You are on page 1of 23

PRIMER

Kidney stones
Saeed R. Khan1, Margaret S. Pearle2, William G. Robertson3,4, Giovanni Gambaro5,
Benjamin K. Canales1, Steeve Doizi6, Olivier Traxer6 and Hans-Göran Tiselius7
Abstract | Kidney stones are mineral deposits in the renal calyces and pelvis that are found free or
attached to the renal papillae. They contain crystalline and organic components and are formed when
the urine becomes supersaturated with respect to a mineral. Calcium oxalate is the main constituent
of most stones, many of which form on a foundation of calcium phosphate called Randall’s plaques,
which are present on the renal papillary surface. Stone formation is highly prevalent, with rates of up
to 14.8% and increasing, and a recurrence rate of up to 50% within the first 5 years of the initial stone
episode. Obesity, diabetes, hypertension and metabolic syndrome are considered risk factors for stone
formation, which, in turn, can lead to hypertension, chronic kidney disease and end-stage renal
disease. Management of symptomatic kidney stones has evolved from open surgical lithotomy to
minimally invasive endourological treatments leading to a reduction in patient morbidity, improved
stone-free rates and better quality of life. Prevention of recurrence requires behavioural and
nutritional interventions, as well as pharmacological treatments that are specific for the type of stone.
There is a great need for recurrence prevention that requires a better understanding of the
mechanisms involved in stone formation to facilitate the development of more-effective drugs.

Kidney stones (calculi) are mineral concretions in the implicated in the deaths of dogs and cats7,8 and caused
renal calyces and pelvis (FIG. 1) that are found free or a major health emergency in China in 2008. Melamine
attached to the renal papillae. By contrast, diffuse renal adulteration of infant formula led to the development
parenchymal calcification is called nephro­calcinosis1. of stones and sand-like calculi in the urinary tracts of
Stones that develop in the urinary tract (known as >294,000 infants9,10, >50,000 of whom were hospitalized;
nephro­lithiasis or urolithiasis) form when the urine six patients died as a result.
becomes excessively supersaturated with respect to a Stone formation is a common disease, with an esti­
mineral, leading to crystal formation, growth, aggre­ mated 5‑year recurrence rate of up to 50%11. The preva­
gation and retention within the kidneys2. Globally, lence of stones has been consistently increasing over the
approximately 80% of kidney stones are composed of past 50 years and further increases are expected owing
calcium oxalate (CaOx) mixed with calcium phos­ to changing lifestyle, dietary habits and global warm­
phate (CaP). Stones composed of uric acid, struvite ing 12–14. Obesity 15, diabetes16–18, hypertension13,17,19 and
and cystine are also common and account for approx­ metabolic syndrome20 are considered risk factors for
imately 9%, 10% and 1% of stones, respectively 3. Urine stone formation; conversely, stone formers are at risk
can also become supersaturated with certain relatively of hypertension19,21, chronic kidney disease (CKD) and
­insoluble drugs or their metabolites, leading to crystal­ end-stage renal disease (ESRD)22–25. The costs associated
lization in the renal collecting ducts (iatrogenic stones). with stone disease have also risen, increasing from an
For ­example, patients with HIV who are treated with estimated US$2 billion in 2000 to over US$10 billion in
protease inhibitors such as indinavir and atazanavir 2006 in the United States alone26.
Correspondence to S.R.K.
Department of Urology,
are at risk for developing nephrolithiasis4. Both indina­ Major advances have been made in the medical and
University of Florida, vir and atazanavir are metabolized by the liver, with a surgical management of patients with kidney stones.
1600 SW Archer Road, consider­able proportion of the drug excreted in the urine Stones can be fragmented using shockwave lithotripsy
Gainesville, Florida 32610, unchanged, leading to their crystallization and the for­ (SWL) to enable them to pass in the urine, or surgically
USA.
mation of kidney stones5. Even when given as part of a removed using percutaneous nephrolithotomy (PCNL)
khan@pathology.ufl.edu
multiple drug regimen, atazanavir can ­crystallize in the or retrograde intrarenal surgery (RIRS). PCNL involves
Article number: 16008 urine and form kidney stones6. direct endoscopic access into the kidney through an
doi:10.1038/nrdp.2016.8
Published online 25 Feb 2016; Poorly soluble dietary contaminants can also crystal­ incision in the flank, whereas RIRS is performed using
corrected online 12 Jan 2017 lize and form stones. For example, melamine has been a flexible fibre-optic ureteroscope to access the upper

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 1


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Author addresses Racial and ethnic differences in stone prevalence


have long been recognized. In the United States, non-­
1
Department of Urology, University of Florida, 1600 SW Hispanic white individuals have the highest prevalence
Archer Road, Gainesville, Florida 32610, USA. among racial and ethnic groups (10.3%), followed by
2
Department of Urology, UT Southwestern Medical Center, Hispanics (6.4%) and non-Hispanic African Americans
Dallas, Texas, USA.
(4.3%)12. Comparison of NHANES II (1988–1994)
3
Nuffield Department of Surgical Sciences, University of
Oxford, Oxford, UK. with NHANES III (2007–2010) data has shown that
4
Stone Biochemist, Lithoscreen Service, London, UK. the rise in kidney stone prevalence among Hispanics
5
Division of Nephrology and Dialysis, Columbus-Gemelli and African Americans was nearly double that of their
University Hospital Catholic University, School of white counterparts12,28.
Medicine, Rome, Italy. Geographical variation in stone disease typically
6
Department of Urology, Tenon Hospital, Hôpitaux reflects environmental risk factors, with higher stone
Universitaires Paris-EST, Assistance Publique Hôpitaux de prevalence in hot, arid climates. In the United States, kid­
Paris, Université Pierre et Marie Curie Paris 6, Paris, France. ney stones are most prevalent in the south and southeast
7
Division of Urology, Department of Clinical Sciences, regions and are lowest in the west of the country 28,34–37.
Intervention and Technology, Karolinska Institutet,
After controlling for other factors, ambient temper­ature
Stockholm, Sweden.
and sunlight have been shown to be independently
­associated with stone prevalence35.
urinary tract through natural passageways. Medical Numerous systemic diseases and factors have been
therapies are being used to ease stone passage, pro­ associated with an increased risk of kidney stones.
mote expulsion and reduce stone recurrence. Important Weight, weight gain, body mass index 15,38,39 and dia­
advances have also been made in our understanding of betes18,40 have been shown in large prospective cohort
stone pathogenesis. This Primer focuses on the medical studies to correlate with the risk of incident kidney
and surgical management currently practiced, as well as stones, with a greater effect in women than in men in
the contemporary understanding of stone pathogenesis. some cohorts. A multivariable model based on recent
NHANES data showed that obesity and diabetes were
Epidemiology associated with a 55% (95% CI: 1.25–1.94; P < 0.001) and
A recent review of epidemiological data from seven a 59% (95 %CI: 1.22–2.07; P < 0.001) increased risk of
countries revealed incidence rates for kidney stones of kidney stones, respectively 12. Metabolic syndrome has
114–720 per 100,000 individuals and prevalence rates also been linked to risk of kidney stones, with NHANES
of 1.7–14.8%, and in nearly all countries, the rates seem data indicating that the number of metabolic traits
to be rising 27. According to data from the National corre­lates with the risk of stones41. Jeong and colleagues42
Health and Nutrition Examination Survey (NHANES), detected a 25% higher rate of radiographically detected
the self-reported prevalence of kidney stones in the kidney stones among individuals with metabolic syn­
United States has increased nearly threefold, from drome in a screened population in Asia, after adjusting
3.2% in the period 1976–1980 to 8.8% in 2007–2010 for confounding variables (95% CI: 1.03–1.50; P < 0.001).
(REFS 12,28). The lifetime prevalence of kidney stones in Finally, risk of cardiovascular disease has been associ­
the United Kingdom increased by 63% (7.14–11.62%) ated with a history of kidney stones, although a cause and
between 2000 and 2010 (REF. 29). effect relationship has not been definitively established.
The propensity to form stones varies according to Ferraro and colleagues43 showed a modest increased risk
sex, ethnicity and geography. Although historically of incident cardiovascular disease among women with a
stones have been 2–3‑times more common in men history of stones (an adjusted multi­variable hazard ratio
than in women, recent data indicate that this dis­ of 1.30; 95% CI: 1.04–1.62) but not in men in three large
parity is diminishing. For example, data from the US prospective cohorts. Similarly, among individuals regis­
Nationwide Inpatient Sample revealed a decline in the tered in the Canadian health care system, a 63% higher
male to female ratio for hospital discharges for stones, risk of incident myocardial infarction was detected
from 1.7 in 1997 to 1.3 in 2002 (REF. 30). The male to among stone formers (95% CI: 1.51–1.76), with a greater
female ratio of incident kidney stones also declined in effect in women than in men44. A matched pair analysis,
Rochester, Minnesota, USA, from 3.1 to 1.3, between adjusted for confounding factors, revealed a 31% higher
1970 and 2000 (REF. 31). In Florida (USA), analysis of risk of myocardial infarction in stone formers than in
resource use related to procedures for stones revealed the general population in Olmstead County, Minnesota,
that the increase in rates in women was greater than USA (95% CI: 1.02–1.69)45.
that in men between 1998 and 2004 (REF. 32). In Canada,
a 48% increase in stone treatment between 1991 and Mechanisms/pathophysiology
2010 was primarily accounted for by an increase in Data from patients with stone disease provide visual and
procedures among women33. The reason for the surge histological observations of the kidneys, microscopic
in stone disease in women is not precisely understood, and biochemical analyses of the urine and determination
but some have speculated that it might be attributable of the crystalline nature of the stones. Such observational
to changes in lifestyle and diet, resulting in increased data only produce a ‘snapshot’ at the end of a long pro­
obesity among women, a known risk factor for cess of the stone formation. To understand the mech­
stone formation30. anistic details theoretical, animal and cell culture models

2 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Kidney a b
Glomerulus

Distal
tubule

Proximal Apatite
Upper tubule
pole
Vasa recta Thick
ascending
Middle limb
pole Pelvis
Thin
descending
limb
Lower
pole Terminal Randall’s
Loop of collecting plug
Henle duct (apatitic)
Papilla Cortex Pelvis
Randall’s
Stone plaque (apatitic) Stone
Ureter Medulla

Figure 1 | Macroscopic and microscopic morphology of human kidneys and location of stones.
Nature Reviewsa | According
Disease Primers
to
the fixed-particle mechanism, stones begin as depositions of calcium phosphate (CaP) in the interstitium (apatite),
grow outwards reaching the renal papillary surface, become exposed to the pelvic urine and establish a nucleus for the
deposition of calcium oxalate (CaOx), leading to the formation of CaOx stones attached to a CaP base, known as
Randall’s plaques. b | By contrast, in the free-particle mechanism, for example, CaP, uric acid or cystine crystals form in
the renal tubules, move with the urine, aggregate and plug the terminal collecting ducts. These plugs, called Randall’s
plugs or lesions, are exposed to the pelvic urine. Deposition of CaOx crystals on the CaP plugs leads to the formation of
CaOx kidney stones.

have been developed. Indeed, results obtained from Calculi types. The four main types of stones are named
such studies are providing a better understanding of the after their major constituents. Calcium stones are the
pathogenesis of stone formation and are described here. most common and occur as CaOx and CaP crystals,
alone or in combination. Most kidney stones are par­
Microstructure of kidney stones tially or completely composed of CaOx, which exists
Kidney stones are solid masses, ranging in size from a as a monohydrate or dihydrate. Individual crystals of
grain of sand to a pearl (or larger) — a stone does not have CaOx monohydrate (COM) are thin and plate-like, and
to cause symptoms. Depending on their composition, generally acquire a ‘dumb-bell’ shape through twinning,
stones are either yellow or brown in colour and smooth as seen in urinary sediments. Inside the stones, COM
or jagged in appearance. They are composed of crystals crystals are arranged radially into fan-shaped profiles
(FIG. 2) and a ubiquitous organic matrix (FIG. 3), which not with distinct concentric laminations, showing out­
only coats the crystals but is also present inside the crystals ward growth of the crystals and stones. CaOx dihydrate
and the inter-crystalline spaces46–48. The matrix of calcific (COD) crystals have character­istic tetragonal bipyrami­
stones contains many macromolecules, including osteo­ dal shape both in urinary sediment and in kidney stones.
pontin (which also has a role in the biomineralization CaOx stones are small with shiny exteriors and generally
of bone), inter‑α‑inhibitor (which is a plasma protein) contain both COM and COD crystals. COM stones are
and urinary prothrombin fragment 1 (UPTF1) — all of more common than the pure COD stones61. In mixed
which are normally present in the urine49, albeit in small stones, COD crystals are predominantly present on the
quantities49–51. The matrix also contains various forms of stone surface, which appears jagged. By contrast, pure
lipids, which have been shown to induce crystal nucle­ COM stones have smooth surfaces. CaOx stone forma­
ation52–56. The association between the crystals and the tion is a multistep process (see below). Hypercalciuria,
matrix seems to start early upon crystal nucleation and hyperoxaluria and h ­ ypocitraturia are major risk factors.
continues throughout the formative and growth phases of CaP is mainly found as basic CaP (apatite), calcium
the developing stone. Although some urinary molecules, hydrogen phosphate dihydrate (brushite) or tricalcium
such as UPTF1, are considered crystallization inhibitors, phosphate (whitlockite). Pure CaP stones are rare62.
others such as osteopontin can act as both inhibitors and Apatite is the most common crystal in kidney stones
promoters of crystallization57. These molecules seem to and is often a powdery mass that fills the spaces in
be produced as a protective response against mineral­ between other types of crystals, mainly CaOx crystals.
ization. However, both CaOx and CaP crystals have been Whitlokite is very rare in both kidney stones and uri­
shown to induce the production of macromolecules that nary sediments. Brushite frequently occurs in kidney
inhibit and/or modulate crystallization51,58–60. stones and is present as rosettes of radially arranged

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 3


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

thin blade-like crystals. Hypercalciuria, hypocitraturia such as gout (gouty diathesis). Increased purine catabo­
and increased urinary pH are major risk factors for CaP lism (which can occur in those with myeloproliferative
stone formation63. disorders or in those receiving chemotherapy) and the use
Uric acid stones comprise 8–10% of all kidney stones of drugs that prevent renal reabsorption of uric acid are
worldwide, with a disproportionate prevalence in stone also contributing factors. Most uric acid stones are com­
formers who are obese and insulin resistant — two of pact, appearing like pebbles, with a central core of loosely
the main components of metabolic syndrome. Unlike aggregated anhydrous uric acid crystals surrounded by
calcium stone types, overly acidic urine (a pH of <5.5) is radiating columnar anhydrous uric acid crystals organ­
recog­nized as the main abnormality responsible for uric ized in concentric laminations66,67. Some stones display a
acid nephrolithiasis64. In addition to the insolubility of compact outer layer enclosing a porous friable interior
uric acid at low urinary pH and dehydration, conditions consisting of anhydrous uric acid, uric acid ­dihydrate and
that lead to excessive urinary uric acid excretion, known COM crystals mixed with organic material.
as hyperuricosuria, have also been associated with uric Struvite stones, also known as ‘infection stones’, rep­
acid stone formation. These high levels might be due resent 7–8% of all stones worldwide and are typically
to excess dietary intake of purine-rich foods65 or endo­ caused by increased production of ammonia secondary
genous uric acid overproduction, as occurs in conditions to infection with urease-producing organisms, such as
Proteus or Klebsiella. The subsequent alkaline urine leads
to the formation of magnesium ammonium phosphate
a b hexa­hydrate crystals68. Struvite and associated carbo­
nate apatite crystals can grow quickly into large stones
referred to as staghorn calculi, appropriately named
for their horn-like projections that occupy the renal
­pelvis and renal calyces. Although historically feared
for their association with high mortality, struvite stones
and their association with urosepsis and infections are
now treatable with surgical intervention and antibiotics.
In the modern era, these stone types are better known
for their preponderance for recurrence, particularly in
immuno­compromised individuals with incomplete stone
removal. Struvite stones are large aggregates of ortho­
rhombic ‘coffin-lid’-shaped struvite crystals covered with
spherulitic carbonate a­ patite crystals and mixed
with ­cellular debris, which often included bacteria67.
Finally, cystine stones form as result of an autosomal
500 μm 500 μm
recessive defect in the renal transporter of the amino
acid cystine69. The lack of cystine reabsorption leads to
c d increased urinary cystine excretion. At normal urinary
pH, cystine is insoluble and forms cystine crystals that
can aggregate to form recurrent kidney and bladder
stones. Cystine stones are compact, amber coloured,
slightly opaque and with homogenous interiors. Higher
magnification of the stone and urinary deposits reveals
a unique and characteristic hexagonal structure of the
cystine crystals62.

