SlideShare a Scribd company logo
www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 1
Review
Lancet Diabetes Endocrinol 2016
Published Online
March 2, 2016
http://dx.doi.org/10.1016/
S2213-8587(16)00033-4
Nephrology Section,
Department of Internal
Medicine, Ghent University
Hospital, Ghent, Belgium
(Prof RVanholder MD,
Prof G Glorieux PhD);
Department of Nephrology-
Nutrition-Dialysis, Centre
Hospitalier Lyon Sud,
Carmen-CENS, Université
Claude Bernard Lyon 1, Lyon,
France; F-CRIN-INI-CRCT,
France (Prof D Fouque MD);
Department of Internal
Medicine IV, Saarland
University Medical Centre,
Homburg, Germany
(Prof G H Heine MD);
Department of Medicine,
Division of Nephrology, Koc
University School of Medicine,
Istanbul,Turkey
(Prof M Kanbay MD);
Nephrology, Dialysis and
Transplantation Unit, and
CNR-IFC Clinical Epidemiology
and Pathophysiology of Renal
Diseases and Hypertension,
Ospedali Riuniti, Reggio
Calabria, Italy
(Prof F Mallamaci MD,
Prof C Zoccali MD); Division of
Nephrology, Ambroise Paré
University Hospital (APHP),
University of Paris Ouest,
Versailles-Saint-Quentin-en-
Yvelines (UVSQ),
Boulogne-Billancourt, Paris,
France (Prof Z A Massy MD);
INSERM U1018, Research
Centre in Epidemiology and
Population Health (CESP),
UVSQ,Villejuif, France
(Prof Z A Massy); Division of
Nephrology, IIS-Fundacion
Jimenez Diaz, Madrid, Spain
(Prof A Ortiz MD); INSERM
Centre d’Investigations
Cliniques (CIC)-1433, and
INSERM U1116, Nancy, France
(Prof P Rossignol MD); Institut
Lorrain du Cœur et des
Vaisseaux, CHU Nancy,
Vandoeuvre lès Nancy, France
(Prof P Rossignol); Université de
Clinical management of the uraemic syndrome in chronic
kidney disease
RaymondVanholder, Denis Fouque, Griet Glorieux, Gunnar H Heine, Mehmet Kanbay, Francesca Mallamaci, Ziad A Massy, Alberto Ortiz,
Patrick Rossignol, AndrzejWiecek, Carmine Zoccali, Gérard Michel London, for the European Renal Association European Dialysis and
Transplant Association (ERA-EDTA) European Renal and Cardiovascular Medicine (EURECA-m) working group
The clinical picture of the uraemic syndrome is a complex amalgam of accelerated ageing and organ dysfunction,
which progress in parallel to chronic kidney disease. The uraemic syndrome is associated with cardiovascular disease,
metabolic bone disease, inflammation, protein energy wasting, intestinal dysbiosis, anaemia, and neurological and
endocrine dysfunction. In this Review, we summarise specific, modern management options for the uraemic
syndrome in chronic kidney disease. Although large randomised controlled trials are scarce, based on data from
randomised controlled trials and observational studies, as well as pathophysiological reasoning, a therapeutic
algorithm can be developed for this complex and multifactorial condition, with interventions targeting several
modifiable factors simultaneously.
Introduction
Kidney failure is associated with deterioration of body
functions. The clinical picture as a whole—the uraemic
syndrome—is named after urea, the most abundant
metabolite retained in kidney failure and the first uraemic
retention product identified. The uraemic syndrome can
be caused by chronic kidney disease or acute kidney
injury, and affects almost every organ system (panel 1).1
The syndrome results from the biological effects of
metabolites that are not excreted or metabolised by the
kidneys and are retained within the body.2
Such
metabolites are named uraemic retention products, or
uraemic toxins if they exert biological or toxic effects. The
deterioration of renal endocrine function (production of
erythropoietin, active vitamin D, or renin), the
deregulation of kidney electrolyte homoeostasis, and
functional alterations resulting from chronic kidney
disease and its causes (eg, diabetes, autoimmune
disorders) also contribute to the syndrome. The clinical
picture worsens with kidney failure, with coma and death
(end-stage kidney disease; table 1) the ultimate result if the
patient is left untreated. However, since the 1940s, renal
replacement therapies (dialysis or transplantation) have
extended the life expectancy of patients with this
potentially fatal condition.
