, 217–226 (2014). 22, 513–519 (1993).
kidney disease. Am. J. Nephrol. 21, 98–103 (2001). 112. van Keimpema, L. et al. Lanreotide reduces the 134. Grampsas, S. A. et al. Anatomic and metabolic risk
91. Kidney Disease: Improving Global Outcomes (KDIGO) volume of polycystic liver: a randomized, double-blind, factors for nephrolithiasis in patients with autosomal
Chronic Kidney Disease Work Group. KDIGO 2012 placebo-controlled trial. Gastroenterology 137, dominant polycystic kidney disease. Am. J. Kidney Dis.
clinical practice guideline for the evaluation and 1661–1668.e2 (2009). 36, 53–57 (2000).
management of chronic kidney disease. Kidney Int. 113. Hogan, M. C. et al. Somatostatin analog therapy for 135. Umbreit, E. C. et al. Percutaneous nephrolithotomy for
Suppl. 3, 1–150 (2013). severe polycystic liver disease: results after 2 years. large or multiple upper tract calculi and autosomal
92. Klahr, S. et al. Dietary protein restriction, blood Nephrol. Dial. Transplant. 27, 3532–3539 (2012). dominant polycystic kidney disease. J. Urol. 183,
pressure control, and the progression of polycystic 114. Meijer, E. et al. Rationale and design of the DIPAK 1 183–187 (2010).
kidney disease. J. Am. Soc. Nephrol. 5, 2037–2047 study: a randomized controlled clinical trial assessing 136. Mallett, A., Patel, M., Tunnicliffe, D. J. & Rangan, G. K.
(1995). the efficacy of lanreotide to Halt disease progression KHA-CARI autosomal dominant polycystic kidney
93. Appel, L. J. et al. Intensive blood-pressure control in in autosomal dominant polycystic kidney disease. disease guideline: management of renal stone disease.
hypertensive chronic kidney disease. N. Engl. J. Med. Am. J. Kidney Dis. 63, 446–455 (2014). Semin. Nephrol. 35, 603–606.e3 (2015).
363, 918–929 (2010). 115. Lantinga, M. A. et al. Hepatic cyst infection during use 137. Penniston, K. L., Wertheim, M. L., Nakada, S. Y. &
94. Ku, E. et al. Association between strict blood pressure of the somatostatin analog lanreotide in autosomal Jhagroo, R. A. Factors associated with patient recall
control during chronic kidney disease and lower dominant polycystic kidney disease: an interim of individualized dietary recommendations for kidney
mortality after onset of end-stage renal disease. analysis of the randomized open-label multicenter stone prevention. Eur. J. Clin. Nutr. 70, 1062–1067
Kidney Int. 87, 1055–1060 (2015). DIPAK‑1 study. Drug Saf. 40, 153–167 (2017). (2016).