Chemistry of stone formation


Several models of kidney stone formation have been
proposed; the two dominating mechanisms for the initi­
ation of stones are commonly described by the terms
‘free particle’ (in which crystals form ‘Randall’s plugs’
in the tubule) and ‘fixed particle’ (in which stones grow
50 μm 5 μm
on so-called Randall’s plaques; FIG. 1). Although these
models encompass all the possible hypothetical models
of how stones begin, no single model can rationalize the
Figure 2 | Calcium oxalate kidney stones examined Nature using scanning
Reviews | electron
Disease Primers
evidence observed from all patients with stones — many
microscopy. a | A low magnification image showing the outer nodular appearance of
factors probably contribute. Regardless of the model,
a calcium oxalate (CaOx) monohydrate stone. b | The fractured surface showing CaOx
monohydrate crystals organized in concentric laminations and radial striations. the chemical processes of nucleation and crystal growth
c | The surface of a stone with an outer layer of CaOx dihydrate crystals. Bipyramidal are essential for the initiation and development of all
CaOx dihydrate crystals protruding on the surface are shown (arrows) and are powdered stone types70. Stone formation is caused by an abnor­
with tiny calcium phosphate crystals. d | The surface of a CaOx monohydrate stone. mal combination of factors that influence the thermo­
The tabular morphology of CaOx monohydrate crystals is clearly visible. dynamic driving force (supersaturation) and the kinetic

4 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

a b Supersaturation. Estimating the degree of supersatur­


ation of urine with respect to each stone-forming
mineral requires the measurement of 15 urinary constit­
uents, including any crystals that may have precipitated
before or after micturition (urination)2,70–75. Using an
iterative ion speciation programme2,72–75, these data can
be used to calculate the concentrations of the various
soluble complexes formed between these urinary con­
stituents, the ionic strength of the urine and the activity
10 μm 5 μm ­co­efficients of all the ions involved. From this informa­
tion, the relative supersaturation (RSS) of the urine with
c d respect to the mineral concerned can be calculated73;
this is the theoretical RSS that would be achieved if all
potential crystalline material in the urine remained
in solution.
The RSS level at which nucleation occurs is known
as the formation product of the mineral concerned; it
is not a fixed thermodynamic constant, but covers a
range of supersaturation values within which de novo
crystal nucleation can take place. Its value depends on
0.5 μm 0.5 μm
several factors. First, the length of time of incubation
affects the RSS. The longer a given supersaturated solu­
Figure 3 | Scanning electron microscopy and transmission Natureelectron
Reviewsmicroscopy
| Disease Primers
of tion is left to stand, the more likely it is to precipitate
kidney stones. These images demonstrate the ubiquitous nature of the organic matrix, crystals. The higher the initial RSS, the shorter is the
and its intimate association with the crystalline components of the stone. Stone
nucle­ation time76. Low initial levels of RSS generally
fragments were demineralized and then processed for electron microscopy. The stone
and crystals maintained their architecture even after the removal of the crystalline require longer incubation times before nucleation takes
components. a | A scanning electron microscopy image of the organic matrix of calcium place, which is important when considering the likeli­
oxalate (CaOx) dihydrate crystals in a CaOx stone. The organic matrix is organized in hood of crystal nucleation occurring during the short
layers, but the overall bipyramidal architecture of the CaOx dihydrate crystal is period when urine is forming in the kidneys77,78. The
maintained even after demineralization. b | The fractured surface of CaOx monohydrate second factor that can affect the RSS value is the pos­
crystal ‘ghosts’ showing internal spaces (arrows) created by a loss of plate-like CaOx sible presence of nucleation inhibitors in the urine that
monohydrate crystals. c | A transmission electron microscopy image of the organic could retard the process by ‘poisoning’ active growth
matrix, showing at least two layers of radially organized CaOx monohydrate crystal sites on n ­ ascent crystals. Most studies on the effect of
ghosts. d | A higher magnification of the CaOx monohydrate crystal ghosts from part c calcium salt nucleation inhibitors have been carried out
stained with antibodies against osteopontin (black dots), demonstrating its substantial
using relatively long incubation periods (24–72 hours)
presence in the matrix.
and low RSS levels79,80 that do not mirror the situation
in the kidneys. However, many of these presumed
(rate-controlling) processes involved in the crystalliza­ inhibitors have been shown to have little effect on the
tion of the various stone-forming minerals. The princi­ nucleation of calcium salts under conditions that mimic
pal thermo­dynamic driving force for both stages is the urine crystallization (that is, high RSS levels and short
degree of supersaturation of the fluid within which initi­ incubation (3–4 minutes)). Furthermore, the RSS of
ation occurs70,71. Whether this takes place intracellularly tubular fluid with respect to calcium salts often exceeds
or extracellularly, the laws of crystallization ­chemistry their 3–4‑minute formation product values78,81. Thus,
must apply. the concept that inhibitors of nucleation play a part
in preventing calcium-­containing stones is question­
Crystal nucleation. Nucleation (in which solute mol­ able because their effect may be overwhelmed by high
ecules dispersed in a solvent begin to cluster) is the first ­ambient levels of supersaturation.
stage in crystallization and can occur homogeneously
or heterogeneously. Homogeneous nucleation requires a Crystal growth and agglomeration. Once a crystal
high degree of supersaturation with respect to the min­ nucleus is established inside the kidneys71, exposure
eral concerned; in vitro this would normally take place in to the urine enables the stone to grow by encrust­
a pure solution containing no particulate matter and in a ation82,83. There are two basic pathways (free-particle
receptacle that is chemically inert. By contrast, hetero­ and fixed-particle mechanisms) for the establishment
geneous nucleation is the much more likely mechanism of a stone nucleus, both of which can be active in any
through which crystal initiation occurs in the urine71. stone former, although stones from idiopathic stone
The process can occur in the presence of particulates formers are generally formed attached to plaques84,85
consisting of proteins, other organic polymers or crystals (FIG. 1).
of another mineral, and is contained within receptacles In the free-particle mechanism71,77, crystals nucle­
lined with chemically active cell surfaces. Heterogeneous ate, grow and aggregate within the urine of the renal
nucleation requires a lower level of supersaturation than tubules. Once crystals aggregate to form large particles,
homogeneous nucleation for crystal initiation. they are retained inside the kidneys either by becoming

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 5


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

too large to pass through the tubular lumens or by High-resolution radiography of cadaveric kidneys
attaching themselves to the tubular epithelium86. In was performed by Stoller et al.105. They reported that
the presence of high supersaturation, crystal deposits 57% of the kidneys had subepithelial Randall’s plaques
occlude the collecting ducts87 forming Randall’s type 2 that extended deep within the papillae and were inti­
lesions or plugs88, which protrude out into the renal mately associated with the collecting ducts and the vasa
pelvis and become exposed to the pelvic urine. Once recta. Furthermore, von Kossa-positive (which stains for
tubular openings are blocked, stasis can promote the CaP) spherical CaP deposits were identified scattered in
formation of small stones behind the plugs. Similarly, the interstitium as well as around the collecting ducts
unattached stones in the renal c­ alyces can also form and blood vessels. Evan et al.96 examined renal papillae
through the free-particle mechanism. from patients with various causes who form stones. They
Support for the free-particle mechanism comes concluded that all idiopathic calcium stones develop
from animal models and tissue culture studies. attached to subepithelial Randall’s plaques109, and con­
Experimentally induced hyperoxaluria or hyper­ firmed the earlier observations of Cooke et al.103,108 about
calciuria leads to increased urinary supersaturation the involvement of the basement membrane of the
and the formation of CaOx or CaP crystals in the renal loop of Henle in the development of Randall’s plaques.
tubular lumen1,89,90. Crystals are retained at sites where Osteopontin3,110, heavy chain of inter‑α-inhibitor 111,112,
urin­ary flow is impeded, either through a narrowing collagen103,104,107 and zinc113 have been identified in these
of the tubule as occurs where the proximal tubule meets interstitial plaques. Interestingly, all Randall’s plaques
the loop of Henle or at the papillary base where renal do not develop into stones — plaques have also been
tubules bend91. Interestingly, the openings of the collect­ ­identified in the kidneys of non-stone formers107.
ing ducts are slit-like and narrower than the luminal Evan et al.96,114 further hypothesized that deposits
diameter of the duct, which hinders urinary passage migrate from the basement membrane of the loop of
and might play a part in ductal plugging 77,86. Crystal Henle into the surrounding interstitium and become
retention is also accomplished by attaching to the renal associated with type 1 collagen, fusing into a sync­
epithelium, as well as the basement membrane, which tium in which islands of mineral ‘float’ in an organic
has become exposed by epithelial sloughing 1,92. ‘sea’. Microscopic and analytical studies of plaques
This mechanism is probably involved in the forma­ have shown that interstitial CaP deposition occurs in
tion of apatite, brushite and cystine stones, as well as close association with cellular degradation products,
CaOx stones associated with primary hyperoxaluria membrane-­bound vesicles, some unidentified fibril­
and hyperoxaluria after bariatric surgery 93–98. Epithelial lary mat­erial and collagen fibres100 (FIG. 4). Some vesicles
injury and sloughing is common in patients with brush­ contained needle-shaped crystals, presumably CaP100.
ite stones and in hyperoxaluric CaOx stone formers96,99. Accordingly, crystal formation has been proposed to
The plugs act as substrates for future crystal deposition begin in membrane-bound vesicles, propagated through
and stone growth and can have the same or different the mineralization of collagen, leading to the move­
crystalline composition96,97. ment of a mineralization ‘front’ to the subepithelium
The alternative mechanism for stone development of the renal papillae and the development of Randall’s
is the fixed-particle mechanism in which stones form plaques100. Thus, there are differences of opinions about
attached to calcific plaques on the papillary surface. The the beginning of the Randall’s plaque. Does it start in the
plaques, called Randall’s plaques, start with CaP crystal basement membrane of the loop of Henle, the vasa recta
formation and deposition in the renal interstitium88,100. or the collecting ducts, or simply in the interstitium from
A substantial number of idiopathic CaOx stones are where it grows outwards and in the process involves all
formed attached to Randall’s plaques87,101,102. The CaP components of the renal papillae? What provokes the
deposits comprising the plaque seem to start deep in formation of Randall’s plaques is unknown. It has been
the renal interstitium, in the basement membrane of the proposed that the formation of Randall’s plaques might
loop of Henle103,104, or are associated with the collecting be similar to vascular calcification115,116 (see below).
ducts and vasa recta (the blood vessels that lie ­parallel One of the key differences between the free-­particle
to the loop of Henle)105,106. Haggitt and Pitcock107 exam­ and fixed-particle mechanisms is the location of the ­initial
ined kidneys from 100 randomly selected cadavers nidus (FIG. 1). Randall’s plugs (free particle) are tubular
and performed light microscopy and transmission deposits — usually consisting of CaP — that are con­
electron microscopy (TEM) on selected specimens. tinuously exposed to the urine and grow accordingly.
They found alizarin red-positive (which stains for cal­ Conversely, Randall’s plaques (fixed particle) are sub­
cium) laminated spherulitic crystals in the interstitium, epithelial deposits that must have their surfaces breached
which, on closer examination by TEM, were shown to before the crystalline mass is exposed to the urine for
be associ­ated with collagen fibres in the interstitium further growth (FIG. 5). Such a breach can happen with the
as well as the basement membrane of the collecting involvement of matrix metalloproteinases and/or owing to
ducts. Furthermore, Cooke and colleagues103,108 studied the sheer force of the growing plaque88,117. Biological apa­
62 normal kidneys and found calcification in four, all tite is the main constituent of the plaques, whereas plugs
of which occurred in the basement membrane of the can be made of any solid that can precipitate out into the
loop of Henle and extended into the medullary inter­ supersaturated urine. However, both plugs and plaques
stitium. Some collecting ducts and blood vessels were are coated with an organic matrix, which (as mentioned
also involved. earlier) is made of macromolecules produced by renal

6 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Rate of crystal growth. The rate of crystal growth of a par­


a
ticular mineral is mainly dependent on its RSS level in the
SCaP
urine but might also be influenced by certain modifiers
of crystallization. Some modifiers have been shown to
retard the rate of growth and/or agglomeration of crystals
of calcium salts in vitro. These modifiers include mag­
nesium122, citrate123, pyrophosphate79,80, ADP124, ATP124,
phosphopeptides125, various glycosaminoglycans126,127,
non-polymerized Tamm–Horsfall protein (also known
CaP as uromodulin)128,129, nephrocalcin130,131, osteopontin132,133,
calgranulin134, α1‑­microglobulin135, β2‑microglobulin136,
SCaP
UPFT1 (REFS 137,138) and inter‑α-inhibitor (bikunin
light chain)111,139. However, only citrate and magnesium
have been found to be excreted in lower amounts by some
calcium stone formers than by non-stone formers140.
Collagen A second group of modifiers is postulated to stimu­
late the crystallization of calcium salts. Known as crystal­
lization promoters, they include matrix substance A135,
various uncharacterized urinary proteins and glyco­
proteins141–143 and the polymerized form of Tamm–
Basement Horsfall protein (uromucoid)144–146. So far, stone formers
membrane 2 μm have not been shown to excrete an excess of any of these
macromolecules, but some researchers still believe that
b they might have a role in the growth phase of many
stones and research continues on their possible role in
­promoting the development of stones.

Stone formation and vascular calcification


Several analyses have suggested a link between vascu­
lar calcification and the formation of idiopathic kidney
stones147–151. Deposition of CaP in association with col­
lagen is similar to what is seen at sites of vascular calcifi­
Collagen CaP cation, which is currently considered to be an active
process in which vascular smooth muscle cells acquire
100 nm an osteogenic phenotype152–154. Exposure of vascular
smooth muscle cells to high levels of calcium and phos­
phate triggers this transformation154,155, which activates
Figure 4 | The renal interstitium of a calcium oxalate Nature
stone former
Reviews with Randall’s
| Disease Primers
plaque. a | The tubular epithelium is separated from the interstitium by the basement
bone morphogenetic proteins (BMPs) and WNT signal­
membrane. The interstitium contains collagen fibres, spherulitic calcium phosphate ling pathways via upregulation of Runt-related transcrip­
(SCaP) reminiscent of membrane-bound vesicles with crystals, as well as dense and tion factor 2 (RUNX2) and msh homeobox 2 (MSX2)
compacted calcium phosphate (CaP) crystals. There is a close association between transcription factors156. Transformation is also mediated
collagen and dense CaP. b | A portion of the image in part a, but at a higher magnification. by reactive oxygen species157,158; the transformed cells
Collagen fibres are intimately associated with dense deposits of CaP. produce matrix proteins154,159. Some evidence supports
the theory that renal epithelial cells have the capacity
to become osteogenic116. Basal levels of BMP2, RUNX2
epithelial cells in response to their exposure to tubular and osterix (also known as Sp7; another transcription
deposition of crystals. Pelvic urine is normally metastable factor) are higher in genetic hypercalciuric rats that pro­
with respect to CaOx and would support heterogeneous duce intrarenal CaP deposits160. Indeed, Madin–Darby
nucleation and growth of CaOx. Animal model and tissue canine kidney cells produce CaP microliths on the basal
culture studies have shown that continuous exposure of side when grown in monolayers161,162, and exposure of
epithelial cells to crystals leads to the production of large these cells to high oxalate levels, as well as CaOx and
amounts of macromolecules. These molecules travel CaP crystals, leads to the activation of NADPH oxidase
down with the urine and, when coating the crystal aggre­ and the production of reactive oxygen species60,163–167,
gates, promote further crystallization and stone growth. resulting in an osteogenic phenotype. Evidence has
The formation and growth of CaOx crystals over plaques shown that genes considered to be involved in epithe­
or plugs would eventually lead to the development of a lial transformation and bone morphogenesis — includ­
calcific stone that is attached to the renal papillary tip. CaP ing those that encode RUNX2, osterix, BMP2, BMP7,
by itself is also a good nucleator of CaOx 118 and has been BMP receptor type 2, collagen, osteocalcin, osteo­
shown to transform itself into CaOx through dissolution nectin, osteopontin, matrix-gla-protein, osteoprotegrin,
and recrystallization119–121. Thus, stone formation is the ­cadherin, fibronectin and vimentin — are upregulated
end product of a cascade of events (FIG. 6). in hyperoxaluric rats168, all of which are markers of the

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 7


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

osteogenic phenotype. Overall, some evidence supports urinary tract by imaging studies or surgery. Otherwise,
that the abnormal urinary conditions of hyperoxaluria, other possible causes of the above manifestations should
hypercalciuria, hypocitraturia and renal oxidative stress be investigated.
trigger the transformation of renal epithelial cells into Symptoms of nephrolithiasis in children are often
osteoblastic phenotypes. The dedifferentiation pro­ unclear. The only manifestations are microhaematuria
motes the deposition of CaP crystals and the formation or recurrent episodes of macrohaematuria; pollachiuria–­
of Randall’s plaques88. dysuria; acute or subacute episodes of recurrent abdom­
inal pain, located in the flank in older patients or diffuse
Diagnosis, screening and prevention in the abdomen in younger (<8 years of age) patients;
Patients with urinary stones generally present with the urinary tract infections; or, finally, nonspecific manifest­
typical reno-ureteral colic and less frequently with loin ations such as irritability, vomiting, unmotivated crying
pain; associated manifestations could be gross haema­ and motor agitation in those <2 years of age. In the
turia, vomiting and sometimes fever. However, patients absence of a clear demonstration of a stone, the only
can also be asymptomatic. A diagnosis of nephrolithiasis clue that these manifestations are due to urinary stones
is only confirmed when a stone has been passed, has could be the observation of concurrent hypercalciuria
been extracted or destroyed, or has been identified in the or hyperuricosuria.
A detailed medical history and physical examination
is part of the evaluation of patients with a suspected stone.
a Clinical diagnosis generally needs to be supported by
appropriate imaging. Renal and urinary tract ultrasono­
Papillary
surface
graphy can identify stones that are situated in the calices,
the pelvis and the pyeloureteric and vesico­ureteric junc­
Crystalline tions; it may also identify indirect signs of a stone such
shell as pyeloureteric dilation and a perirenal film of extrava­
sated urine. Indeed, ultrasonography has been shown
to identify renal stones with a sensitivity of 70% and a
Protruding
crystal specificity of 94%169. For ureteral stones, the sensitivity
and specificity of ultrasonography is lower at 57.3% and
97.5%, respectively 170. Plain abdominal X‑ray imaging is
generally not used anymore in the evaluation of a flank
pain given its modest diagnostic performance (sensitivity
and specificity of 44–77% and 80–87%, respectively)171.
However, plain abdominal X‑ray imaging might still
have a role in distinguishing between r­ adiopaque and
­radiolucent stones and in follow‑up care172.
100 μm
Intravenous urography, which was the historical
gold-standard imaging technique for urolithiasis, has been
b replaced by non-contrast-enhanced CT (NCCT) owing
to its higher sensitivity and specificity for identify­ing ure­
Protruding Fibrous
crystal material teral stones regardless of location, size and composition;
its lack of contrast agents; and because it can recognize
extraurinary causes of renal colic in 30% of patients173.
Moreover, NCCT can determine the stone density and
inner structure, as well as the skin‑to‑stone distance,
which are useful ahead of extracorporeal SWL174.
12 μm
Categories of stone formers
c Patients with kidney stones are generally classified in
different categories according to the composition of
stones and the history of previous stone episodes. This
classification influences the diagnostic work up and
preventive treatment. However, both of these aspects
have limitations. For example, classification according
6.7 μm to stone composition has the caveat that chemical meth­
ods are imprecise and do not distinguish the crystalline
Naturemonohydrate
Reviews | Disease Primers forms175; infrared spectroscopy or X‑ray diffraction are
Figure 5 | The renal papillary surface of a calcium oxalate stone
former examined using scanning electron microscopy. a | A crystalline entity is the preferred methods.
protruding through the papillary surface, causing epithelial cells to be pushed apart Classification according to a history of previous
(arrow). The protrusion is covered with fibrous material and surrounded by a layer of stone episodes is easier because a single stone former
crystalline shell. b | Fibrous material covering the protrusion. c | An intact portion of the is defined as one who seeks advice for a single kidney
papillary surface epithelium. stone episode; by contrast, a recurrent stone former is a