Although dialysis and transplantation extend the life
expectancy of patients with uraemia, mortality remains
substantially higher than in age-matched populations
with normal kidney function;3,4
general and cardiovascular
mortality tend to rise even before patients need dialysis.5,6
In this Review, we discuss several therapeutic options to
treat the consequences of the uraemic syndrome in
chronic kidney disease, based on the pathophysiology of
the uraemic syndrome and taking into account newly
detected, pathological pathways.
Although we have followed the principles of evidence-
based medicine as much as possible in this Review,
much of the data cited are from observation studies.
Randomised controlled trials in kidney disease are
scarce,7
and many studies have had negative results.8
This fact is largely attributable to the complex and
multifactorial nature of the disease, which makes it
difficult to recruit large patient groups with uniform
pathophysiological backgrounds. Additionally, because
of multi-layered pathophysiology, the effect of
therapeutic options that correct one aspect of disease
(eg, hypercholesterolaemia) can be masked by the effect
of other factors (eg, hypertension, fluid overload) on
outcome measures.
Traditional and non-traditional risk factors
Cardiovascular and non-cardiovascular mortality contri-
bute equally to the high mortality seen in people with
chronic kidney disease.9
Socioeconomic and
geographical factors, including access to therapy,
explain the variable mortality in chronic kidney disease
and end-stage kidney disease populations.10
Cardio-
vascular disease in patients with chronic kidney disease
is characterised by immunity-driven inflammatory
changes that cause vessel wall stiffening, arteriopathy,
and cardiomyopathy leading to heart failure,
arrhythmia, and cardiac arrest. Risk factors span from
traditional (Framingham) factors to an expanding list of
non-traditional risk factors (panel 2).8
Among non-
cardiovascular causes of death, infection, cancer,
cachexia, suicide, and refusal of treatment account for
the largest share of fatalities.9
The gap between the predictive value of traditional
risk factors and real cardiovascular mortality in chronic
kidney disease11
is to a large extent shown by indicators
of kidney dysfunction, such as estimated glomerular
filtration rate (eGFR) and albuminuria.12
Hence, factors
related to a decline in kidney function, such as
subclinical volume expansion and uraemic solute
retention, might play a part in this process. Many factors
that are affected by the uraemic status have been
associated with causes of cardiovascular damage, such
as inflammation, oxidative stress, macrophage
infiltration, endothelial dysfunction, thrombogenesis,
arterial calcification, or osteodystrophy.
2 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4
Review
Lorraine, Nancy, France
(Prof P Rossignol); Association
Lorraine pour leTraitement de
l’Insuffisance Rénale,
Vandoeuvre lès Nancy, France
(Prof P Rossignol); Department
of Nephrology,Transplantation
and Internal Medicine, Medical
University of Silesia, Katowice,
Poland (Prof AWiecek MD); and
INSERM U970, Hôpital
Européen Georges Pompidou,
Paris (Prof G M London MD)
Correspondence to:
Prof RaymondVanholder,
Nephrology Section, University
Hospital, De Pintelaan
185, B9000 Ghent, Belgium
raymond.vanholder@ugent.be
See Online for appendix
Uraemic retention products
Information about uraemic retention products has
increased continuously in the past few decades. At least
150 uraemic retention products have been described so
far,13,14
and with developments in metabolomics and
proteomics in the past decade, each new study has the
potential to add dozens of new substances to this list.