18 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REVIEWS

138. Fink, H. A. et al. Medical management to prevent 157. Dougher, C. E. et al. Spot urine sodium measurements 177. Lacquaniti, A. et al. Apelin and copeptin: two opposite
recurrent nephrolithiasis in adults: a systematic review do not accurately estimate dietary sodium intake in biomarkers associated with kidney function decline
for an American College of Physicians Clinical chronic kidney disease. Am. J. Clin. Nutr. 104, and cyst growth in autosomal dominant polycystic
Guideline. Ann. Intern. Med. 158, 535–543 (2013). 298–305 (2016). kidney disease. Peptides 49, 1–8 (2013).
139. Casteleijn, N. F. et al. Novel treatment protocol for 158. Thomas, M. C. et al. The association between dietary 178. Kocer, D., Karakukcu, C., Ozturk, F., Eroglu, E. &
ameliorating refractory, chronic pain in patients with sodium intake, ESRD, and all-cause mortality in Kocyigit, I. Evaluation of fibrosis markers: apelin and
autosomal dominant polycystic kidney disease. patients with type 1 diabetes. Diabetes Care 34, transforming growth factor‑β1 in autosomal dominant
Kidney Int. 91, 972–981 (2017). 861–866 (2011). polycystic kidney disease patients. Ther. Apher. Dial.
140. Miskulin, D. C. et al. Health-related quality of life in 159. Mente, A., O’Donnell, M. J. & Yusuf, S. Sodium and 20, 517–522 (2016).
patients with autosomal dominant polycystic kidney cardiovascular disease — Authors’ reply. Lancet 388, 179. Helal, I. et al. Serum uric acid, kidney volume and
disease and CKD stages 1‑4: a cross-sectional study. 2113–2114 (2016). progression in autosomal-dominant polycystic kidney
Am. J. Kidney Dis. 63, 214–226 (2014). 160. O’Donnell, M. et al. Dietary sodium and disease. Nephrol. Dial. Transplant. 28, 380–385
141. Casteleijn, N. F. et al. A stepwise approach for effective cardiovascular disease risk. N. Engl. J. Med. 375, (2013).
management of chronic pain in autosomal-dominant 2404–2406 (2016). 180. Kocyigit, I. et al. Serum uric acid levels and endothelial
polycystic kidney disease. Nephrol. Dial. Transplant. 29 161. O’Donnell, M., Mente, A. & Yusuf, S. Commentary: dysfunction in patients with autosomal dominant
(Suppl. 4), iv142–iv153 (2014). Accepting what we don’t know will lead to progress. polycystic kidney disease. Nephron Clin. Pract. 123,
142. World Health Organisation. Cancer pain relief: with a Int. J. Epidemiol. 45, 260–262 (2016). 157–164 (2013).
guide to opiod availability — 2nd ed. (WHO, 1996). 162. Campbell, N. R. Dissidents & dietary sodium: 181. Akiyama, K., Mochizuki, T., Kataoka, H., Tsuchiya, K.
143. Ballantyne, J. C., Kalso, E. & Stannard, C. WHO concerns about the commentary by O’Donnell et al. & Nitta, K. Fibroblast growth factor 23 and soluble
analgesic ladder: a good concept gone astray. BMJ Int. J. Epidemiol. 46, 362–366 (2017). Klotho in patients with autosomal dominant polycystic
352, i20 (2016). 163. Graudal, N., Jurgens, G., Baslund, B. & kidney disease. Nephrology http://dx.doi.org/10.1111/
144. Tellman, M. W., Bahler, C. D., Shumate, A. M., Alderman, M. H. Compared with usual sodium intake, nep.12862 (2016).
Bacallao, R. L. & Sundaram, C. P. Management of low- and excessive-sodium diets are associated with 182. Pavik, I. et al. Soluble klotho and autosomal dominant
pain in autosomal dominant polycystic kidney disease increased mortality: a meta-analysis. Am. J. Hypertens. polycystic kidney disease. Clin. J. Am. Soc. Nephrol. 7,
and anatomy of renal innervation. J. Urol. 193, 27, 1129–1137 (2014). 248–257 (2012).
1470–1478 (2015). 164. Vegter, S. et al. Sodium intake, ACE inhibition, and 183. Pavik, I. et al. Secreted Klotho and FGF23 in chronic
145. PKD Foundation forum. Living with PKD: pain relief and progression to ESRD. J. Am. Soc. Nephrol. 23, kidney disease Stage 1 to 5: a sequence suggested
medical marijuana. PKD Foundation http://forums. 165–173 (2012). from a cross-sectional study. Nephrol. Dial. Transplant.
pkdconnection.org/viewtopic.php?f=3&t=109 (2017). 165. Lambers Heerspink, H. J. et al. Moderation of dietary 28, 352–359 (2013).
146. Walsh, N. & Sarria, J. E. Management of chronic pain sodium potentiates the renal and cardiovascular 184. Meijer, E. et al. Association of urinary biomarkers with
in a patient with autosomal dominant polycystic kidney protective effects of angiotensin receptor blockers. disease severity in patients with autosomal dominant
disease by sequential celiac plexus blockade, Kidney Int. 82, 330–337 (2012). polycystic kidney disease: a cross-sectional analysis.
radiofrequency ablation, and spinal cord stimulation. 166. Maroni, B. J., Steinman, T. I. & Mitch, W. E. A method Am. J. Kidney Dis. 56, 883–895 (2010).
Am. J. Kidney Dis. 59, 858–861 (2012). for estimating nitrogen intake of patients with chronic 185. Parikh, C. R. et al. Evaluation of urine biomarkers of
147. Loukas, M. et al. A review of the thoracic splanchnic renal failure. Kidney Int. 27, 58–65 (1985). kidney injury in polycystic kidney disease. Kidney Int.