8 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER
a b 1
1 Supersaturation of the tubular fluid

2 3
Basement
membrane

Calcified
2 interstitial
Collagen collagen
Mineralization
front

Plaque initiation

Papillary surface epithelium


Matrix 5 4
vesicle 3

6
5

Calcification

Urinary
CaP macromolecules
CaOx

Figure 6 | Randall’s plaque and calcium oxalate stone formation. On the basis of experimental and available clinical data,
one can surmise that stone formation is a multistep process, probably involving the formation Nature
ofReviews Disease
Randall’s| plugs andPrimers
plaques.
For simplicity, we have separated out these steps, but a combination of factors probably contributes to stone formation and
growth. a | In Randall’s plaque formation, supersaturation of tubular fluid with respect to calcium phosphate (CaP) and/or
calcium oxalate (CaOx) occurs in renal tubules at the end of the loop of Henle and at the beginning of the colleting duct
system (step 1). From here, two alternate pathways have been suggested. Supersaturation with respect to CaP leads to
the deposition of CaP in the basement membrane of the loop of Henle (step 2), initiating the process of plaque formation104
(FIG. 1). Alternatively, crystal formation occurs in the renal tubules (step 3). CaP crystals translocate into the interstitium
(step 4) or are internalized by the cells, dissolved and re‑precipitated in the tubular basement membrane121. Another
possibility is that renal epithelial cells, when exposed to CaP and/or CaOx, produce reactive oxygen species and, probably,
a range of factors associated with osteogenesis, such as Runt-related transcription factor 2 (RUNX2), osterix (also known as
Sp7), bone morphogenetic protein 2 (BMP2), BMP7, BMP receptor type 2 (BMPR2), collagen and osteopontin. Epithelial cells
produce matrix vesicles on the basal side (step 5) followed by their calcification (step 6; REFS 88,168). b | Once CaP crystals
have been deposited in the basement membrane of the loop of Henle and/or collecting ducts, mineralization continues.
Collagen fibres and membranous vesicles become calcified (step 1). The mineralization front reaches the renal papillary
surface and a subepithelial plaque is established (step 2). The papillary surface epithelium is disrupted (step 3) and plaques
rupture, exposing the CaP crystals to the pelvic urine metastable with respect to CaOx (step 4). Urinary macromolecules
are deposited over the exposed CaP crystals, promoting the deposition of CaOx crystals on the CaP base (step 5).

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 9


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

We suggest that stone disease should not be evaluated


Box 1 | Categories of patients with renal stones only in reference to stone composition and clinical activ­
Non-calcium stones ity, but the risk of complications should also be appraised
• Struvite stones in the diagnostic process and patient c­ ategorization
(BOX 1).
• Matrix stones
• Uric acid stones
Diagnosis
• Other purine stones (such as xanthine and Diagnosing and developing effective prevention strat­
2,8‑dihydroxyadenine)
egies requires both understanding the metabolic back­
• Cystine stones ground that promotes lithogenesis and assessing the
• Drug stones patient’s risk of CKD and metabolic bone disease (MBD).
Calcium stones Metabolic evaluation of patients with stones aims to esti­
• Single and sporadic stone formers who are not at risk of mate the propensity of the urine to crystallize, investigate
chronic kidney disease and/or metabolic bone disease the metabolic mechanisms of nephrolithiasis, diagnose
• Single and sporadic stone formers who are at risk of underlying systemic causes of nephrolithiasis, determine
chronic kidney disease and/or metabolic bone disease the risk of CKD and MBD and to achieve insights on
• Recurrent stone formers nutritional habits.
Specific software for estimating the urine propensity
for crystallization have been proposed (for example, the
patient who has passed many temporally spaced stones. EQUIL and JESS programs)73,178. These programs cal­
Given that no metabolic markers are available to distin­ culate the urinary supersaturation of numerous salts by
guish between single and recurrent stone formers, a long considering the concentration in the urine of different
observation period is required to determine whether ionic species. The probability of stone formation can also
a patient is a single or a recurrent stone former. The be evaluated by algorithms based on urine excretion of
majority of stone formers will pass only a single stone a limited number of parameters140. Although these
in their lifetime or over a long observation period176. methods can be of help, they are not compulsory for a
Furthermore, recurrence of stone formation in most ­successful metabolic assessment and follow‑up.
first-time calcium stone formers occurs in a period that
could be as long as 5 years, and only 15–20% of calcium Risk factors of CKD and MBD. The categorization
stone formers recur ≥4 times176. Accordingly, the term of first-time or sporadic stone formers, according to
recurrent calcium stone former has been proposed to CKD or MBD risk, aims to identify those who require
describe a patient who has experienced three or more pharmacological prevention and/or special attention
episodes of passing stones in a 5‑year period176. Other during follow‑up care. Risk factors for MBD in stone
patients with some stone recurrence can be identified as formers include distal renal tubular acidosis (complete
occasionally recurrent or sporadic stone formers. or incomplete), medullary sponge kidney, primary
Although the number of stones formed by a patient hyperparathyroidism, malabsorptive syndromes, fast­
denotes the clinical activity of the stone disease, the con­ ing hypercalciuria and genetic disorders (for example,
cept of metabolic activity of the stone disease is different. sodium-phosphate transporter disorders)179. In calcium
Metabolic activity in stone disease refers to an ongoing stone formers, the risk of developing CKD is increased
crystallization-driving process that will probably lead to as a result of systemic or renal tubular disorders, high
new stones or to the growth of the existing stones. It is recurrence rates or in those with MBD. All patients
this concept that should ideally drive the clinician’s deci­ with non-calcium nephrolithiasis are at risk of develop­
sion on how to treat the patient. Unfortunately, we do ing CKD and recurrent stones. In first-time or sporadic
not have reliable metabolic markers. Patients with their stone formers, stone composition, family and personal
first (and most likely only) stone might have the same recall, imaging and laboratory data would suggest that
metabolic urine abnormalities observed in those with these individuals have disorders that put them at a
recurrent stones177; the same is probably the case for the higher risk of CKD (BOX 2). Children (and young adults)
occasional recurrent stone former. Thus, we must rely on should be considered at high risk of developing CKD
a surrogate marker of metabolic activity: clinical activity, and MBD180.
which has limitations. A recurrent calcium stone former, CKD can also result from whatever has caused stones
who has passed many new stones in a relatively short to precipitate, renal damage from stone complications
period of time, should be considered as metabolically (pyelonephritis and obstructive nephropathy) or can be
active, as should a patient who exhibits stone growth on the result of urological treatment 181. Referral to a neph­
serial imaging even if it is their first stone. By contrast, rologist is highly recommended in patients with stones
a patient with multiple kidney stones discovered on the and CKD.
same occasion could not be clinically active because
the cluster of stones could have formed previously in a Stone analysis. Knowledge of stone composition is
short time-frame as a result of time-concentrated, non- fundamental in non-calcium stone formers; in these
long-lasting and unique metabolic derangements. Thus, cases, it is a formidable pathway to diagnosis. For
when the stones formed, the patient was metabolically instance, finding that a stone is pure cystine or con­
active but might not be at the moment of diagnosis. tains even a small percentage of cystine is diagnostic of

10 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

cystinuria (an inherited autosomal recessive disease). also focus on CKD, MBD and ESRD. Examples that
Furthermore, discovering a composite stone of struvite such an approach is possible, at least with respect to
and CaOx is a strong hint of the existence of a meta­ MBD, have been reported187,188. Comprehensive meta­
bolic disorder 182. In calcium stones, the precise recog­ bolic evaluation is followed by a range of preventive
nition of the mineral components is much less relevant. measures, including specific pharmacological interven­
The identification of apatite as a major component of a tions in addition to advice for a change in lifestyle and
stone could suggest renal tubular acidosis or primary nutritional habits.
hyperparathyroidism, but this is not always the case.
Finding brushite, another crystalline form of CaP, sug­ Recurrent calcium stones. Although prevention of new
gests more-active stone disease that is less amenable to calcium stones is possible, there is no pharmacological
preventive treatments and increases the risk of CKD183. treatment that can dissolve existing calcium stones. In
Most calcium stones have mixed composition; this is non-idiopathic calcium nephrolithiasis, the primary
believed to depend on the pathogenetic mechanisms conditions should be addressed with specific treatments.
involved in lithogenesis (that is, low levels of apatite In these cases, preventive measures are supportive. In
in a CaOx stone suggests lithogenesis on Randall’s the majority of patients with idiopathic stone disease,
plaques) and/or on the superimposition of a different behavioural and nutritional interventions are poten­
form of lithogenesis (for example, a mixed CaOx and tially helpful and should be the first step of stone pre­
uric acid stone). The calcium composition of a stone vention189. Nutritional advice for patients with calcium
can be deduced by the plain abdominal X‑ray imaging
of radiopaque stones. Dual-energy CT shows promise
as a diagnostic tool for uric acid stones184. Box 2 | Stones and risk of chronic kidney disease
The conditions reported in this box complicate more or
Laboratory evaluations. In all stone formers, the initial less frequently with chronic kidney disease (CKD). The
metabolic work up is best performed ≥3 weeks after different categories of CKD risk are arbitrarily attributed
the acute episode, such as renal colic with or without by the author (G.G.) on the basis of personal experience
stone passage, obstruction, stone removal or concomi­ and the literature. No or very few studies have
tant urinary tract infection. In calcium stone formers, investigated the prevalence or incidence of CKD in the
the diagnostic algorithm differs according to the his­ listed conditions; for some of the rarest stones, the risk
tory of previous stones and/or co-morbidities (TABLE 1). attribution was based on few case reports.
Conversely, the diagnostic algorithm for individuals Possible risk of CKD
with non-calcium stones (TABLE 2) is dependent on • Xanthine stones
stone composition only because many of these con­ • Indinavir stones
ditions are recurrent and/or have a high risk of CKD • Distal renal tubular acidosis (incomplete)
and ESRD. If the stone is not analysed or its composi­ • Primary hyperparathyroidism
tion is unknown, the patient should be investigated as
• Eating disorders and laxative abuse
a calcium stone former (TABLE 1). Children who have
• Medullary sponge kidney
passed stones with calcium or of unknown composi­
tion should be evaluated intensively as recurrent stone Moderate risk of CKD
formers. However, in these patients, 24‑hour urine col­ • Brushite stones
lection and MBD-related evaluations could be difficult • 2,8‑Dihydroxyadenine stones
to undertake; thus, analyses of the spot urine might • Sarcoidosis
be sufficient. • Pyelo-ureteral or ureteral strictures
High risk of CKD
Genetic investigations. A recent study has concluded
that in up to 11.4% of adult and 20.8% of paediatric • Cystine stones
patients with renal stones and nephrocalcinosis, pre­ • Struvite stones
viously unsuspected monogenic disorders such as • Stones in a single kidney
cystinuria were eventually diagnosed185. This finding • Distal renal tubular acidosis (complete)
is at odds with several investigations that did not find • Secondary hyperoxaluria (bariatric surgery,
carriers of mutated genes in large series of patients inflammatory bowel disease, bowel resection and
with renal stones and/or hypercalciuria186. Accordingly, malabsorptive syndromes)
a systematic investigation of patients with stones for • Other forms of nephrocalcinosis (often associated with
genetic mutations is probably not warranted. Instead, genetic conditions with hypercalciuria)
these findings implicitly suggest that patients should be • Anatomical abnormalities of the kidney and urinary
thoroughly investigated for the clinical manifestations of tract (for example, horseshoe kidney, ureterocele
inherited disorders180. and vesicoureteral reflux)
• Neurological bladder
Prevention Very high risk of CKD
Stone formation can be a recurring disease that may • Primary hyperoxaluria
have severe consequences, such as CKD, MBD and
• Autosomal dominant polycystic kidney
ESRD. Thus, the aim of preventive strategies should

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 11


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Table 1 | Laboratory assessment of calcium stones


Patient category Stone Blood‡ 24‑hour urine First morning spot Others
analysis* collection§ urine
First-time or sporadic Yes Creatinine, calcium, phosphate Not suggested pH, routine urinalysis According to the blood
stone former and potassium levels and eGFR and urine culture findings
First-time or sporadic Yes Creatinine, calcium, Oxalate, calcium and Calcium to creatinine Renal acidification test¶,
stone former at risk of phosphate, potassium and citrate levels and volume ratio|| bone turnover markers#
CKD and/or MBD bicarbonate levels and eGFR and bone densitometry
Recurrent stone Yes Creatinine, calcium, Oxalate, calcium, pH, routine urinalysis, Renal acidification test¶,
former phosphate, potassium, phosphate, citrate, urine culture, cystine parathyroid hormone
bicarbonate and uric acid uric acid, urea, sodium (qualitative) levels levels‡‡, bone turnover
levels and eGFR and potassium** levels and calcium to markers#,§§ and bone
and volume creatinine ratio|| densitometry
CKD, chronic kidney disease; eGFR, estimated glomerular filtration rate; MBD, metabolic bone disease. *Stone (and stone fragment) analysis should be performed
on all available stones. ‡The determination of ion concentrations aims to recognize secondary forms of urolithiasis and risk of CKD and MBD. High calcium and/or
low phosphate levels suggest hyperparathyroidism; low or marginally low potassium levels suggest renal tubular acidosis. Low bicarbonate levels suggest renal
tubular acidosis. §Data from at least two 24‑hour urine collections in the usual conditions of diet and fluid intake should be obtained. Collections should be
performed in plastic bottles following laboratory instructions. The accuracy of the collection could be checked by evaluating creatininuria. ||The urinary calcium to
creatinine ratio in a morning (fasting) spot urine sample is performed in patients with hypercalciuria; it is a marker of the obligatory urinary loss of calcium.
Values of >0.25 mg per mg suggest an increased bone turnover. ¶The test is optional to confirm renal tubular acidosis, which should be suspected in patients with
or without systemic acidosis and recurrent nephrolithiasis of pure calcium phosphate (CaP) stones or stones mainly composed of CaP. Renal tubular acidosis should
also be considered in recurrent calcium stone formers (both calcium oxalate and CaP) with hypocitraturia and a urinary pH of >5.8 and in patients with
nephrocalcinosis. #Urinary hydroxyproline, or urinary or serum deoxypyridinoline crosslinks. Assessment can help to monitor the effects of treatment on MBD280.
**The 24‑hour urinary excretion of sodium, potassium, phosphate and urea provide important information on the nutritional habits of the patient, and the daily
protein intake can be estimated. ‡‡When blood tests suggest hyperparathyroidism, serum parathyroid hormone levels should be measured and parathyroid imaging
should be performed. §§In patients with hypercalciuria.

stones include increased water intake (>2 l per day and increased fluid intake have not been successful in
>3 l per day in the summer) to provide 24‑hour diuresis prevent­ing stone recurrence. Allopurinol or febuxostat
of >2 l190, and maintenance of a balanced diet with cal­ could be useful in patients with calcium stones who are
cium intake not <800–1,000 mg per day, reduced meat hyperuricosuric195; the former was shown to be effect­
and poultry intake (≤0.8 g per kg of body weight)191, ive in reducing urinary uric acid and stone recurrence
reduced salt intake (<2 g per day, which is equivalent to in hyper­uricosuric CaOx stone formers without other
5 g of table salt (sodium chloride)), avoidance of excess metabolic abnormalities197. Although no data support
food intake, increased vegetable consumption192 and its use, the hypouricosuric effect of febuxostat suggests
avoidance of soda beverages193. that this drug could be ­effective in stone formers who
Low calcium diets should be avoided in the majority cannot tolerate allopurinol198.
of patients because they increase intestinal absorption Citrate (generally potassium citrate) use to increase
and urinary excretion of oxalate, thereby increasing citraturia, which raises the inhibitory activity against cal­
lithogenesis194; furthermore, such diets can cause or cium crystallization199, has been shown to be effective in
worsen MBD in calcium stone formers. Low oxalate two randomized trials200,201. Citrate is indicated in those
diets are difficult to attain because of the presence of with recurrent CaOx stones with decreased urinary cit­
oxalate in many common foods. Only foods with very rate excretion; in patients with complete or incomplete
high oxalate content should be limited or avoided (see distal renal tubular acidosis, chronic diarrhoeal states,
https://regepi.bwh.harvard.edu/health/Oxalate/files). drug-induced or diet-induced hypocitraturia; and in
The concomitant consumption of foods that are rich in patients with MBD who form stones. In general, potas­
oxalate and calcium is a possible strategy to decrease the sium citrate is preferred to sodium citrate because it
absorption of oxalate195. attenuates calciuria and, therefore, is likely to be more
Drug treatment could be considered if stones con­ effective in preventing calcium stones202. However, some
tinue to recur despite the above measures, or if the concern pertains to overtreatment with citrate, which
CKD and/or MBD risks are considerable, or in cer­ in theory might increase the risk of forming new CaP
tain groups of people (for example, flying airline per­ stones because it raises urinary pH (via its metabolism
sonnel) and in those who have severe urine metabolic to bicarbonate by the liver). However, in patients with
abnormal­ities. For example, thiazides reduce calciuria medullary sponge kidney and/or distal renal tubular
and might improve bone mineral density 179 and should acidosis and pre-existing high urinary pH, kidney stone
be considered in patients with high or relatively high recurrence rate is decreased rather than increased after
urine calcium levels and recurrent calcium stones. treatment with citrate203,204.
However, thiazides have also been shown to decrease
stone activity in individuals with normocalciuria196. Uric acid stones. To prevent frequently occurring uric
Indeed, the American Urological Association guide­ acid stones, uric acid supersaturation in the urine must
lines195 suggest that lowering calciuria with thiazides be decreased. This can be achieved by increasing uri­
might be effective regardless of the absolute rate of nary volume (>2 l per day), increasing urinary pH to
calcium excretion. Thiazides are appropriate for both approximately 7.0, decreasing uricosuria and admin­
CaOx and CaP stones when dietary measures and istering sodium bicarbonate or potassium citrate.