The idea that removal of one single solute would be
sufficient to solve the problem of uraemic toxicity has
long since been abandoned. By contrast, the notion that
all these molecules interact is developing.15,16
In the past 30 years, based on the physicochemical
characteristics that affect their elimination during
dialysis (the main removal strategy until now), uraemic
retention products have been divided into three major
categories: small, water-soluble compounds; protein-
bound compounds; and larger, middle molecules.13
The
characteristics of these classes and some of the main
compounds in the groups are summarised in the appendix.
Pathophysiology
Scope of Review
Since the pathophysiology of the uraemic syndrome
affects the function of almost every organ (panel 1), this
Review is restricted to elements for which relevant
information is available and that imply specific therapeutic
approaches. Definition of which pathophysiological events
are directly caused by uraemic retention products and
which, as a whole or in part, are caused by other aspects of
kidney dysfunction is not always easy. We will summarise
which elements are linked to uraemic retention and which
ones to kidney dysfunction. Since the links are sometimes
very complex, we can only focus on what we deem the
most important aspects.
Progression of kidney failure
Progression of chronic kidney disease varies depending
on the underlying cause, disease-specific pathology, and
predisposing risk factors (figure 1).17
Independent of the
initial cause, any loss of functional kidney parenchyma
leads to compensatory hyperfiltration and intraglomerular
hypertension of the remaining nephrons, which causes
fibrosis and progressive decline in kidney function.
Furthermore, uraemia-specific and non-specific nephro-
toxins, inflammation, tissue ischaemia, and pro-coagulant
mechanisms damage glomeruli and tubules. Proteinuria
increases the risk of chronic kidney disease progression
by causing tubular injury that leads to inflammatory
macrophage infiltration and tubulointerstitial fibrosis.
Angiotensin II release increases intraglomerular pressure
and oxidative stress, which alters podocyte function and
promotes synthesis of chemokines and cytokines.18
Several uraemic retention products have been linked to
progression of kidney disease such as indoxyl sulfate,
p-cresyl sulfate, asymmetrical dimethylarginine (ADMA),
and several cytokines. Factors not linked to uraemic toxins
are exacerbations of primary kidney disease, acute kidney
injury, proteinuria, hyperglycaemia, external nephrotoxic
agents (radiocontrast, non-steroidals), compensatory
hyperfiltration, and ischaemia. Hypertension is a partly
retention-independent factor—ie, it can be caused by
uraemic toxins or by other factors (figure 1).
Inflammation, metabolic bone disease, and
cardiovascular disease
Non-traditional risk factors, such as low-grade
inflammation and oxidative stress play a fundamental part
in chronic kidney disease-associated cardiovascular
complications, and to a larger extent than in the general
population.19
A key element in this proinflammatory
status is the activation of the redox-sensitive nuclear
transcription factor kappa B (NF-κB) (figure 2), in response
to several factors including oxidative stress (reactive
oxygen species [ROS]), mitochondrial dysfunction,
Panel 1: Consequences of the uraemic syndrome
Cardiovascular
Hypertension, fluid overload, cardiac decompensation,
vascular damage and stiffness, cardiovascular events,
pericarditis
Haematological
Anaemia, erythrocyte fragility, immune dysfunction
(susceptibility to infections, low response to vaccination),
inflammation, hypercoagulability, bleeding tendency
Endocrine
Hyperparathyroidism, insulin resistance, impotence,
infertility, thyroid dysfunction, hyperaldosteronism, growth
disturbance, adipokine dysbalance, klotho deficiency and
FGF-23 excess, active vitamin D deficiency
Osteoarticular problems
Osteomalacia, osteodystrophy, adynamic bone disease,
β2-microglobulin amyloidosis, muscle weakness, fractures,
bone pain, calciphylaxis and cardiovascular calcification
Neurological
Polyneuropathy, coordination disturbances, tremor, cognitive
dysfunction, decreased attention span, coma
Gastrointestinal
Anorexia, gastroparesis, nausea, vomiting
Dermatological
Skin atrophy, pruritus, calciphylaxis
Stomatological
Periodontitis, stomatitis
Nephrological
Renal tubular damage, progression of kidney failure
Other
Malnutrition, changes in drug protein binding, changes in
metabolism, hyperkalaemia, metabolic acidosis
www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 3
Review
several uraemic retention products, infection, vasoactive
substances, dyslipidaemia, and inflammation. Activation
of NF-κB is associated with the release of proinflammatory
cytokines (tumour necrosis factor α, interleukin 1β,
interleukin 6, soluble tumour necrosis factor-like weak
inducer of apoptosis) and activation of monocyte
chemoattractant protein-1 and profibrotic transforming
growth factor β1.