nerves and celiac ganglia. Clin. Anat. 23, 512–522 167. Ko, G. J., Obi, Y., Tortorici, A. R. & Kalantar-Zadeh, K. 81, 784–790 (2012).
(2010). Dietary protein intake and chronic kidney disease. 186. Harskamp, L. R. et al. Urinary EGF receptor ligand
148. Casteleijn, N. F., de Jager, R. L., Neeleman, M. P., Curr. Opin. Clin. Nutr. Metab. Care 20, 77–85 excretion in patients with autosomal dominant
Blankestijn, P. J. & Gansevoort, R. T. Chronic kidney (2017). polycystic kidney disease and response to tolvaptan.
pain in autosomal dominant polycystic kidney disease: 168. Kalantar-Zadeh, K. et al. North American experience Clin. J. Am. Soc. Nephrol. 10, 1749–1756 (2015).
a case report of successful treatment by catheter- with low protein diet for non-dialysis-dependent 187. Zschiedrich, S. et al. Secreted frizzled-related protein
based renal denervation. Am. J. Kidney Dis. 63, chronic kidney disease. BMC Nephrol. 17, 90 (2016). 4 predicts progression of autosomal dominant
1019–1021 (2014). 169. Ma, Y. et al. A dietary quality comparison of popular polycystic kidney disease. Nephrol. Dial Transplant 31,
149. Valente, J. F., Dreyer, D. R., Breda, M. A. & weight-loss plans. J. Am. Diet Assoc. 107, 1786–1791 284–289 (2016).
Bennett, W. M. Laparoscopic renal denervation for (2007). 188. Ruggenenti, P. et al. Effect of sirolimus on disease
intractable ADPKD-related pain. Nephrol. Dial. 170. Levey, A. S. et al. Effect of dietary protein restriction progression in patients with autosomal dominant
Transplant. 16, 160 (2001). on the progression of kidney disease: long-term polycystic kidney disease and CKD stages 3b‑4.
150. Chapuis, O., Sockeel, P., Pallas, G., Pons, F. & follow‑up of the Modification of Diet in Renal Disease Clin. J. Am. Soc. Nephrol. 11, 785–794 (2016).
Jancovici, R. Thoracoscopic renal denervation for (MDRD) Study. Am. J. Kidney Dis. 48, 879–888
intractable autosomal dominant polycystic kidney (2006). Acknowledgements
disease-related pain. Am. J. Kidney Dis. 43, 161–163 171. Fouque, D. & Laville, M. Low protein diets for chronic M.B.L. is supported by the American Society of Nephrology
(2004). kidney disease in non diabetic adults. Cochrane Jared J. Grantham fellowship.
151. Bhatt, D. L. et al. A controlled trial of renal denervation Database Syst Rev, CD001892 (2009).
for resistant hypertension. N. Engl. J. Med. 370, 172. Boertien, W. E. et al. Short-term effects of tolvaptan Author contributions
1393–1401 (2014). in individuals with autosomal dominant polycystic Both authors contributed to all aspects of the preparation of
152. Haseebuddin, M. et al. Long-term impact of kidney disease at various levels of kidney function. this manuscript.
laparoscopic cyst decortication on renal function, Am. J. Kidney Dis. 65, 833–841 (2015).
hypertension and pain control in patients with 173. Wang, C. J., Grantham, J. J. & Wetmore, J. B. The Competing interests statement
autosomal dominant polycystic kidney disease. J. Urol. medicinal use of water in renal disease. Kidney Int. 84, A.B.C. declares an association with Otsuka Pharmaceutical
188, 1239–1244 (2012). 45–53 (2013). Group through membership of the TEMPO steering ­committee.
153. Millar, M. et al. Surgical cyst decortication in 174. Barash, I., Ponda, M. P., Goldfarb, D. S. & Skolnik, E. Y. M.B.L. declares no competing interests.
autosomal dominant polycystic kidney disease. A pilot clinical study to evaluate changes in urine
J. Endourol. 27, 528–534 (2013). osmolality and urine cAMP in response to acute Publisher’s note
154. Cook, N. R. et al. Long term effects of dietary sodium and chronic water loading in autosomal dominant Springer Nature remains neutral with regard to jurisdictional
reduction on cardiovascular disease outcomes: polycystic kidney disease. Clin. J. Am. Soc. Nephrol. 5, claims in published maps and institutional affiliations.
observational follow‑up of the trials of hypertension 693–697 (2010).
prevention (TOHP). BMJ 334, 885–888 (2007). 175. Wang, C. J., Creed, C., Winklhofer, F. T. &
155. Torres, V. E. et al. Dietary salt restriction is beneficial Grantham, J. J. Water prescription in autosomal DATABASES
to the management of autosomal dominant polycystic dominant polycystic kidney disease: a pilot study. ADPKD Mutation Database (PKDB): http://pkdb.mayo.edu/
kidney disease. Kidney Int. 91, 493–500 (2017). Clin. J. Am. Soc. Nephrol. 6, 192–197 (2011). Mayo Clinic ADPKD classification: http://www.mayo.edu/
156. McMahon, E. J., Campbell, K. L., Bauer, J. D. & 176. Higashihara, E. et al. Does increased water intake research/documents/pkd-center-adpkd-classification/
Mudge, D. W. Altered dietary salt intake for people prevent disease progression in autosomal dominant doc-20094754
with chronic kidney disease. Cochrane Database Syst. polycystic kidney disease? Nephrol. Dial. Transplant.
ALL LINKS ARE ACTIVE IN THE ONLINE PDF
Rev., CD010070 (2015). 29, 1710–1719 (2014).

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 19

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.

You might also like