12 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Table 2 | Laboratory assessment of non-calcium stones


Stone type Stone Blood 24‑hour urine collection* Spot urine
analysis
Struvite stone‡ Yes, stone Creatinine, bicarbonate, calcium, potassium Calcium and oxalate levels, pH pH, routine urinalysis§ and
culture and inorganic phosphate levels and eGFR and volume urine culture
Matrix stone|| N/A Creatinine levels and eGFR Volume and pH pH, routine urinalysis and
urine culture
Uric acid stone Yes Creatinine and uric acid levels and eGFR Uric acid levels, pH and volume pH and routine urinalysis§
Cystine stone Yes Creatinine levels and eGFR Cystine levels (quantitative), pH# and routine urinalysis§
proteinuria¶, pH and volume
Other purine stones Yes Creatinine and uric acid levels and eGFR Uric acid levels and volume pH and routine urinalysis§
(such as xanthine and
2,8‑dihydroxyadenine)
Drug stones Yes Creatinine levels and eGFR pH and volume pH and routine urinalysis§
eGFR, estimated glomerular filtration rate; N/A, not applicable. *Data from at least two 24‑hour urine collections in the usual conditions of diet and fluid intake
should be obtained. Collections should be performed in plastic bottles following laboratory instructions. The accuracy of the collection could be checked by
evaluating creatinine levels in the urine. ‡Given that struvite stones could be a complication of common calcium stones, especially in men, the common metabolic
risk factors for calcium stones should be examined. §Typical crystals of the corresponding condition can be observed. ||It has the same background conditions of
infection stones. ¶Total proteinuria has to be checked in patients who are treated or are going to be treated with 6‑mercaptopropionyl glycine or d‑penicillamine.
#
The determination of pH in spot urine during the day is useful for treatment guidance.

Although not supported by data from clinical trials, Struvite stones. The risk of recurrence of struvite stones
allopurinol or febuxostat can be used if the patient has is high. Particularly in men, these ‘infection stones’ are
hyper­uricosuria and dietary measures fail to normalize a complication of common calcium stones182; thus, it is
­urinary uric acid205. important to look for and treat the metabolic abnor­
malities that are typical of calcium nephrolithiasis in
Cystine stones. Reducing the concentration of cystine these patients. Complete removal of struvite stones is
in the urine and increasing its solubility will prevent a prerequisite for successful recurrence prevention.
stone formation in this highly recurrent stone dis­ Indeed, it is almost impossible to cure infection in the
ease. The preventive strategy involves increasing water presence of the stone because it represents a micro­
intake to >3 l per day and the administration of sodium organism reservoir. After clearing the stones, long-
bicarbo­nate or potassium citrate to raise urinary pH and term (several months) targeted antibiotic treatment
increase the solubility of cystine. Early in the treatment might be necessary to keep urine sterile and prevent
course, urinary pH should be checked multiple times new stone formation205.
per day to titrate the quantity of alkali; at a later stage of
treatment, the urinary pH should be monitored less fre­ Management
quently. The target pH is 7.0–8.0 (REF. 195). If cystinuria Surgical management
is >300 μmol per mmol of creatinine (>2,600 μmol per g Over the past 30 years, the management of paediatric
of creatinine) or if the aforementioned measures fail to and adult patients with symptomatic kidney stones has
prevent new stones, 6‑mercaptopropionyl glycine can evolved from open surgical lithotomy to minimally
be administered, with d‑penicillamine as an alternative invasive endourological approaches. The three most
treatment 195. Given that both drugs can cause protein­ common treatment modalities for renal stones include
uric glomerular diseases, urine should be periodically extracorporeal SWL (40–50% worldwide use), rigid or
monitored for proteinuria. Ideally, cystine solubility flexible retrograde ureteroscopic stone fragment­ation
should be assessed; numerous methods have been pro­ and retrieval (30–40%) and PCNL (5–10%). Each of
posed206–208, but few laboratories have this capacity. Thus, these therapies has its own particular adverse-effect
generally, treatment is guided by cystine stone formation profile and expected success rate depending on the
and/or growth. experi­ence of the treating physician, stone factors (size,
Patients with cystine stones require close follow‑up location and composition) and patient characteristics
because of the high metabolic activity of the disease (with (body habitus, medical co-morbidities and anatomy).
a very high risk of stone recurrence, the rapid growth of With appropriate counselling and proper procedure
these stones, staghorn stone formation, need of surgical selection, patients should expect high stone clearance
procedures and CKD occurrence) and because of the rates, low associated morbidity and quick recovery times.
possible adverse effects of treatment. Indeed, a French A general decision-tree algorithm of the most common
cohort study of 442 patients showed that 26.7% had of these approaches, stratified by stone l­ocation, stone
CKD and only 22.5% had a (normal) estimated glomeru­ size and stone density, is presented in FIG. 7.
lar filtration rate (eGFR) of >90 ml/min/1.73 m2; a his­
tory of staghorn stones and multiple open surgical stone Shockwave lithotripsy. SWL involves the non-invasive
procedures constituted a relevant risk factor for CKD delivery of high-energy acoustic waves that fragment
and nephrectomy 209. Accordingly, patients should be a kidney stone. The shockwave, created by electro­
monitored for urinary pH and volume. hydraulic, electromagnetic or other types of energy

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 13


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Symptomatic kidney stone anatomical factors (long, narrow lower-pole infundibu­


lum) and the dependent position of the calculi limit­
ing the passage of fragments213. Owing to these factors
Elective stone removal
and the poor stone passage rates for larger stones, most
clinicians do not perform SWL for lower-pole stones that
Lower pole Upper or middle pole are >1 cm in size214,215.
In addition to lower-pole limitations, SWL might
require repeated treatments to match the efficacies of
<1 cm 1–1.5 cm >1.5 cm <2 cm ≥2 cm or staghorn PCNL and ureteroscopic retrieval211,216. This retreat­
ment risk is associated with obesity (a body mass index
Low High
of >30) and with extremely dense stones. Patients who
attenuation attenuation are obese are believed to have lower SWL success rates
because their kidneys — and, accordingly, stone depth
— exceed the lithotripter focal length (shockwaves
only penetrate 12–14 cm) and/or their body habitus
prevents adequate stone visualization at the time of
lithotripsy 217,218. Dense stones, such as cystine, brushite
or COM, are more resistant to SWL fragmentation. As
Shockwave lithotripsy Ureteroscopic retrieval Percutaneous nephrolithotomy stone composition is usually unknown before surgery,
CT stone attenuation values in Hounsfield units (HUs)
Stone location Stone size or morphology CT evaluation Treatment are commonly used as preoperative surrogates for stone
density. Although variable, most urologists use high
Figure 7 | Algorithm for the most common approaches to surgical
Nature Reviews treatment
| Disease of
Primers attenuation values of >1,000 HU as a predictor of renal
kidney stones. The decision of which surgical strategy to use is dictated by the location
stone disintegration failure that should be considered
of the stone (with lower-pole stones being more difficult to treat), stone size and
before undertaking SWL219.
stone density.
Overall, most of the SWL shortcomings are sur­
mounted by the excellent quality-of-life (QOL) meas­
sources, travels through the patient and is focused on ures and low morbidity associated with the procedure.
the stone using an acoustic lens. When these shock­ Patients who undergo SWL have repeatedly been shown
waves approach and pass through the calculus, energy to have faster return to work, shorter recovery times
is released resulting in internal structure disruption and and higher satisfaction scores than those who undergo
stone fragmentation. Fluoroscopic or ultrasonographic ureteroscopic retrieval, especially if SWL occurs with­
guidance is routinely used during SWL to aid in calculus out stenting 215. SWL also has a low complication pro­
targeting and for precise acoustic-wave focusing. Deep file, including a 5% rate of steinstrasse (that is, stone
sedation or general anaesthesia is commonly used for fragment build‑up within the ureter) and a 2% rate of
intra-­operative analgesia, as well as to control respir­ urinary tract infection (TABLE 3). Major complications
atory renal movement. Although recent Canadian and such as sepsis or profound haemorrhage are rare but
US medical claims data have demonstrated a marked deserve mention. The development of sepsis following
decrease in use over the past decade, SWL remains the SWL is low in absolute terms (<1% of patients), but is
most commonly performed endourological kidney stone considerably higher in the presence of staghorn or col­
procedure worldwide210,211. onized stones (up to 10% of patients)220. To mitigate this
The success of SWL is typically determined risk, patients with urinary obstruction or positive urine
1–3 months after the procedure by plain abdominal cultures before SWL should be completely treated. The
X‑ray with or without renal ultrasonography. As small majority of patients who undergo SWL develop tran­
residual fragments of <4 mm in size within the kidney sient haematuria that resolves within days, and imaging
are considered to be passable, patients with these clin­ studies in asymptomatic patients post-procedure have
ically insignificant stones are often referred to as stone shown a haematoma rate of 25%221. However, sympto­
free by most classification systems212. This misnomer matic fluid collections (perirenal, subcapsular or intra­
becomes confusing when comparing stone-free rates renal haematomas) are rare (<1% of patients), and the
among studies using plain abdominal X-ray versus CT rate of post‑SWL blood transfusions are very low (<0.2%
imaging because CT is more sensitive than X‑ray or renal of patients).
ultrasonography for assessing residual kidney stones.
Despite these discrepancies, stone-free rates for SWL are Ureteroscopic fragmentation and retrieval. Uretero­
considered equivalent to those of ureteroscopic retrieval scopy consists of retrograde passage of an endoscope
(50–80% success) for small radiopaque stones (<2 cm in from the urethra proximally towards the affected ure­
size) located in non-dependent portions of the kidney ter and kidney, enabling access to the stone as well
(upper pole, middle pole or renal pelvis; TABLE 3). Stones as delivery of other instruments, such as guidewires,
located in the lower pole of the kidney remain the most ­balloon dilators, laser fibres and baskets. Although
daunting clinical challenge for endourologists perform­ fairly non-invasive, ureteroscopy requires spinal or
ing SWL. Multiple explanations have been offered for general anaesthesia to minimize pain and the vis­
the poor stone-free rates for lower-pole stones, including ceral response to ureteral and renal dilation. Rigid

14 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

ureteroscopes are reserved for distal ureteral stones, Compared with SWL, ureteroscopy is associated
whereas flexible uretero­scopes, with their deflection with higher procedure-related complication rates and
ability, are used to reach the extremes of the renal longer hospital stays (TABLE 3). Many of the symptoms
collect­ing system and negotiate access to anatomically that raise complication rates are secondary to the ure­
difficult renal calyceal variants. Some urologists place teral stent that is left in place following the procedure213.
ureteral access sheaths (long reinforced hollow tubes) These small, hollow polyurethane tubes are designed
from the urethra to the renal pelvis to enable repetitive with proximal and distal coils to maintain their position
passage of the ureteroscope while minimizing uro­thelial within the kidney and the bladder. Unfortunately, the
trauma. These sheaths also enable the continuous flow distal bladder coil can be felt by the majority of patients,
of irrigation fluid, improving stone visualization and causing haematuria and irritative voiding symptoms
facilitating a low-pressure system. Although flexible that range from mild to intolerable215. Ureteroscopy
electrohydraulic lithotripters are available, the holmium is favoured over SWL in the setting of multiple or
yttrium-aluminium-­garnet (Ho:YAG) laser remains the radio­lucent stones (stones that are not visible on plain
preferred method of lithotripsy in most centres in devel­ film), hydronephrosis, obesity or high-density stones
oped countries owing to its rapid absorption in water (holmium lasers are able to fragment all stone types).
and minimal tissue penetration213. Ureteroscopy for renal stones during pregnancy or in
A recent meta-analysis of seven large randomized patients with bleeding diathesis is unusual but would
controlled trials totalling >1,200 patients demonstrated be considered the safest approach, if necessary (TABLE 3).
that ureteroscopic retrieval achieved a higher ureteral and
kidney stone-free rate and a lower need for retreatment Percutaneous nephrolithotomy. PCNL involves the
than did SWL222. As such, for ureteral stones <10 mm direct passage of an endoscope percutaneously through
in size, SWL and ureteroscopy are considered first-line skin, muscle and perirenal fat, and into the kidney and
therapy. For ureteral stones >10 mm in size, ureteroscopic is generally performed for stones >2 cm in size. With the
fragmentation results in higher stone-free rates and fewer patient under general anaesthesia in either the prone or
procedures223. For renal stones in non-­dependent loca­ supine position, the kidney is located using anatomical
tions such as the lower pole, uretero­scopy stone-free markings. Renal access is achieved under fluoroscopic
rates are comparable to those of SWL, if not slightly and/or ultrasonographic guidance and can be combined
­better (TABLE 3). Like SWL, uretero­scopic manage­ment of with endoscopic or radiographic imaging techniques.
lower-­pole stones is frequently more challenging than for For the posterior approach, renal access below the 12th
stones located elsewhere in the kidney. Not only can acute rib is preferred to avoid the pleural cavity and intercostal
infundibular angles make deflection and manoeuvr­ing of vessels and nerves. A needle puncture into a posterior
the scope difficult but also the passage of the accessory calyx rather than the renal pelvis is also recommended
instruments through the ureteroscope working channel to avoid posterior renal artery or vein branch injury.
reduces the ability of the surgeon to actively deflect the This ‘access’ can be performed preoperatively as an out­
ureteroscope, creating a scenario in which a lower-pole patient procedure in an angiography suite (percutaneous
stone can be visualized but not manipulated213. With nephrostomy tube placement) or preferably during the
the advent of smaller flexible endoscopes and tipless same setting as PCNL.
stone baskets, many endo­urologists relocate lower-pole Worldwide, approximately 70% of all renal access for
stones into a more-favourable upper‑pole location PCNL occurs in the lower-pole calyx, and prior cysto­
before fragmentation224. scopic placement of a ureteral catheter for retrograde

Table 3 | Indications, success rates, risks and contraindications for renal stone procedures
Location Stone Success Risks Absolute and relative contraindications
size rate
Shockwave lithotripsy
Upper pole, middle ≤2 cm 50–70% • >15% require retreatment Coagulopathy, pregnancy, obesity, multiple
pole and renal pelvis • 2–5% experience steinstrasse or radiolucent stones, obstruction, cystine
• <2% experience obstructions composition and dense stones*
Lower pole ≤1 cm 50% • 2% experience urinary tract infections, haematomas
and sepsis
Ureteroscopy
Upper pole, middle <2 cm 60–80% • >25% experience stent pain Bladder reconstruction, ileal conduit, renal
pole and renal pelvis • 5% experience urinary tract infections, ureter transplantation, large median prostate
injuries or require retreatment lobes, ureteral stricture, re-implantation
Lower pole ≤1.5 cm 50–60% • <1% experience sepsis or avulsion or pathology
Percutaneous nephrolithotomy
Upper pole, middle >2 cm 80–95% • 2–5% experience sepsis or require blood transfusion Coagulopathy, pregnancy, high pulmonary
pole and renal pelvis • 1% require intubation and 0.3% experience arterial risk, bowel overlying renal access and an
embolization inability to tolerate prone position
Lower pole ≥1.5 cm 70–80%
*Defined as stones of >1,000 HU on CT.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 15