Inflammation and oxidative stress are tightly
interconnected. NF-κB activation initiates production of
ROS and vice versa, but under normal conditions,
nuclear factor-erythroid-2-related factor 2 (a regulator of
resistance to oxidants via expression of antioxidants
and cytoprotective agents) is also activated. This balance
might be disrupted in chronic kidney disease.20
A substantial number of uraemic toxins (eg, indoxyl
sulfate, p-cresyl sulfate, the cytokines, advanced glycation
end products, trimethylamine N-oxide) are pro-
inflammatory, as are compounds that until 5 years ago
were thought to be inert, like urea (for a more
comprehensive list, see appendix). Furthermore, several
other factors that are inflammatory are partly or entirely
unrelated from uraemic retention products: infection,
dialysis fluid contamination, dialysis bioincompatibility,
concomitant diseases (eg, autoimmune disorders), and
vasoactive agents (figure 2).
Cardiac and arterial complications are the principal
complications of the uraemic syndrome. The
cardiomyopathy in chronic kidney disease is characterised
by left ventricular hypertrophy, capillary rarefaction, and
interstitial fibrosis, with predominant diastolic dys-
function. Arrhythmias, sudden death, and heart failure
are the main consequences of these cardiac com-
plications. NF-κB activation by vasoactive substances
affects cardiomyocyte growth, fibrosis, and apoptosis,
inducing hypertrophy.19
In addition to the proinflammatory factors mentioned
above, some uraemic toxins and several factors that are
independent of uraemic toxins are damaging for heart
and vessels: indoxyl sulfate, ADMA, endothelin, oxalate
(uraemic retention products), hypertension, sodium and
fluid retention, primary diseases (eg, amyloidosis),
infections (endocarditis).
Compared with the general population, arterial disease
in populations with chronic kidney disease is premature,
and characterised by endothelial dysfunction, arterial
remodelling with arterial calcifications (figure 3A, 3B),
vascular stiffness, and the transition of the vascular
smooth muscle cells to an osseous phenotype.21
Vascular
stiffness, which is closely linked to endothelial
dysfunction, precedes arterial calcifications that further
exacerbate stiffening.22
Arterial calcifications are due to an
GFR* Definition Therapeutic approaches Pathophysiological changes Mortality
Stage 1 >90 Normal kidney function, but
urine abnormalities (eg,
haematuriaor
albuminuria†), proven
structural kidneydamage,
or genetictrait
Observation, blood pressure and
cardiovascular risk factor control,
preventionof cardiovasculardisease
Klothodeficiency Increasedoverall and
cardiovascular
mortality +
Stage 2 60–89 Mildly reduced kidney
function associatedwith
other findings (as in stage 1)
Observation, blood pressure and
cardiovascular risk factor control,
preventionof cardiovasculardisease
Klothodeficiency, elevated FGF-23 Increasedoverall and
cardiovascular
mortality +
Stage 3A
Stage 3B
45–59;
30–44
Moderately reduced kidney
function
Observation, blood pressure and
cardiovascular risk factor control,
preventionof cardiovasculardisease,
treatmentof anaemia and metabolic bone
disease, preventionof chronic kidney
disease progression
Klothodeficiency, elevated FGF-23
and PTH, malnutrition, hypertension,
left ventricular hypertrophy, anaemia
Increasedoverall and
cardiovascular
mortality ++
Stage 4 15–29 Severely reduced kidney
function
Planning for renal replacementtherapy,
blood pressure and cardiovascular risk
factor control, preventionof cardiovascular
disease,treatmentof anaemia and
metabolic bonedisease, preventionof
chronic kidneydisease progression
Klothodeficiency, elevated FGF-23
and PTH, malnutrition, hypertension,
left ventricular hypertrophy, anaemia,
hypertriglyceridaemia,
hyperphosphataemia, metabolic
acidosis, hyperkalaemia
Increasedoverall and
cardiovascular
mortality +++
Stage 5 <15 Very severe kidney failureor
end-stage kidneydisease
Renal replacementtherapyor conservative
approach, blood pressure and
cardiovascular risk factor control,
preventionof cardiovasculardisease,
treatmentof anaemia and metabolic bone
disease
Klothodeficiency, elevated FGF-23
and PTH, malnutrition, hypertension,
left ventricular hypertrophy, anaemia,
hypertriglyceridaemia,
hyperphosphataemia, metabolic
acidosis, hyperkalaemia, need for
renal replacementtherapy
Increasedoverall and
cardiovascular
mortality ++++
FGF-23=fibroblast growth factor-23. GFR=glomerular filtration rate (mL/min per 1·73 m²). PTH=parathyroid hormone. *GFR can be directly measured (eg, clearance of inulin,
iothalamate, or ethylene diamino acetic acid), or calculated based on serum markers, such as creatinine, cystatin C, or both and anthropometric parameters (estimated GFR–
eGFR). †Albuminuria is classified as moderately increased (30–300 mg/g or 3–30 mg/mmol creatinine) and severely increased (>300 mg/g or >30 mg/mmol) and increases
risk by one level unless the highest risk category has been reached. Severity of mortality is indicated as mild (+), mild-moderate (++), moderate (+++), and severe (++++).
Table 1: Chronic kidney disease stages and their implications
4 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4
Review
imbalance of calcification inducers and inhibitors, and
development of a senescence-associated secretory
phenotype (SASP) in vascular smooth muscle cells.23
Inducers of arterial calcifications and SASP are all linked
to secretion of proinflammatory cytokines. Inflammation
activates the BMP2:BMP4, Msx2, and Wnt pathways,
which promote increased transcription of genes associated
with conversion of vascular smooth muscle cells to the
osteochondrogenic phenotype (Runx2, osterix, Sox9).24
Several chronic kidney disease-linked and biochemical
changes related to mineral bone disorder (figure 3C), such
as hyperphosphataemia, are associated with arterial
calcifications. Hyperphosphataemia also stimulates the
production of ROS and thereby activation of NF-κB. The
resultant inflammation decreases the concentration of
calcification inhibitors and reduces klotho expression,
which induces resistance to the phosphaturic effect
of fibroblast growth factor 23 (FGF-23),25
bone resistance
to parathyroid hormone (PTH), secondary hyperpara-
thyroidism,26
and accelerated ageing.27
Telomere
shortening, which is typical for ageing, is associated with
cardiovascular disease manifestations.28
Inflammatory
epigenetic changes, which, in part, involve inhibited gene
methylation of s-adenosinemethionine, also contribute to
inflammation and cardiovascular lesions.29,30
Uraemic bone disease is mainly triggered by increased
concentrations of the uraemic toxin phosphate, which
results in the enhanced generation of several
compensatory factors to maintain serum phosphate con-
centration, such as FGF-23 and PTH, at the cost of loss of
bone structure and function. Independent of metabolite
retention, ageing, inflammation, bone ischaemia, and
inadequate renal production of active vitamin D
metabolites further enhance bone degradation. Some
patients develop adynamic bone disease, whereby low
PTH hinders calcium deposition in the bone, favouring
vascular calcification. In rare cases, patients develop
calciphylaxis—a severe syndrome consisting of vascular
calcification, thrombosis, and skin necrosis.