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

injection of a contrast has been shown to dramatically Hydration should be normal and intravenous fluids
improve targeting and access to the kidney 225. Once are only indicated in the case of protracted vomiting
entry into the kidney has been achieved, a guidewire because it does not favour stone expulsion but instead
is then advanced antegrade to the bladder to maintain increases pain and the risk of complications (renal pelvic
access and enable passage of either sequential or balloon rupture and urine extravasation)233. α‑Adrenergic recep­
dilators to expand the tract. After dilation and placement tor antagonists (mainly tamsulosin)234,235 and calcium
of a working sheath, both rigid and flexible nephro­ channel blockers234 have been demonstrated to be an
scopes can be passed into the kidney for stone removal effective medical expulsive therapy, believed to be due
and lithotripsy. After the procedure, nephrostomy tubes to their ability to dilate the distal ureter and increase the
with or without a ureteral catheter or a ureteral stent probability of spontaneous stone passage. The efficacy
by itself (‘tubeless’ stents) are left in place to facilitate of these agents in promoting the passage of small distal
­maximal renal drainage226. ureteral stones (<5 mm in size) has recently been decried
PCNL generally offers very high stone-free rates by two well-designed, randomized, placebo-controlled
in the 80–90% range, which can be attributed to the trials, one of which found efficacy only for larger stones
shorter working distance, the introduction of relatively (≥5 mm in size) and the other found no efficacy for
large working sheaths (30 French or 10 mm) and more-­ stones of any size236,237.
effective instruments for the removal of larger stone
fragments (TABLE 3). For patients with a stone burden Oral and percutaneous dissolution therapy of stones.
of >2 cm or staghorn calculi, PCNL is considered the Oral dissolution of existing stones is generally effective
standard of care, mirroring the successes of open surgery only with uric acid stones. Two-thirds of these stones
while decreasing the length of hospital stay by 75%227. can be at least partially dissolved by following the same
PCNL is regarded as being more effective than uretero­ rules suggested for their prevention: modulating the pH
scopic procedures or SWL for most stones, but is more of urine to 7.0, increasing urinary volume and decreas­
invasive. For ureteral stones, antegrade access to the ure­ ing uricosuria with allopurinol or febuxostat 238,239. Few
ter can also be achieved with high success depending on obser­vations also suggest that cystine stones might be
the ureteral diameter. However, ureteral stone manage­ amen­able to oral chemolysis with 6‑­mercaptopropionyl
ment is typically reserved for less-invasive approaches glycine, high urine output and alkalinization240. Finally,
and smaller stones. the use of percutaneous solutions, such as 10% hemi­
Owing to its more-invasive nature, especially for acidrin or Suby’s solution, to turn insoluble kidney
larger and branched stones, PCNL has a greater compli­ stones into more-water-soluble forms was popularized
cation profile than less-invasive endoscopic techniques, in the 1960s but is rarely used today. Although some­
including a 2% sepsis risk and a 5% risk of requiring what effective for infection, uric acid, cystine and brush­
blood transfusion 228. Other complications are also ite stones, percutaneous dissolution of kidney stones
reported with varying frequency, such as pneumothorax is labour intensive and considerably less effective than
(1% of patients, usually with upper-pole access), arterial contemporary minimally invasive removal techniques241.
injury requiring embolization (0.3% of patients) and
colonic perforation (rare)213. As stone clearance relies Quality of life
on visibility, nuisance bleeding can obstruct visibil­ As indicated earlier, the prevalence of urolithiasis and
ity, requiring procedure termination, placement of a nephrolithiasis has increased over the past decades and
nephrostomy tube and a return operating room appoint­ affects approximately 9% of the American adult popula­
ment at a later date. Anatomical limitations, such as tion12. Although many kidney stones remain asympto­
­narrow infundibulum or entrances at acute angles to the matic for long periods, the incidence of stone episodes,
tract, can also make it difficult to access the entire caly­ such as pain, infection or obstruction, has increased27.
ceal system through one tract. In these cases, a second Furthermore, following an initial stone event, >50%
or even third access tract is required to obtain complete of patients experience a recurrent stone in the follow­
stone clearance, occasionally even requiring a combined ing 5 years242. Thus, for these patients, treatment (both
retrograde approach. medical and surgical) and implementation of lifestyle
­modifications can significantly affect patient QOL.
Medical management The most commonly used QOL questionnaire in the
Treatment of renal colic and medical expulsive therapy. literature is the 36‑Item Short-Form Health Survey (SF‑36)
Treatment of the pain associated with kidney stones questionnaire, which consists of self-administered ques­
(renal colic) is based on the use of NSAIDs as a first tions (a total of 36) divided into eight domains including:
choice in the absence of contraindications229,230 and, in physical function, role limitations due to physical health
case of failure in relieving pain, opioids. Intravenous problems, bodily pain, general health perception, vitality,
para­cetamol (acetaminophen) also seems to be as effect­ social function, role limitations due to emotional problems
ive as morphine231. The use of antispasmodics does not and mental health243. The eight domains are scored from
seem to have a significant effect 229,232. If analgesia c­ annot 0 (worst) to 100 (optimal). Three US studies using this
be achieved with the previous measures, drainage of questionnaire compared stone formers to healthy adults
the renal pelvis through percutaneous nephrostomy or and found lower scores in stone formers in two (general
ureteral stenting and eventually stone removal should health perception and bodily pain), five (physical function,
be performed. role limitations due to physical health problems, general

16 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

health perception, social function and role limit­ations due Residual fragments in the kidney are frequently
to emotional problems) and six (all except role limitations encountered, not only following SWL255–257 but also after
due to emotional problems and mental health) of the other minimally invasive procedures258–262. The course
eight domains, respectively 244–246. Among stone formers, of residual fragments is variable, but their presence is
one study reported that cystine stone formers had the problematic for both the patient and the treating physi­
worst QOL owing to role limitations caused by emotional cian and requires careful follow‑up or, often, repeated
problems247. When comparing scores between men and intervention. Given that most residual fragments are
women, most of the studies reported lower QOL in female composed of CaOx, will it be possible to accomplish
stone formers244–247. However, these results have to be inter­ fragment elimination by enzymatic dissolution with
preted with caution because the timing of the question­ oxalate decarboxyl­ase and formate dehydrogenase262?
naire in relation to the last stone event has a signifi­cant Although such an approach certainly can be carried out
effect on the SF‑36 score. When adjusting results accord­ by percutaneous irrigation in selected patients, the ques­
ing to the interval from the last stone episode, patients tion is whether or not it might be possible to incorporate
with a recent stone event (within the past month) had the enzymes in stents or other intracorporeal devices
lower scores (role limitations due to physical health with a slow-release function. Furthermore, the medical
problems, bodily pain, general health perception and advances of managing patients with stone disease have
social function) than the general American population, remained behind the times. The literature currently
whereas patients with a remote stone event (>1 month indicates a faint awaken­ing interest in factors that are
ago) had scores simi­lar to the general ­population with the responsible for stone formation and recurrence preven­
exception of general health perception244,245. tion242,263–265. However, the enthu­siasm among urologists
In addition to QOL evaluation for urolithiasis, it is to deal with metabolic and m ­ edical aspects of stone
also important to evaluate QOL in regard to m ­ anagement ­disease, ­nevertheless, is too weak.
because some patients might require surgical treatment. Another therapy is that of inversion (head down)
The current guidelines for the management of uro­lithiasis therapy, which is a procedure carried out after stone
are based on the stone-free rates between the different disintegration with the aim of favourably changing the
surgical modalities but do not incorporate QOL as an position of the lower calyces. This step would facilitate
additional end point. One study directly compared SWL, fragment elimination by gravitation. The altered position
ureteroscopy and PCNL a few months after surgery to of the kidneys is often combined with vibration on the
a control group using the SF‑36 questionnaire248. The body surface to add some kinetic energy. This treatment
global score after treatment was higher in the patients approach can be used in patients with residual fragments
under­going surgery than in controls for physical func­ in the lower pole of the kidneys266–269, although the results
tion, role limitations due to physical health problems, have been conflicting 270.
vitality, role limitations due to emotional problems and In another recently published report, ultrasonic
mental health; the only exception to this finding was for propulsion was used successfully to facilitate fragment
bodily pain, which was lower after treatment248. Two other elimination, which is a new and interesting concept 271.
studies did not find differences between patients who Although promising, this technology is not US FDA
underwent surgical treatment and controls249,250. When approved and is being performed in patients only in
comparing the different surgical modalities, patients experimental protocols. To facilitate fragment elimin­
who underwent PCNL had worse scores in all QOL ation, a pharmacological agent could be used to pro­
domains, except for bodily pain, whereas the patients mote muscle contraction in the calyx. Poor local smooth
who underwent SWL reported the highest QOL scores248. ­muscle activity might, hypothetically, be associated with
Furthermore, increasing numbers of surgical procedures poor clearance of fragments. Whereas fragments from
were associated with lower QOL244,248. Stent placement, small disintegrated stones are relatively easily eliminated,
often used at the end of the surgical pro­cedure, has fragments from large stones are not. A possible explan­
also been reported to impair QOL without a difference ation for this discrepancy is that the large stone in the
between ureteral and nephrostomy stents244,251–253. In the calyx has hindered muscle contraction. This research area
case of residual fragments, one study found that fragments has not been addressed hitherto272.
of >4 mm in size might affect QOL254. Finally, dietary It is interesting to note that present treatment trends
changes and medical therapy have not been reported to comprise relatively aggressive, albeit minimally inva­
affect QOL except for alkali therapy 244,245. sive, methods for fragment removal. Such a step is of
course necessary in patients with infection stones and
Outlook stones composed of uric acid, cystine and brushite, but
John Wickham (a pioneer of stone surgery) once said at the question is whether such procedures are necessary
an early SWL congress — when he realized the conse­ in asymptomatic patients with minor residual frag­
quences of the procedure — that we had made an almost ments composed of CaOx. Given the high recurrence
unbelievable journey from open surgery via minimally risk that these patients experience despite meticulous
invasive techniques to non-invasive stone removal. stone clearance, this is a pertinent question regarding
Although this development reflected a giant step in stone overtreatment. In an epidemiological study 273, a 10‑year
treatment, his vision was that the final goal would be recurrence risk of about 25% was shown for patients who
entirely medical. Three decades later, it is obvious that we had formed only one stone at the start of follow‑up, but
still have a long way to go before that dream comes true. was approximately 70% for patients with more than one

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 17


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Production of promoter Reduce or eliminate oxidative stress current treatment options, we are only addressing the
final steps in the chain of abnormal crystallization events
Reduce intratubular in idiopathic patients who form calcium stones. Whereas
CaP nucleation CaOx nucleation* APCaP and/or APCaOx recurrence prevention in patients who are stone free can
be accomplished by reducing supersaturation of CaOx in
the final urine; how can we approach the remaining CaP
CaP precipitation in the Intratubular deposits? Would it be possible to decrease CaP super­
interstitial tissue transport of CaP Dilute urine in
the thin and thick saturation in the nephron urine and interstitial tissues
segments of with a short-term intensive treatment and hope for nat­
the loop of Henle ural elimination of CaP by the action of, for example,
CaP plaques CaP plugs
macrophages49,277? Or can we interfere with any other
step in CaP precipitation that would eliminate this risk
CaP dissolution in acidic urine Increase pH in final urine factor? In this regard, radiological improvements might
enable the detection and quantification of papillary
CaP deposits278.
CaOx nucleation Reduce APCaOx in final urine The initiation of stone formation in most patients is
an occasional event. Accordingly, optimal compliance
CaOx aggregation Increase urinary pH, can only be expected with regimens based on strictly
reduce ion strength and individualized analysis of risk factors and risk periods.
increase urinary citrate
The information obtained from 24‑hour urine collec­
CaOx growth Maintain low APCaOx and tions and other long-term urine collections gives only
high pH during periods rough average information on specific risk variables
of particularly high risk of
CaOx precipitation
­during the period. Indeed, imprecise data on urinary pH
Stone are the rule rather than the exception. Better analyti­cal
Nature Reviews | Disease Primers focus on periods assumed to represent particularly high
Figure 8 | Potential methods to interfere with abnormal crystallization and stone
risk is necessary. Such information can be obtained by
formation. Oxidative stress in the proximal tubules might induce lipid peroxidation and
damage to the brush border membrane at this level of the nephron. Released membrane combining a careful medical history with analysis of
fragments and vesicles containing phospholipids are considered important as promoters risk factors during periods when supersaturation with
for both calcium phosphate (CaP) and calcium oxalate (CaOx) precipitation at CaOx occurs simultaneously with low urinary pH. Such
supersaturation levels lower than those otherwise required for crystallization. an approach might enable the design of individualized
Precipitation of CaOx higher in the nephron is only expected when very high recurrence prevention279.
concentrations of oxalate are available. Dilution of the urine at all levels of the Today, it is very attractive to remove kidney stones
nephron is most certainly associated with a reduced risk of intratubular, as well as with flexible ureteroscopy 265 and, if that is not optimal,
interstitial, precipitation of CaP and CaOx. *Indicates that precipitation of CaOx at high to use smaller and smaller instruments for percutane­
nephron levels only occurs when very high concentrations of oxalate are available. ous removal of smaller and smaller stones. However,
AP, ion-activity product.
it seems logical that for each treatment decision a bal­
ance is reached between the intention of the treatment
stone at the same point in time. The average recurrence and the efforts required to reach that goal. Active or
risk for patients with calcium stones was estimated to be pharmaco­logical removal of residual fragments should
approximately 50% during a 10‑year period after their be considered in view of the risk of the procedures and
first stone episode273. of the recurrence rate. There is no doubt that recurrence
Recent research has revived the role of Randall’s preventions need to be further developed, fine-tuned
plaques and plugs 88. In the final process of CaOx and used in an individually designed manner. With
stone formation, high supersaturation with CaOx and further increased understanding of the mechanisms
low pH seem to be important factors that need to be behind stone formation, it is possible that some gen­
­corrected121. The goal, therefore, should be to reduce eral treatment can be prescribed for many patients with
the risk of precipi­tation or growth of CaOx on the sur­ ­idiopathic calcium stone disease.
face of CaP3,274–276 (FIG. 8). The formation of CaOx stones It stands to reason, however, that further progress in
requires a complex interaction of nucleation of both CaP the management of patients who form stones cannot be
and CaOx, with subsequent growth and aggregation. made without a genuine interest and responsibility by
Interference with the precipitation of these two essential urologists who regularly see these patients. The need
crystal phases at different levels of the nephron seems for close interaction between stone removing and recur­
necessary to arrest or counteract the formation of the rence preventive procedures are absolute prerequisites
most commonly encountered stones in patients. for success. Basic as well as clinical research must have
Numerous recurrence preventive efforts for cal­ its focus on both aspects. In this regard, the urologist,
cium stone formation have been described and tested, during various endoscopic procedures, has a unique
but none have shown the capacity to definitely arrest possibility to visually observe and record the clinical
stone formation. One contributing factor to insufficient features of the pathology involved in stone formation. It
success is the lack of long-term patient compliance264. cannot be too strongly emphasized that the optimal care
Another important explanation is the shortage of means of patients with stone disease is not only surgical, in its
to counteract the initial precipitation of CaP54. With widest sense, but also medical.