In summary, inflammation, oxidative stress, and bone
disease show specific features linked to chronic kidney
disease and uraemia resulting in accelerated vascular
damage and ageing.
Malnutrition and dysbiosis
Malnutrition and dysbiosis are two features of the
uraemic syndrome that relate to digestive function
(figure 4). Some uraemic retention products (such as
cytokines leptin, ghrelin, and neuropeptide Y) can be
implicated in reduced appetite and nutrient intake.
p-cresyl sulfate causes insulin resistance, disturbances of
lipid metabolism, and aberrant distribution of fat cells
throughout the body. Inflammation—a common epi-
phenomenon of chronic kidney disease—is associated
with reduced appetite, at all chronic kidney disease stages,
including chronic kidney disease stage 5 on dialysis.31
Other factors implicated in reduced appetite, independent
of solute retention, are changes in taste and smell
perception, reduced intestinal motility and absorption
capacity, depression, and loss of aminoacids and protein
via dialysis (figure 4). Consequently, patients might have a
spontaneous reduction in energy intake, which can
induce protein energy wasting even before starting
dialysis (figure 3D). Although decreased protein intake
could reduce generation of uraemic retention products,
Panel 2: Non-traditional risk factors in the uraemic syndrome5
• Anaemia
• Volume overload
• Metabolic bone disorder and related mediators
• Hyperphosphataemia
• High levels of FGF-23
• Low expression of the anti-ageing factor α-klotho
• Secondary hyperparathyroidism
• Active vitamin D deficiency
• Low grade inflammation
• Oxidative stress
• Uraemic retention products*
• Post-translational protein modifications
• Accumulation of atherogenic remnant lipoproteins
• Endogenous nitric oxide synthase (NOS) inhibitors
• High sympathetic activity
• Insulin resistance
• Exposure to bioincompatible dialysis conditions
• Immune response activating membranes
• Contaminated haemodialysate
• Peritoneal dialysate containing glycation products
• Haemodynamic instability during haemodialysis
• Infections
• General
• Vascular access: arteriovenous fistula, catheters, and arteriovenous grafts
• Dyskalaemia (hypokalaemia and hyperkalaemia)
*For details, see appendix.
Figure 1: Role of progression of kidney failure in defining the uraemic syndrome
↑ Angiotensin II
↑ Catecholamines
↑ Endothelin
Glomerular hypertension
and hyperfiltration
Podocyte injury and
proteinuria
Glomerular and
tubulo-interstitial
fibrosis
Progressive kidney
dysfunction
Inflammation
Uraemic retention products
Chronic
kidney
disease
www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 5
Review
muscle protein catabolism, indirectly caused by acidosis
and inflammation, might enhance the accumulation of
toxic metabolites. Thus, a diet containing the appropriate
nutrients in the appropriate quantity and balance is
crucial for wellbeing and to reduce metabolic poisoning
in patients with chronic kidney disease.
The intestinal microbiota plays an important part in
regulating the host’s nutritional status, metabolism, and
different aspects of immunity. Dysbiosis has been linked
to several chronic diseases. The few studies of dysbiosis
in patients with chronic kidney disease suggest changes
in the abundance and metabolic characteristics of
intestinal microbiota as compared with healthy
individuals.32
These changes induce the preferential and
abundant generation of uraemic retention products with
toxic effects. Additionally, chronic kidney disease and
uraemic retention cause structural and functional
changes of the intestinal epithelial barrier, inducing
inflammation via different pathways.33
Although
progression of kidney disease is clearly linked to changes
in intestinal microbiota, whether or not uraemic
retention products play a part in these changes is not
clear. Other conditions that are associated with kidney
disease could also affect intestinal microbiota—eg,
ageing, diabetes, obesity, changes in nutrient intake and
diet, and antibiotic intake.