18 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

1. Khan, S. R. Nephrocalcinosis in animal models with 28. Stamatelou, K. K., Francis, M. E., Jones, C. A., 54. Khan, S. R., Glenton, P. A., Backov, R. & Talham, D. R.
and without stones. Urol. Res. 38, 429–438 (2010). Nyberg, L. M. & Curhan, G. C. Time trends in Presence of lipids in urine, crystals and stones:
2. Finlayson, B. Physicochemical aspects of urolithiasis. reported prevalence of kidney stones in the United implications for the formation of kidney stones.
Kidney Int. 13, 344–360 (1978). States: 1976–1994. Kidney Int. 63, 1817–1823 Kidney Int. 62, 2062–2072 (2002).
3. Evan, A. P. Physiopathology and etiology of stone (2003). 55. Khan, S. R., Shevock, P. N. & Hackett, R. L. In vitro
formation in the kidney and the urinary tract. 29. Turney, B. W., Reynard, J. M., Noble, J. G. & precipitation of calcium oxalate in the presence of
Pediatr. Nephrol. 25, 831–841 (2010). Keoghane, S. R. Trends in urological stone disease. whole matrix or lipid components of the urinary
4. Tattevin, P. et al. Increased risk of renal stones in BJU Int. 109, 1082–1087 (2012). stones. J. Urol. 139, 418–422 (1988).
patients treated with atazanavir. Clin. Infect. Dis. 56, 30. Scales, C. D. et al. Changing gender prevalence of 56. Khan, S. R., Shevock, P. N. & Hackett, R. L.
1186 (2013). stone disease. J. Urol. 177, 979–982 (2007). Membrane-associated crystallization of calcium
5. Izzedine, H., Lescure, F. X. & Bonnet, F. HIV 31. Lieske, J. C. et al. Renal stone epidemiology in oxalate in vitro. Calcif. Tissue Int. 46, 116–120
medication-based urolithiasis. Clin. Kidney J. 7, Rochester, Minnesota: an update. Kidney Int. 69, (1990).
121–126 (2014). 760–764 (2006). 57. Hunter, G. K. Role of osteopontin in modulation
6. Raheem, O. A. et al. Prevalence of nephrolithiasis in 32. Strope, S. A., Wolf, J. S. & Hollenbeck, B. K. Changes of hydroxyapatite formation. Calcif. Tissue Int. 93,
human immunodeficiency virus infected patients on in gender distribution of urinary stone disease. 348–354 (2013).
the highly active antiretroviral therapy. J. Endourol. Urology 75, 543–546.e1 (2010). 58. Khan, S. R., Johnson, J. M., Peck, A. B.,
26, 1095–1098 (2012). 33. Ordon, M. et al. A population based study of the Cornelius, J. G. & Glenton, P. A. Expression of
7. Bischoff, K. & Rumbeiha, W. K. Pet food recalls and changing demographics of patients undergoing osteopontin in rat kidneys: induction during ethylene
pet food contaminants in small animals. Vet. Clin. definitive treatment for kidney stone disease. J. Urol. glycol induced calcium oxalate nephrolithiasis. J. Urol.
North Am. Small Anim. Pract. 42, 237–250 (2012). 193, 869–874 (2015). 168, 1173–1181 (2002).
8. Cianciolo, R. E. et al. Clinicopathologic, histologic, and 34. Curhan, G. C., Rimm, E. B., Willett, W. C. & 59. Khan, S. R., Joshi, S., Wang, W. & Peck, A. B.
toxicologic findings in 70 cats inadvertently exposed Stampfer, M. J. Regional variation in nephrolithiasis Regulation of macromolecular modulators of urinary
to pet food contaminated with melamine and cyanuric incidence and prevalence among United States men. stone formation by reactive oxygen species:
acid. J. Am. Vet. Med. Assoc. 233, 729–737 (2008). J. Urol. 151, 838–841 (1994). transcriptional study in an animal model of
9. Gabriels, G., Lambert, M., Smith, P., Wiesner, L. 35. Soucie, J. M., Thun, M. J., Coates, R. J., McClellan, W. hyperoxaluria. Am. J. Physiol. Renal Physiol. 306,
& Hiss, D. Melamine contamination in nutritional & Austin, H. Demographic and geographic variability F1285–F1295 (2014).
supplements — is it an alarm bell for the general of kidney stones in the United States. Kidney Int. 46, This is the first study to demonstrate the
consumer, athletes, and ‘Weekend Warriors’? Nutr. J. 893–899 (1994). involvement of reactive oxygen species in the
14, 69 (2015). 36. Mandel, N. S. & Mandel, G. S. Urinary tract stone regulation of macromolecular production.
10. Ding, J. Childhood urinary stones induced by disease in the United States veteran population. II. 60. Aihara, K., Byer, K. J. & Khan, S. R. Calcium
melamine-tainted formula: how much we know, Geographical analysis of variations in composition. phosphate-induced renal epithelial injury and stone
how much we don’t know. Kidney Int. 75, 780–782 J. Urol. 142, 1516–1521 (1989). formation: involvement of reactive oxygen species.
(2009). 37. Mandel, N. S. & Mandel, G. S. Urinary tract stone Kidney Int. 64, 1283–1291 (2003).
11. Fink, H. A. et al. Medical management to prevent disease in the United States veteran population. I. 61. Daudon, M., Doré, J.‑C., Jungers, P. & Lacour, B.
recurrent nephrolithiasis in adults: a systematic review Geographical frequency of occurrence. J. Urol. 142, Changes in stone composition according to age and
for an American College of Physicians Clinical 1513–1515 (1989). gender of patients: a multivariate epidemiological
Guideline. Ann. Intern. Med. 158, 535–543 (2013). 38. Curhan, G. C., Willett, W. C., Rimm, E. B., Speizer, F. E. approach. Urol. Res. 32, 241–247 (2004).
12. Scales, C. D., Smith, A. C., Hanley, J. M. & Saigal, C. S. & Stampfer, M. J. Body size and risk of kidney stones. 62. Khan, S. R. & Hackett, R. L. Identification of urinary
Prevalence of kidney stones in the United States. J. Am. Soc. Nephrol. 9, 1645–1652 (1998). stone and sediment crystals by scanning electron
Eur. Urol. 62, 160–165 (2012). 39. Sorensen, M. D. et al. Activity, energy intake, obesity, microscopy and X‑ray microanalysis. J. Urol. 135,
13. Obligado, S. H. & Goldfarb, D. S. The association and the risk of incident kidney stones in 818–825 (1986).
of nephrolithiasis with hypertension and obesity: postmenopausal women: a report from the Women’s 63. Siener, R., Netzer, L. & Hesse, A. Determinants of
a review. Am. J. Hypertens. 21, 257–264 (2008). Health Initiative. J. Am. Soc. Nephrol. 25, 362–369 brushite stone formation: a case–control study.
14. Brikowski, T. H., Lotan, Y. & Pearle, M. S. Climate- (2014). PLoS ONE 8, e78996 (2013).
related increase in the prevalence of urolithiasis in 40. Chung, S.‑D., Chen, Y.‑K. & Lin, H.‑C. Increased risk 64. Sakhaee, K., Adams-Huet, B., Moe, O. W.
the United States. Proc. Natl Acad. Sci. USA 105, of diabetes in patients with urinary calculi: a 5‑year & Pak, C. Y. C. Pathophysiologic basis for
9841–9846 (2008). followup study. J. Urol. 186, 1888–1893 (2011). normouricosuric uric acid nephrolithiasis. Kidney Int.
15. Taylor, E. N., Stampfer, M. J. & Curhan, G. C. Obesity, 41. West, B. et al. Metabolic syndrome and self-reported 62, 971–979 (2002).
weight gain, and the risk of kidney stones. JAMA 293, history of kidney stones: the National Health and 65. Fellström, B. et al. The influence of a high dietary
455–462 (2005). Nutrition Examination Survey (NHANES III) intake of purine-rich animal protein on urinary urate
16. Daudon, M. & Jungers, P. Diabetes and 1988–1994. Am. J. Kidney Dis. 51, 741–747 excretion and supersaturation in renal stone disease.
nephrolithiasis. Curr. Diab. Rep. 7, 443–448 (2007). (2008). Clin. Sci. (Lond.) 64, 399–405 (1983).
17. Lieske, J. C. et al. Diabetes mellitus and the risk of 42. Jeong, I. G. et al. Association between metabolic 66. Grases, F., Villacampa, A. I., Costa-Bauzá, A.
urinary tract stones: a population-based case–control syndrome and the presence of kidney stones in a & Söhnel, O. Uric acid calculi: types, etiology and
study. Am. J. Kidney Dis. 48, 897–904 (2006). screened population. Am. J. Kidney Dis. 58, 383–388 mechanisms of formation. Clin. Chim. Acta 302,
18. Taylor, E. N., Stampfer, M. J. & Curhan, G. C. Diabetes (2011). 89–104 (2000).
mellitus and the risk of nephrolithiasis. Kidney Int. 68, 43. Ferraro, P. M. et al. History of kidney stones and the 67. Khan, S. R., Hackett, R. L. & Finlayson, B. Morphology
1230–1235 (2005). risk of coronary heart disease. JAMA 310, 408–415 of urinary stone particles resulting from ESWL
19. Strazzullo, P. et al. Past history of nephrolithiasis and (2013). treatment. J. Urol. 136, 1367–1372 (1986).
incidence of hypertension in men: a reappraisal based 44. Alexander, R. T. et al. Kidney stones and 68. Griffith, D. P. & Osborne, C. A. Infection (urease)
on the results of the Olivetti Prospective Heart study. cardiovascular events: a cohort study. Clin. J. Am. Soc. stones. Miner. Electrolyte Metab. 13, 278–285
Nephrol. Dial. Transplant. 16, 2232–2235 (2001). Nephrol. 9, 506–512 (2014). (1987).
20. Johri, N. et al. An update and practical guide to renal 45. Rule, A. D. et al. Kidney stones associate with 69. Biyani, C. S. & Cartledge, J. J. Cystinuria — diagnosis
stone management. Nephron Clin. Pract. 116, increased risk for myocardial infarction. J. Am. Soc. and management. EAU–EBU Updat. Ser. 4, 175–183
c159–c171 (2010). Nephrol. 21, 1641–1644 (2010). (2006).
21. Cappuccio, F. P., Strazzullo, P. & Mancini, M. Kidney 46. Khan, S. R. & Hackett, R. L. Role of organic matrix 70. Robertson, W. G., Peacock, M. & Nordin, B. E.
stones and hypertension: population based study of in urinary stone formation: an ultrastructural study Calcium oxalate crystalluria and urine saturation in
an independent clinical association. BMJ 300, of crystal matrix interface of calcium oxalate recurrent renal stone-formers. Clin. Sci. 40, 365–374
1234–1236 (1990). monohydrate stones. J. Urol. 150, 239–245 (1993). (1971).
22. Rule, A. D., Krambeck, A. E. & Lieske, J. C. Chronic 47. Ryall, R. L., Chauvet, M. C. & Grover, P. K. 71. Finlayson, B. & Reid, F. The expectation of free and
kidney disease in kidney stone formers. Clin. J. Am. Intracrystalline proteins and urolithiasis: a comparison fixed particles in urinary stone disease. Invest. Urol.
Soc. Nephrol. 6, 2069–2075 (2011). of the protein content and ultrastructure of urinary 15, 442–448 (1978).
23. El‑Zoghby, Z. M. et al. Urolithiasis and the risk of calcium oxalate monohydrate and dihydrate crystals. 72. Robertson, W. G. Measurement of ionized calcium
ESRD. Clin. J. Am. Soc. Nephrol. 7, 1409–1415 BJU Int. 96, 654–663 (2005). in biological fluids. Clin. Chim. Acta 24, 149–157
(2012). 48. McKee, M. D., Nanci, A. & Khan, S. R. Ultrastructural (1969).
24. Shoag, J., Halpern, J., Goldfarb, D. S. & Eisner, B. H. immunodetection of osteopontin and osteocalcin as 73. Werness, P. G., Brown, C. M., Smith, L. H. &
Risk of chronic and end stage kidney disease major matrix components of renal calculi. J. Bone Finlayson, B. EQUIL2: a BASIC computer program
in patients with nephrolithiasis. J. Urol. 192, Miner. Res. 10, 1913–1929 (1995). for the calculation of urinary saturation. J. Urol. 134,
1440–1445 (2014). 49. Khan, S. R. & Kok, D. J. Modulators of urinary stone 1242–1244 (1985).
25. Keddis, M. T. & Rule, A. D. Nephrolithiasis and loss of formation. Front. Biosci. 9, 1450–1482 (2004). 74. May, P. M. & Muray, K. JESS, a joint expert speciation
kidney function. Curr. Opin. Nephrol. Hypertens. 22, 50. Atmani, F. & Khan, S. R. Role of urinary bikunin in the system‑II. The thermodynamic database. Talanta 38,
390–396 (2013). inhibition of calcium oxalate crystallization. J. Am. Soc. 1419–1426 (1991).
26. Department of Health and Human Services USA, Nephrol. 10, S385–S388 (1999). 75. Brown, C. M., Ackermann, D. K. & Purich, D. L.
National Institutes of Health & National Institute of 51. Ryall, R. L. Macromolecules and urolithiasis: parallels EQUIL93: a tool for experimental and clinical
Diabetes and Digestive and Kidney Diseases. Urologic and paradoxes. Nephron Physiol. 98, 37–42 (2004). urolithiasis. Urol. Res. 22, 119–126 (1994).
Diseases in America (US Government Printing Office, 52. Khan, S. R. et al. Lipids and membranes in the organic 76. Robertson, W. G. Factors affecting the precipitation
2012). matrix of urinary calcific crystals and stones. of calcium phosphate in vitro. Calcif. Tissue Res. 11,
27. Romero, V., Akpinar, H. & Assimos, D. G. Kidney Calcif. Tissue Int. 59, 357–365 (1996). 311–322 (1973).
stones: a global picture of prevalence, incidence, 53. Khan, S. R. & Glenton, P. A. Increased urinary 77. Kok, D. J. & Khan, S. R. Calcium oxalate
and associated risk factors. Rev. Urol. 12, e86–e96 excretion of lipids by patients with kidney stones. nephrolithiasis, a free or fixed particle disease.
(2010). Br. J. Urol. 77, 506–511 (1996). Kidney Int. 46, 847–854 (1994).

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 19


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

78. Robertson, W. G. Potential role of fluctuations in the 103. Cooke, S. A. The site of calcification in the human renal 128. Robertson, W. G., Scurr, D. S. & Bridge, C. M. Factors
composition of renal tubular fluid through the nephron papilla. Br. J. Surg. 57, 890–896 (1970). influencing the crystallisation of calcium oxalate in
in the initiation of Randall’s plugs and calcium oxalate 104. Evan, A. P. et al. Randall’s plaque of patients with urine — critique. J. Cryst. Growth 53, 182–194
crystalluria in a computer model of renal function. nephrolithiasis begins in basement membranes of thin (1981).
Urolithiasis 43 (Suppl. 1), 93–107 (2015). loops of Henle. J. Clin. Invest. 111, 607–616 (2003). 129. Worcester, E. M., Nakagawa, Y. & Coe, F. L.
79. Fleisch, H. & Bisaz, S. The inhibitory effect of 105. Stoller, M. L., Low, R. K., Shami, G. S., McCormick, V. D. Glycoprotein calcium oxalate crystal growth inhibitor in
pyrophosphate on calcium oxalate precipitation and & Kerschmann, R. L. High resolution radiography of urine. Miner. Electrolyte Metab. 13, 267–272 (1987).
its relation to urolithiasis. Experientia 20, 276–277 cadaveric kidneys: unraveling the mystery of Randall’s 130. Nakagawa, Y., Ahmed, M., Hall, S. L., Deganello, S.
(1964). plaque formation. J. Urol. 156, 1263–1266 (1996). & Coe, F. L. Isolation from human calcium oxalate
80. Fleisch, H. & Bisaz, S. Isolation from urine of This paper provided the first suggestion that renal stones of nephrocalcin, a glycoprotein inhibitor
pyrophosphate, a calcification inhibitor. Am. J. Physiol. plaques might start in the vasa recta. of calcium oxalate crystal growth. Evidence that
203, 671–675 (1962). 106. Stoller, M. L., Meng, M. V., Abrahams, H. M. nephrocalcin from patients with calcium oxalate
81. Asplin, J. R., Mandel, N. S. & Coe, F. L. Evidence of & Kane, J. P. The primary stone event: a new nephrolithiasis is deficient in gamma-carboxyglutamic
calcium phosphate supersaturation in the loop of hypothesis involving a vascular etiology. J. Urol. 171, acid. J. Clin. Invest. 79, 1782–1787 (1987).
Henle. Am. J. Physiol. 270, F604–F613 (1996). 1920–1924 (2004). 131. Hess, B., Nakagawa, Y. & Coe, F. L. Inhibition of
82. Khan, S. R. & Hackett, R. L. Developmental 107. Haggitt, R. C. & Pitcock, J. A. Renal medullary calcium oxalate monohydrate crystal aggregation
morphology of calcium oxalate foreign body stones calcifications: a light and electron microscopic study. by urine proteins. Am. J. Physiol. 257, F99–F106
in rats. Calcif. Tissue Int. 37, 165–173 (1985). J. Urol. 106, 342–347 (1971). (1989).
83. Khan, S. R. & Hackett, R. L. Urolithogenesis of mixed 108. Weller, R. O., Nester, B. & Cooke, S. A. Calcification 132. Shiraga, H. et al. Inhibition of calcium oxalate crystal
foreign body stones. J. Urol. 138, 1321–1328 in the human renal papilla: an electron-microscope growth in vitro by uropontin: another member of the
(1987). study. J. Pathol. 107, 211–216 (1972). aspartic acid-rich protein superfamily. Proc. Natl Acad.
84. Linnes, M. P. et al. Phenotypic characterization of 109. Miller, N. L. et al. A formal test of the hypothesis that Sci. USA 89, 426–430 (1992).
kidney stone formers by endoscopic and histological idiopathic calcium oxalate stones grow on Randall’s 133. Tsuji, H. et al. Urinary concentration of osteopontin
quantification of intrarenal calcification. Kidney Int. plaque. BJU Int. 103, 966–971 (2009). and association with urinary supersaturation and
84, 818–825 (2013). 110. Evan, A. P. et al. Apatite plaque particles in inner crystal formation. Int. J. Urol. 14, 630–634 (2007).
85. Wang, X. et al. Distinguishing characteristics of medulla of kidneys of calcium oxalate stone formers: 134. Pillay, S. N., Asplin, J. R. & Coe, F. L. Evidence
idiopathic calcium oxalate kidney stone formers osteopontin localization. Kidney Int. 68, 145–154 that calgranulin is produced by kidney cells and is
with low amounts of Randall’s plaque. Clin. J. Am. (2005). an inhibitor of calcium oxalate crystallization.
Soc. Nephrol. 9, 1757–1763 (2014). 111. Evan, A. P. et al. Renal inter-α-trypsin inhibitor heavy Am. J. Physiol. 275, F255–F261 (1998).
86. Khan, S. R. Experimental calcium oxalate chain 3 increases in calcium oxalate stone-forming 135. Morse, R. M. & Resnick, M. I. A new approach to the
nephrolithiasis and the formation of human urinary patients. Kidney Int. 72, 1503–1511 (2007). study of urinary macromolecules as a participant in
stones. Scanning Microsc. 9, 89–100; discussion 112. Evan, A., Lingeman, J., Coe, F. L. & Worcester, E. calcium oxalate crystallization. J. Urol. 139, 869–873
100–101 (1995). Randall’s plaque: pathogenesis and role in calcium (1988).
87. Randall, A. The etiology of primary renal calculus. oxalate nephrolithiasis. Kidney Int. 69, 1313–1318 136. Dussol, B. et al. Analysis of the soluble organic matrix
Int. Abstr. Surg. 71, 209–240 (1940). (2006). of five morphologically different kidney stones.
88. Khan, S. R. & Canales, B. K. Unified theory on the 113. Carpentier, X. et al. High Zn content of Randall’s Evidence for a specific role of albumin in the
pathogenesis of Randall’s plaques and plugs. plaque: a μ‑X‑ray fluorescence investigation. J. Trace constitution of the stone protein matrix. Urol. Res. 23,
Urolithiasis 43 (Suppl. 1), 109–123 (2015). Elem. Med. Biol. 25, 160–165 (2011). 45–51 (1995).
89. Bushinsky, D. A., Frick, K. K. & Nehrke, K. Genetic 114. Evan, A. P., Lingeman, J. E., Coe, F. L. & 137. Stapleton, A. M. et al. Further evidence linking
hypercalciuric stone-forming rats. Curr. Opin. Nephrol. Worcester, E. M. Role of interstitial apatite plaque urolithiasis and blood coagulation: urinary
Hypertens. 15, 403–418 (2006). in the pathogenesis of the common calcium oxalate prothrombin fragment 1 is present in stone matrix.
90. Khan, S. R. & Canales, B. K. Ultrastructural stone. Semin. Nephrol. 28, 111–119 (2008). Kidney Int. 49, 880–888 (1996).
investigation of crystal deposits in Npt2a knockout 115. Khan, S. R. & Gambaro, G. Role of osteogenesis in the 138. Grover, P. K. & Ryall, R. L. Inhibition of calcium oxalate
mice: are they similar to human Randall’s plaques? formation of Randall’s plaques. Anat. Rec. (Hoboken) crystal growth and aggregation by prothrombin and its
J. Urol. 186, 1107–1113 (2011). 299, 5–7 (2015). fragments in vitro: relationship between protein
91. Khan, S. R. & Hackett, R. L. Retention of calcium 116. Mezzabotta, F. et al. Spontaneous calcification process structure and inhibitory activity. Eur. J. Biochem. 263,
oxalate crystals in renal tubules. Scanning Microsc. 5, in primary renal cells from a medullary sponge kidney 50–56 (1999).
707–711; discussion 711–712 (1991). patient harbouring a GDNF mutation. J. Cell. Mol. 139. Dawson, C. J., Grover, P. K. & Ryall, R. L. Inter-alpha-
92. Khan, S. R., Finlayson, B. & Hackett, R. L. Med. 19, 889–902 (2015). inhibitor in urine and calcium oxalate urinary crystals.
Experimental calcium oxalate nephrolithiasis in 117. Khan, S. R., Glenton, P. A. & Byer, K. J. Modeling Br. J. Urol. 81, 20–26 (1998).
the rat. Role of the renal papilla. Am. J. Pathol. 107, of hyperoxaluric calcium oxalate nephrolithiasis: 140. Robertson, W. G. A risk factor model of stone-
59–69 (1982). experimental induction of hyperoxaluria by formation. Front. Biosci. 8, s1330–s1338 (2003).
93. Evan, A. P. et al. Crystal-associated nephropathy in hydroxy‑l‑proline. Kidney Int. 70, 914–923 (2006). 141. Spector, A. R., Gray, A. & Prien, E. L. Kidney stone
patients with brushite nephrolithiasis. Kidney Int. 67, 118. Meyer, J. L., Bergert, J. H. & Smith, L. H. Epitaxial matrix. Differences in acidic protein composition.
576–591 (2005). relationships in urolithiasis: the calcium oxalate Invest. Urol. 13, 387–389 (1976).
94. Evan, A. P. et al. Renal crystal deposits and monohydrate–hydroxyapatite system. Clin. Sci. Mol. 142. Lian, J. B., Prien, E. L., Glimcher, M. J. & Gallop, P. M.
histopathology in patients with cystine stones. Med. 49, 369–374 (1975). The presence of protein-bound gamma-
Kidney Int. 69, 2227–2235 (2006). 119. Sethman, I., Grohe, B. & Kleebe, H.‑J. Replacement of carboxyglutamic acid in calcium-containing renal
95. Evan, A. P. et al. Renal intratubular crystals and hydroxyapatite by whewellite: implications for kidney calculi. J. Clin. Invest. 59, 1151–1157 (1977).
hyaluronan staining occur in stone formers with stone formation. Miner. Mag. 78, 91–100 (2014). 143. Jones, W. T. & Resnick, M. I. The characterization of
bypass surgery but not with idiopathic calcium 120. Højgaard, I., Fornander, A. M., Nilsson, M. A. soluble matrix proteins in selected human renal calculi
oxalate stones. Anat. Rec. (Hoboken) 291, 325–334 & Tiselius, H. G. The effect of pH changes on the using two-dimensional polyacrylamide gel
(2008). crystallization of calcium salts in solutions with an ion electrophoresis. J. Urol. 144, 1010–1014 (1990).
96. Coe, F. L., Evan, A. P., Lingeman, J. E. & composition corresponding to that in the distal tubule. 144. Rose, G. A. & Sulaiman, S. Tamm–Horsfall
Worcester, E. M. Plaque and deposits in nine human Urol. Res. 27, 409–416 (1999). mucoproteins promote calcium oxalate crystal
stone diseases. Urol. Res. 38, 239–247 (2010). 121. Tiselius, H.‑G. A hypothesis of calcium stone formation in urine: quantitative studies. J. Urol. 127,
97. Evan, A. P., Worcester, E. M., Coe, F. L., Williams, J. formation: an interpretation of stone research during 177–179 (1982).
& Lingeman, J. E. Mechanisms of human kidney stone the past decades. Urol. Res. 39, 231–243 (2011). 145. Robertson, W. G. & Scurr, D. S. Modifiers of calcium
formation. Urolithiasis 43 (Suppl. 1), 19–32 (2015). 122. Borden, T. A. & Lyon, E. S. The effects of magnesium oxalate crystallization found in urine. I. Studies with a
98. Evan, A. E. et al. Histopathology and surgical anatomy and pH on experimental calcium oxalate stone continuous crystallizer using an artificial urine. J. Urol.
of patients with primary hyperparathyroidism and disease. Invest. Urol. 6, 412–422 (1969). 135, 1322–1326 (1986).
calcium phosphate stones. Kidney Int. 74, 223–229 123. Meyer, J. L. & Smith, L. H. Growth of calcium oxalate 146. Grover, P. K., Ryall, R. L. & Marshall, V. R. Does
(2008). crystals. II. Inhibition by natural urinary crystal growth Tamm–Horsfall mucoprotein inhibit or promote
99. Khan, S. R., Finlayson, B. & Hackett, R. Renal papillary inhibitors. Invest. Urol. 13, 36–39 (1975). calcium oxalate crystallization in human urine?
changes in patient with calcium oxalate lithiasis. 124. Meyer, J. L., McCall, J. T. & Smith, L. H. Inhibition Clin. Chim. Acta 190, 223–238 (1990).
Urology 23, 194–199 (1984). of calcium phosphate crystallization by nucleoside 147. Bagga, H. S., Chi, T., Miller, J. & Stoller, M. L.
100. Khan, S. R., Rodriguez, D. E., Gower, L. B. & phosphates. Calcif. Tissue Res. 15, 287–293 (1974). New insights into the pathogenesis of renal calculi.
Monga, M. Association of Randall plaque with 125. Howard, J. E., Thomas, W. C., Barker, L. M., Urol. Clin. North Am. 40, 1–12 (2013).
collagen fibers and membrane vesicles. J. Urol. 187, Smith, L. H. & Wadkins, C. L. The recognition and 148. Fabris, A. et al. The relationship between calcium
1094–1100 (2012). isolation from urine and serum of a peptide inhibitor kidney stones, arterial stiffness and bone density:
This is the first study to discuss the initiation of to calcification. Johns Hopkins Med. J. 120, 119–136 unraveling the stone–bone–vessel liaison. J. Nephrol.
Randall’s plaque through the deposition of CaP (1967). 28, 549–555 (2015).
in membrane-bound vesicles and plaque growth 126. Robertson, W. G., Peacock, M. & Nordin, B. E. 149. Gambaro, G. et al. Crystals, Randall’s plaques and
via the renal interstitium by mineralization of Inhibitors of the growth and aggregation of calcium renal stones: do bone and atherosclerosis teach us
collagen fibres. oxalate crystals in vitro. Clin. Chim. Acta 43, 31–37 something? J. Nephrol. 17, 774–777 (2004).
101. Coe, F. L., Evan, A. P., Worcester, E. M. (1973). 150. Reiner, A. P. et al. Kidney stones and subclinical
& Lingeman, J. E. Three pathways for human kidney 127. Ryall, R. L., Harnett, R. M. & Marshall, V. R. The effect atherosclerosis in young adults: the CARDIA study.
stone formation. Urol. Res. 38, 147–160 (2010). of urine, pyrophosphate, citrate, magnesium and J. Urol. 185, 920–925 (2011).
102. Randall, A. Recent advances in knowledge relating glycosaminoglycans on the growth and aggregation of 151. Taylor, E. R. & Stoller, M. L. Vascular theory of
to the formation, recognition and treatment of kidney calcium oxalate crystals in vitro. Clin. Chim. Acta 112, the formation of Randall plaques. Urolithiasis
calculi. Bull. N. Y. Acad. Med. 20, 473–484 (1944). 349–356 (1981). 43 (Suppl. 1), 41–45 (2015).

20 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

152. Moe, S. M. & Chen, N. X. Mechanisms of vascular 175. Primiano, A. et al. FT‑IR analysis of urinary stones: 195. Pearle, M. S. et al. Medical management of kidney
calcification in chronic kidney disease. J. Am. Soc. a helpful tool for clinician comparison with the stones: AUA guideline. J. Urol. 192, 316–324 (2014).
Nephrol. 19, 213–216 (2008). chemical spot test. Dis. Markers 2014, 176165 196. Yendt, E. R. & Cohanim, M. Prevention of calcium
153. Shanahan, C. M. Mechanisms of vascular calcification (2014). stones with thiazides. Kidney Int. 13, 397–409
in renal disease. Clin. Nephrol. 63, 146–157 (2005). 176. Gambaro, G., Reis-Santos, J. M. & Rao, N. (1978).
154. Shanahan, C. M., Crouthamel, M. H., Kapustin, A. Nephrolithiasis: why doesn’t our ‘learning’ progress? 197. Ettinger, B., Tang, A., Citron, J. T., Livermore, B.
& Giachelli, C. M. Arterial calcification in chronic Eur. Urol. 45, 547–556; discussion 556 (2004). & Williams, T. Randomized trial of allopurinol in the
kidney disease: key roles for calcium and phosphate. 177. Eisner, B. H., Sheth, S., Dretler, S. P., Herrick, B. prevention of calcium oxalate calculi. N. Engl. J. Med.
Circ. Res. 109, 697–711 (2011). & Pais, V. M. Abnormalities of 24‑hour urine 315, 1386–1389 (1986).
155. Kapustin, A. N. et al. Calcium regulates key composition in first-time and recurrent stone-formers. 198. Arowojolu, O. & Goldfarb, D. S. Treatment of calcium
components of vascular smooth muscle cell-derived Urology 80, 776–779 (2012). nephrolithiasis in the patient with hyperuricosuria.
matrix vesicles to enhance mineralization. Circ. Res. 178. Rodgers, A. L., Allie-Hamdulay, S., Jackson, G. J. Nephrol. 27, 601–605 (2014).
109, e1–e12 (2011). & Tiselius, H.‑G. Simulating calcium salt precipitation 199. Pak, C. Y., Sakhaee, K. & Fuller, C. J. Physiological and
156. Shroff, R. C. & Shanahan, C. M. The vascular biology in the nephron using chemical speciation. Urol. Res. physiochemical correction and prevention of calcium
of calcification. Semin. Dial. 20, 103–109 (2007). 39, 245–251 (2011). stone formation by potassium citrate therapy.
157. Tada, Y. et al. Advanced glycation end products- 179. Sakhaee, K., Maalouf, N. M., Kumar, R., Pasch, A. Trans. Assoc. Am. Physicians 96, 294–305 (1983).
induced vascular calcification is mediated by oxidative & Moe, O. W. Nephrolithiasis-associated bone disease: 200. Barcelo, P., Wuhl, O., Servitge, E., Rousaud, A.
stress: functional roles of NAD(P)H‑oxidase. pathogenesis and treatment options. Kidney Int. 79, & Pak, C. Y. Randomized double-blind study of
Horm. Metab. Res. 45, 267–272 (2013). 393–403 (2011). potassium citrate in idiopathic hypocitraturic calcium
158. Byon, C. H. et al. Oxidative stress induces vascular This is an updated overview of the epidemiology nephrolithiasis. J. Urol. 150, 1761–1764 (1993).
calcification through modulation of the osteogenic and mechanisms of MBD in patients with 201. Ettinger, B. et al. Potassium-magnesium citrate is
transcription factor Runx2 by AKT signaling. J. Biol. nephrolithiasis. The effect of treatments for an effective prophylaxis against recurrent calcium
Chem. 283, 15319–15327 (2008). renal stone prevention on the associated MBD oxalate nephrolithiasis. J. Urol. 158, 2069–2073
159. Murshed, M. & McKee, M. D. Molecular determinants and the effect of treatments addressing the (1997).
of extracellular matrix mineralization in bone and bone-on-the-stone disease are 202. Sakhaee, K., Nicar, M., Hill, K. & Pak, C. Y.
blood vessels. Curr. Opin. Nephrol. Hypertens. 19, thoroughly discussed. Contrasting effects of potassium citrate and sodium
359–365 (2010). 180. Ferraro, P. M., D’Addessi, A. & Gambaro, G. When citrate therapies on urinary chemistries and
160. Jia, Z. et al. Does crystal deposition in genetic to suspect a genetic disorder in a patient with renal crystallization of stone-forming salts. Kidney Int. 24,
hypercalciuric rat kidney tissue share similarities stones, and why. Nephrol. Dial. Transplant. 28, 348–352 (1983).
with bone formation? Urology 83, 509.e7–509.14 811–820 (2013). 203. Preminger, G. M., Sakhaee, K., Skurla, C. & Pak, C. Y.
(2014). 181. Kang, H. W. et al. Effect of renal insufficiency on stone Prevention of recurrent calcium stone formation with
161. Naito, Y. et al. Morphological analysis of renal cell recurrence in patients with urolithiasis. J. Korean Med. potassium citrate therapy in patients with distal renal
culture models of calcium phosphate stone formation. Sci. 29, 1132–1137 (2014). tubular acidosis. J. Urol. 134, 20–23 (1985).
Urol. Res. 25, 59–65 (1997). 182. Kristensen, C., Parks, J. H., Lindheimer, M. & Coe, F. L. 204. Fabris, A. et al. Long-term treatment with potassium
162. Kageyama, S. et al. Microlith formation in vitro by Reduced glomerular filtration rate and hypercalciuria citrate and renal stones in medullary sponge kidney.
Madin Darby canine kidney (MDCK) cells. Int. J. Urol. in primary struvite nephrolithiasis. Kidney Int. 32, Clin. J. Am. Soc. Nephrol. 5, 1663–1668 (2010).
3, 23–26 (1996). 749–753 (1987). 205. Skolarikos, A. et al. Metabolic evaluation and
163. Thamilselvan, S., Byer, K. J., Hackett, R. L. 183. Evan, A. P. et al. Contrasting histopathology and recurrence prevention for urinary stone patients:
& Khan, S. R. Free radical scavengers, catalase crystal deposits in kidneys of idiopathic stone formers EAU guidelines. Eur. Urol. 67, 750–763 (2015).
and superoxide dismutase provide protection from who produce hydroxy apatite, brushite, or calcium 206. Daudon, M. et al. Cystine crystal volume
oxalate-associated injury to LLC‑PK1 and MDCK cells. oxalate stones. Anat. Rec. (Hoboken) 297, 731–748 determination: a useful tool in the management of
J. Urol. 164, 224–229 (2000). (2014). cystinuric patients. Urol. Res. 31, 207–211 (2003).
164. Thamilselvan, S., Hackett, R. L. & Khan, S. R. Lipid 184. Ascenti, G. et al. Stone-targeted dual-energy CT: 207. Goldfarb, D. S., Coe, F. L. & Asplin, J. R. Urinary
peroxidation in ethylene glycol induced hyperoxaluria a new diagnostic approach to urinary calculosis. cystine excretion and capacity in patients with
and calcium oxalate nephrolithiasis. J. Urol. 157, AJR Am. J. Roentgenol. 195, 953–958 (2010). cystinuria. Kidney Int. 69, 1041–1047 (2006).
1059–1063 (1997). 185. Halbritter, J. et al. Fourteen monogenic genes account 208. Dello Strologo, L., Laurenzi, C., Legato, A. &
165. Joshi, S., Saylor, B. T., Wang, W., Peck, A. B. for 15% of nephrolithiasis/nephrocalcinosis. J. Am. Pastore, A. Cystinuria in children and young adults:
& Khan, S. R. Apocynin-treatment reverses Soc. Nephrol. 26, 543–551 (2015). success of monitoring free-cystine urine levels.
hyperoxaluria induced changes in NADPH oxidase 186. Gambaro, G. et al. Genetics of hypercalciuria and Pediatr. Nephrol. 22, 1869–1873 (2007).
system expression in rat kidneys: a transcriptional calcium nephrolithiasis: from the rare monogenic to 209. Prot-Bertoye, C. et al. CKD and its risk factors among
study. PLoS ONE 7, e47738 (2012). the common polygenic forms. Am. J. Kidney Dis. 44, patients with cystinuria. Clin. J. Am. Soc. Nephrol. 10,
166. Zuo, J., Khan, A., Glenton, P. A. & Khan, S. R. Effect of 963–986 (2004). 842–851 (2015).
NADPH oxidase inhibition on the expression of kidney 187. Pak, C. Y. et al. Prevention of stone formation and 210. Ordon, M. et al. The surgical management of kidney
injury molecule and calcium oxalate crystal deposition bone loss in absorptive hypercalciuria by combined stone disease: a population based time series analysis.
in hydroxy-l‑proline-induced hyperoxaluria in the male dietary and pharmacological interventions. J. Urol. J. Urol. 192, 1450–1456 (2014).
Sprague-Dawley rats. Nephrol. Dial. Transplant. 26, 169, 465–469 (2003). 211. Scales, C. D. et al. Comparative effectiveness of shock
1785–1796 (2011). 188. Fabris, A. et al. Bone disease in medullary sponge wave lithotripsy and ureteroscopy for treating patients
167. Khan, S. R., Khan, A. & Byer, K. J. Temporal changes kidney and effect of potassium citrate treatment. with kidney stones. JAMA Surg. 149, 648–653
in the expression of mRNA of NADPH oxidase Clin. J. Am. Soc. Nephrol. 4, 1974–1979 (2009). (2014).
subunits in renal epithelial cells exposed to oxalate or Treatment with potassium citrate not only This is one of the few studies comparing large
calcium oxalate crystals. Nephrol. Dial. Transplant. 26, decreased stone recurrences but also improved head-to-head data in SWL versus ureteroscopy.
1778–1785 (2011). mineral bone density in a cohort of patients with 212. Lingeman, J. E. et al. Extracorporeal shock wave
168. Joshi, S., Clapp, W. L., Wang, W. & Khan, S. R. medullary sponge kidney. The effect was probably lithotripsy: the Methodist Hospital of Indiana
Osteogenic changes in kidneys of hyperoxaluric rats. due to the amelioration of the subtle metabolic experience. J. Urol. 135, 1134–1137 (1986).
Biochim. Biophys. Acta 1852, 2000–2012 (2015). acidosis in patients with medullary sponge kidney. 213. Wignall, G. R., Canales, B. K., Denstedt, J. D.
The results of this study show osteogenic changes 189. Hosking, D. H., Erickson, S. B., Van den Berg, C. J., & Monga, M. Minimally invasive approaches to upper
in the kidneys of hyperoxaluric rats. Wilson, D. M. & Smith, L. H. The stone clinic effect in urinary tract urolithiasis. Urol. Clin. North Am. 35,
169. Kanno, T. et al. The efficacy of ultrasonography for patients with idiopathic calcium urolithiasis. J. Urol. 441–454 (2008).
the detection of renal stone. Urology 84, 285–288 130, 1115–1118 (1983). This is a solid review of preoperative considerations
(2014). 190. Borghi, L. et al. Urinary volume, water and recurrences and surgical techniques for urologists who perform
170. Kanno, T. et al. Determining the efficacy of in idiopathic calcium nephrolithiasis: a 5‑year SWL, ureteroscopy and PCNL.
ultrasonography for the detection of ureteral stone. randomized prospective study. J. Urol. 155, 839–843 214. Albala, D. M. et al. Lower pole I: a prospective
Urology 84, 533–537 (2014). (1996). randomized trial of extracorporeal shock wave
171. Heidenreich, A., Desgrandschamps, F. & Terrier, F. 191. Borghi, L. et al. Comparison of two diets for the lithotripsy and percutaneous nephrostolithotomy for
Modern approach of diagnosis and management prevention of recurrent stones in idiopathic lower pole nephrolithiasis-initial results. J. Urol. 166,
of acute flank pain: review of all imaging modalities. hypercalciuria. N. Engl. J. Med. 346, 77–84 (2002). 2072–2080 (2001).
Eur. Urol. 41, 351–362 (2002). 192. Meschi, T. et al. The effect of fruits and vegetables on 215. Pearle, M. S. et al. Prospective, randomized trial
172. Johnston, R., Lin, A., Du, J. & Mark, S. Comparison urinary stone risk factors. Kidney Int. 66, 2402–2410 comparing shock wave lithotripsy and ureteroscopy
of kidney–ureter–bladder abdominal radiography (2004). for lower pole caliceal calculi 1 cm or less. J. Urol. 173,
and computed tomography scout films for identifying 193. Ferraro, P. M., Taylor, E. N., Gambaro, G. & 2005–2009 (2005).
renal calculi. BJU Int. 104, 670–673 (2009). Curhan, G. C. Soda and other beverages and the risk 216. Wiesenthal, J. D., Ghiculete, D., D’A Honey, R. J.
173. Worster, A., Preyra, I., Weaver, B. & Haines, T. The of kidney stones. Clin. J. Am. Soc. Nephrol. 8, & Pace, K. T. A comparison of treatment modalities
accuracy of noncontrast helical computed tomography 1389–1395 (2013). for renal calculi between 100 and 300 mm2:
versus intravenous pyelography in the diagnosis of This is a robust observational study in the general are shockwave lithotripsy, ureteroscopy, and
suspected acute urolithiasis: a meta-analysis. population (the three health professionals percutaneous nephrolithotomy equivalent?
Ann. Emerg. Med. 40, 280–286 (2002). Channing cohorts) that showed that soda J. Endourol. 25, 481–485 (2011).
174. Wiesenthal, J. D., Ghiculete, D., D’A Honey, R. J. beverages increase the risk of becoming a 217. Gupta, N. P., Ansari, M. S., Kesarvani, P., Kapoor, A.
& Pace, K. T. Evaluating the importance of mean stone stone former. & Mukhopadhyay, S. Role of computed tomography
density and skin‑to‑stone distance in predicting 194. Bushinsky, D. A. et al. Increased dietary oxalate does with no contrast medium enhancement in predicting
successful shock wave lithotripsy of renal and ureteric not increase urinary calcium oxalate saturation in the outcome of extracorporeal shock wave lithotripsy
calculi. Urol. Res. 38, 307–313 (2010). hypercalciuric rats. Kidney Int. 55, 602–612 (1999). for urinary calculi. BJU Int. 95, 1285–1288 (2005).