Anaemia
Kidney dysfunction is associated with decreased
concentrations of haemoglobin in blood, mainly due to
deficiency of and resistance to erythropoietin and absolute
and relative iron deficiency (figure 5). The kidneys
Figure 2: Key factors in the microinflammatory condition of chronic kidney disease
All elements lead to the activation of NF-κB and the expression of several proinflammatory factors. ROS=reactive oxygen species. AGEs=advanced glycation end products. ADMA=asymmetrical
dimethylarginine. IS=indoxyl sulfate. pCS=p-cresyl sulfate.TMAO=trimethylamine-N-oxide. LPS=lipopolysaccharide. NF-κB=nuclear factor kappa B. OxLDL=oxidised low density lipoprotein.
MCP-1=monocyte chemoattractant protein-1.TGF-β=transforming growth factor β.
ROS Uraemic retention
products
AGEs, ADMA, IS, pCS,
TMAO, phosphate
Infectious agents
Infection,
endotoxin (LPS),
bacterial DNA,
peptidoglycan
Vasoactive agents
Angiotensin II,
noradrenaline,
endothelin I,
aldosterone
Cytokines and chemokines
Proinflammatory (eg, interleukin 6, MCP-1),
profibrotic (eg,TGF-β)
↑NF-κB
Mitochondrial
dysfunction
Dialysis-related
factors
Bioincompatibility,
dialysis fluid impurities
Dyslipidaemia
OxLDL
Figure 3:Typical clinical problems related to the uraemic syndrome
Vascular calcifications (A, B); rugger-jersey spine: alternating osteosclerosis and osteopenia (C); uraemic malnutrition (D); central lesions: silent infarctions (E),
microbleeds (F); white matter lesions (G).
A B C
E F G
D
6 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4
Review
respond to low tissue oxygen by increasing erythropoietin
production, which induces erythropoiesis. Decline in
kidney function impedes this mechanism. Iron deficiency
is primarily attributable to ineffective intestinal iron
absorption, because inflammation associated with
chronic kidney disease elevates hepcidin, an inhibitor of
iron export from enterocytes to plasma. Insufficient oral
iron intake and blood loss from occult gastrointestinal or
urogenital bleeding, repetitive blood sampling, and
dialysis further drive iron deficiency. Finally, vitamin B12
and folate deficiency, shortened erythrocyte lifespan, and
frequent infections are also contributing factors.34
Erythropoietin and iron deficiencies are the main
drivers of renal anaemia, and these effects are largely
independent of uraemic retention; however, uraemic
toxins can affect erythrocyte production, solidity, and
survival (figure 5).
Neurological dysfunction
High incidence of cerebrovascular diseases (such as
stroke, white matter lesions, and intracerebral
microbleeds; figure 3E–G) and cognitive disorders
(linked to vascular abnormalities, neurodegenerative
abnormalities, or both; figure 6) have been reported in
chronic kidney disease.35
Cerebrovascular disorders
could be linked to traditional and non-traditional
cardiovascular risk factors, such as oxidative stress,
chronic inflammation, endothelial dysfunction, vascular
calcification, anaemia, dialysis-related haemodynamic
instability, and uraemic retention products.36
Uraemic
toxins could affect cerebrovascular diseases or cognition
either by direct functional effects or by modulation of
other factors (such as inflammation and oxidative
stress). β2-microglobulin—a typical uraemic toxin—
was shown to impair cognitive function and act as a
systemic, pro-ageing factor.37
Other uraemic retention
products frequently associated with neurological
dysfunction are guanidines. Phenols and indoles have
also been linked to neurological disturbances. Further
contributing factors that are partly linked to retention
are inflammation, vascular disease, and hypertension.
Factors that are more or less independent of solute
retention are depression, enhanced activity of drugs that
treat psychiatric disorders or have psychological effects
(eg, sleeping pills), and blood pressure changes related
or not to dialysis (figure 6).