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 21


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

218. Wang, L.‑J. et al. Predictions of outcomes of renal 238. Moran, M. E., Abrahams, H. M., Burday, D. E. 262. Thalji, N. K., Richards, N. G., Peck, A. B. &
stones after extracorporeal shock wave lithotripsy & Greene, T. D. Utility of oral dissolution therapy in Canales, B. K. Enzymatic dissolution of calcium and
from stone characteristics determined by unenhanced the management of referred patients with secondarily struvite crystals: in vitro evaluation of biochemical
helical computed tomography: a multivariate analysis. treated uric acid stones. Urology 59, 206–210 requirements. Urology 78, 721.e13–721.e17
Eur. Radiol. 15, 2238–2243 (2005). (2002). (2011).
219. El‑Nahas, A. R., El‑Assmy, A. M., Mansour, O. 239. Trinchieri, A., Esposito, N. & Castelnuovo, C. 263. Qaseem, A., Dallas, P., Forciea, M. A., Starkey, M.
& Sheir, K. Z. A prospective multivariate analysis Dissolution of radiolucent renal stones by oral & Denberg, T. D. Dietary and pharmacologic
of factors predicting stone disintegration by alkalinization with potassium citrate/potassium management to prevent recurrent nephrolithiasis in
extracorporeal shock wave lithotripsy: the value of bicarbonate. Arch. Ital. Urol. Androl. 81, 188–191 adults: a clinical practice guideline from the American
high-resolution noncontrast computed tomography. (2009). College of Physicians. Ann. Intern. Med. 161,
Eur. Urol. 51, 1688–1693; discussion 1693–1694 240. Koide, T., Yoshioka, T., Yamaguchi, S., Utsunomiya, M. 659–667 (2014).
(2007). & Sonoda, T. A strategy of cystine stone management. 264. Tiselius, H.‑G. Recurrence prevention in patients with
220. Müller-Mattheis, V. G., Schmale, D., Seewald, M., J. Urol. 147, 112–114 (1992). urinary tract stone disease. ScientificWorldJournal 4,
Rosin, H. & Ackermann, R. Bacteremia during 241. Gonzalez, R. D., Whiting, B. M. & Canales, B. K. The 35–41 (2004).
extracorporeal shock wave lithotripsy of renal calculi. history of kidney stone dissolution therapy: 50 years 265. Türk, C. et al. Guidelines on urolithiasis. European
J. Urol. 146, 733–736 (1991). of optimism and frustration with renacidin. Association of Urology [online], http://uroweb.org/
221. Dhar, N. B., Thornton, J., Karafa, M. T. & Streem, S. B. J. Endourol. 26, 110–118 (2012). wp-content/uploads/22-Urolithiasis_LR_full.pdf
A multivariate analysis of risk factors associated with 242. Pearle, M. S., Calhoun, E. A. & Curhan, G. C. Urologic (2015).
subcapsular hematoma formation following diseases in America project: urolithiasis. J. Urol. 173, 266. Chiong, E. et al. Randomized controlled study of
electromagnetic shock wave lithotripsy. J. Urol. 172, 848–857 (2005). mechanical percussion, diuresis, and inversion therapy
2271–2274 (2004). 243. Ware, J. E., Kosinski, M. & Gandek, B. SF‑36 Health to assist passage of lower pole renal calculi after shock
222. Aboumarzouk, O. M., Kata, S. G., Keeley, F. X., Survey: Manual and Interpretation Guide wave lithotripsy. Urology 65, 1070–1074 (2005).
McClinton, S. & Nabi, G. Extracorporeal shock wave (Quality Metric Inc., 2003). 267. Lee, S. W.‑H., Chaiyakunapruk, N., Chong, H.‑Y.
lithotripsy (ESWL) versus ureteroscopic management 244. Bensalah, K. et al. Determinants of quality of life & Liong, M.‑L. Comparative effectiveness and safety
for ureteric calculi. Cochrane Database Syst. Rev. 5, for patients with kidney stones. J. Urol. 179, of various treatment procedures for lower pole renal
CD006029 (2012). 2238–2243; discussion 2243 (2008). calculi: a systematic review and network meta-analysis.
223. Preminger, G. M. et al. 2007 guideline for the 245. Bryant, M. et al. Health related quality of life for stone BJU Int. 116, 252–264 (2015).
management of ureteral calculi. J. Urol. 178, formers. J. Urol. 188, 436–440 (2012). 268. Leong, W. S., Liong, M. L., Liong, Y. V., Wu, D. B.‑C.
2418–2434 (2007). 246. Penniston, K. L. & Nakada, S. Y. Health related quality & Lee, S. W. H. Does simultaneous inversion during
224. Kourambas, J., Delvecchio, F. C., Munver, R. of life differs between male and female stone formers. extracorporeal shock wave lithotripsy improve stone
& Preminger, G. M. Nitinol stone retrieval-assisted J. Urol. 178, 2435–2440; discussion 2440 (2007). clearance: a long-term, prospective, single-blind,
ureteroscopic management of lower pole renal calculi. 247. Modersitzki, F., Pizzi, L., Grasso, M. & Goldfarb, D. S. randomized controlled study. Urology 83, 40–44
Urology 56, 935–939 (2000). Health-related quality of life (HRQoL) in cystine (2014).
225. Kamphuis, G. M., Baard, J., Westendarp, M. compared with non-cystine stone formers. Urolithiasis 269. Pace, K. T., Tariq, N., Dyer, S. J., Weir, M. J.
& de la Rosette, J. J. M. C. H. Lessons learned from 42, 53–60 (2014). & D’A Honey, R. J. Mechanical percussion, inversion
the CROES percutaneous nephrolithotomy global 248. Arafa, M. A. & Rabah, D. M. Study of quality of life and diuresis for residual lower pole fragments after
study. World J. Urol. 33, 223–233 (2015). and its determinants in patients after urinary stone shock wave lithotripsy: a prospective, single blind,
226. Akman, T. et al. Tubeless procedure is most important fragmentation. Health Qual. Life Outcomes 8, 119 randomized controlled trial. J. Urol. 166, 2065–2071
factor in reducing length of hospitalization after (2010). (2001).
percutaneous nephrolithotomy: results of univariable 249. Rabah, D. M., Alomar, M., Binsaleh, S. & Arafa, M. A. 270. Albanis, S. et al. Inversion, hydration and diuresis
and multivariable models. Urology 77, 299–304 Health related quality of life in ureteral stone during extracorporeal shock wave lithotripsy: does
(2011). patients: post-ureterolithiasis. Urol. Res. 39, it improve the stone-free rate for lower pole stone
227. Preminger, G. M. et al. Percutaneous 385–388 (2011). clearance? Urol. Int. 83, 211–216 (2009).
nephrostolithotomy vs open surgery for renal calculi. 250. Kurahashi, T. et al. Health-related quality of life in 271. Bailey, M. et al. Ultrasonic propulsion of kidney
A comparative study. JAMA 254, 1054–1058 (1985). patients undergoing lithotripsy for urinary stones. stones: preliminary results of human feasibility study.
228. Xue, W. et al. Management of single large Int. Urol. Nephrol. 40, 39–43 (2008). IEEE Int. Ultrason. Symp. 2014, 511–514 (2014).
nonstaghorn renal stones in the CROES PCNL global 251. Joshi, H. B. et al. Indwelling ureteral stents: evaluation 272. Schulz, E. et al. Disturbed urinary transport in the
study. J. Urol. 187, 1293–1297 (2012). of symptoms, quality of life and utility. J. Urol. 169, pelvi–calyceal system in calcium-oxalate stone
229. Holdgate, A. & Pollock, T. Nonsteroidal anti- 1065–1069; discussion 1069 (2003). patients. Urol. Res. 15, 109–113 (1987).
inflammatory drugs (NSAIDs) versus opioids for 252. Damiano, R. et al. Does the size of ureteral stent 273. Ahlstrand, C. & Tiselius, H. G. Recurrences during
acute renal colic. Cochrane Database Syst. Rev. 2, impact urinary symptoms and quality of life? a 10‑year follow‑up after first renal stone episode.
CD004137 (2005). A prospective randomized study. Eur. Urol. 48, Urol. Res. 18, 397–399 (1990).
230. Afshar, K., Jafari, S., Marks, A. J., Eftekhari, A. 673–678 (2005). 274. Coe, F. L., Evan, A. & Worcester, E. Pathophysiology-
& MacNeily, A. E. Nonsteroidal anti-inflammatory 253. Joshi, H. B., Adams, S., Obadeyi, O. O. & Rao, P. N. based treatment of idiopathic calcium kidney stones.
drugs (NSAIDs) and non-opioids for acute renal colic. Nephrostomy tube or ‘JJ’ ureteric stent in ureteric Clin. J. Am. Soc. Nephrol. 6, 2083–2092 (2011).
Cochrane Database Syst. Rev. 6, CD006027 (2015). obstruction: assessment of patient perspectives using 275. Evan, A. P. et al. Mechanism of formation of human
231. Serinken, M. et al. Intravenous paracetamol versus quality-of‑life survey and utility analysis. Eur. Urol. 39, calcium oxalate renal stones on Randall’s plaque.
morphine for renal colic in the emergency department: 695–701 (2001). Anat. Rec. (Hoboken) 290, 1315–1323 (2007).
a randomised double-blind controlled trial. 254. Sahin, C. et al. Do the residual fragments after shock 276. Tiselius, H.‑G., Lindbäck, B., Fornander, A.‑M.
Emerg. Med. J. 29, 902–905 (2012). wave lithotripsy affect the quality of life? Urology 84, & Nilsson, M.‑A. Studies on the role of calcium
232. Papadopoulos, G. et al. Hyoscine N‑butylbromide 549–554 (2014). phosphate in the process of calcium oxalate crystal
(Buscopan®) in the treatment of acute ureteral colic: 255. Lingeman, J. E., McAteer, J. A., Gnessin, E. formation. Urol. Res. 37, 181–192 (2009).
what is the evidence? Urol. Int. 92, 253–257 (2014). & Evan, A. P. Shock wave lithotripsy: advances in 277. Tsujihata, M. Mechanism of calcium oxalate renal
233. Worster, A. S. & Bhanich Supapol, W. Fluids and technology and technique. Nat. Rev. Urol. 6, 660–670 stone formation and renal tubular cell injury.
diuretics for acute ureteric colic. Cochrane Database (2009). Int. J. Urol. 15, 115–120 (2008).
Syst. Rev. 2, CD004926 (2012). 256. Rajaian, S. et al. Outcome of shock wave lithotripsy as 278. Krambeck, A. E. et al. Current computed tomography
234. Picozzi, S. C. M. et al. Management of ureteral calculi monotherapy for large solitary renal stones (>2 cm in techniques can detect duct of Bellini plugging but not
and medical expulsive therapy in emergency size) without stenting. Indian J. Urol. 26, 359–363 Randall’s plaques. Urology 82, 301–306 (2013).
departments. J. Emerg. Trauma Shock 4, 70–76 (2010). 279. Tiselius, H.‑G. Should we modify the principles of risk
(2011). 257. Zanetti, G., Trinchieri, A., Montanari, E. & Rocco, F. evaluation and recurrence preventive treatment of
235. Campschroer, T., Zhu, Y., Duijvesz, D., Grobbee, D. E. SWL: our twenty-four year experience. Arch. Ital. Urol. patients with calcium oxalate stone disease in view
& Lock, M. T. W. T. Alpha-blockers as medical Androl. 80, 21–26 (2008). of the etiologic importance of calcium phosphate?
expulsive therapy for ureteral stones. Cochrane 258. Srisubat, A., Potisat, S., Lojanapiwat, B., Urolithiasis 43 (Suppl. 1), 47–57 (2015).
Database Syst. Rev. 4, CD008509 (2014). Setthawong, V. & Laopaiboon, M. Extracorporeal 280. Bandeira, F. et al. Bone markers and osteoporosis
236. Pickard, R. et al. Medical expulsive therapy in adults shock wave lithotripsy (ESWL) versus percutaneous therapy. Arq. Bras. Endocrinol. Metabol. 58,
with ureteric colic: a multicentre, randomised, nephrolithotomy (PCNL) or retrograde intrarenal 504–513 (2014).
placebo-controlled trial. Lancet 386, 341–349 surgery (RIRS) for kidney stones. Cochrane Database
(2015). Syst. Rev. 11, CD007044 (2014). Acknowledgements
Perhaps the most controversial publication of the 259. Skolarikos, A. et al. Outcomes of flexible Research funding to S.R.K. is provided by NIH grant numbers
decade in the kidney stone arena, the authors of ureterorenoscopy for solitary renal stones in the RO1‑DK078602 and RO1‑DK092311. S.R.K. thanks
this extremely well-done randomized controlled CROES URS global study. J. Urol. 194, 137–143 P. J. Khan for her assistance in preparing the manuscript.
trial (of tamsulosin, nifedipine and placebo) showed (2015).
that medical expulsive therapy does not alter stone 260. Donaldson, J. F. et al. Systematic review and meta- Author contributions
interventions for patients after 4 weeks of stone analysis of the clinical effectiveness of shock wave Introduction (S.R.K.); Epidemiology (M.S.P.); Mechanism/
passage and declared that these agents should not lithotripsy, retrograde intrarenal surgery, and pathophysiology (W.G.R. and S.R.K.); Diagnosis, screening
be offered to patients with ureteric colic who are percutaneous nephrolithotomy for lower-pole renal and prevention (G.G.); Management (G.G. and B.K.C.);
managed expectantly. stones. Eur. Urol. 67, 612–616 (2015). Quality of life (S.D. and O.T.); Outlook (H.G.T.); Overview of
237. Furyk, J. S. et al. Distal ureteric stones and 261. Ozgor, F. et al. Clinically insignificant residual the Primer (S.R.K.).
tamsulosin: a double-blind, placebo-controlled, fragments after flexible ureterorenoscopy:
randomized, multicenter trial. Ann. Emerg. Med. 67, medium‑term follow‑up results. Urolithiasis 42, Competing interests statement
86–95.e2 (2016). 533–538 (2014). The authors declare no competing interests.

22 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
CORRECTION

CORRECTION

Kidney stones
Saeed R. Khan, Margaret S. Pearle, William G. Robertson, Giovanni Gambaro, Benjamin K. Canales,
Steeve Doizi, Olivier Traxer and Hans-Göran Tiselius
Nature Reviews Disease Primers 2, 16008 (2016)
In the version of the article originally published, a typographical error has now been corrected. The statement now reads:
Exposure of vascular smooth muscle cells to high levels of calcium and phosphate triggers this transformation154,155,
which activates bone morphogenetic proteins (BMPs) and WNT signalling pathways via upregulation of Runt-related
transcription factor 2 (RUNX2) and msh homeobox 2 (MSX2) transcription factors156.

NATURE REVIEWS | DISEASE PRIMERS www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like