Management
The studies, guidelines, position statements, and reviews
referred to in this section are summarised in the
appendix. Treatment aimed at modifying risk factors will
be discussed in broad terms independently of underlying
intermediate mechanisms. Of note, positive interventions
in the general population, could counterintuitively cause
or amplify complications in chronic kidney disease. This
section on management includes 56 original studies
(appendix), of which only 35 (63%) are randomised
controlled trials or meta-analyses. Only six (11%) of these
randomised controlled trials or meta-analyses have an
unambiguously positive intervention in a study taking
into account more than 1000 patients (all but one of these
six are meta-analyses).
Lifestyle
Emphasis is often laid on pharmacological drug
interventions in cardiovascular and kidney protection in
chronic kidney disease, of which the benefit is not
necessarily unequivocal. Several general lifestyle
measures could also be beneficial, and need negligible
societal investment, except for educational initiatives.
In a large meta-analysis, regular exercise training was
associated with improved outcomes in chronic kidney
disease. However, risk of bias of the included studies was
high.38
In a single-centre, observational study, heavy
smoking was associated with increased risk of
progression of chronic kidney disease, especially in
patients with hypertensive and diabetic nephropathy.39
In
a large, cross-sectional follow-up study, no significant
Figure 4: Role of malnutrition and dysbiosis and chronic kidney disease in defining the uraemic syndrome
Figure 5: Role of renal anaemia and chronic kidney disease in defining the uraemic syndrome
↓ Nutritional status
↑ InflammationUraemic
retention products
Chronic kidney
disease
Protein energy wasting
Intestinal modifications
and translocation
↑ Hepcidin
production
↓ Erythropoietin
generation
Iron deficiency
Inflammation,
infection
Deficient iron
intake, blood loss
Anaemia Deficiency of folic acid
and vitamin B12,
erythrocyte fragility
Chronic kidney disease
Figure 6: Role of neurological dysfunction and chronic kidney disease in defining the uraemic syndrome
Cognitive
dysfunction
Neurological
damage
Oxidative stress, inflammation,
endothelial dysfunction,
vascular calcification,
dialysis-related haemodynamic
instability
Uraemic retention
products
Chronic kidney
disease
Cerebrovascular
disease
www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 7
Review
association was recorded between pre-dialysis obesity
and progression of chronic kidney disease.40
Another
cohort study,41
however, showed a clear association
between BMI more than 30 kg/m² and progression of
kidney disease, especially from the age of 40 years on.
The relative survival advantage in patients who are
overweight and on dialysis (obesity paradox) is
remarkable, but can be attributed to poor outcomes of
their malnourished counterparts,42
partly related to the
negative effects of inflammation. Unfortunately, focused
and conclusive studies on lifestyle in chronic kidney
disease are scarce.
Diet
A key factor of the dietary management of the uraemic
syndrome is to supply enough nutrition, but not too
much, because the body needs fuel, but cannot handle
well enough the end products of cellular metabolism.
This notion is particularly true of proteins because they
cannot be stored to adjust for intake fluctuations.
Accumulation of uraemic retention products and
metabolic poisoning in chronic kidney disease can be
ameliorated by reduced protein intake, which, rather
than being left to spontaneous evolution, should be
prescribed as a well balanced, carefully controlled, low-
protein diet. Thus, patients with stages 3–5 chronic
kidney disease who are not yet on dialysis should be
educated to pursue an equilibrium between sufficient
energy intake (at least 30 kcal/kg per day) and reduced
protein intake (0·6 to 0·8 g/kg per day, 50% of which
should be high value proteins of animal origin containing
essential aminoacids (table 2).43
This approach reportedly
reduces serum urea by 30%, and improves insulin
resistance, phosphate and parathyroid metabolism,
blood pressure, and anaemia.43
Reduction of phosphorus
intake allows better control of mineral bone disease and
reduces the need for oral phosphate binders. Sodium
intake should be targeted to a maximum of 6 g sodium
chloride per day and, if possible, lower (table 2), except in
the case of salt-losing nephropathy, in which intake can
be higher. Specialised dietitians should regularly be
involved in the implementation of advice to correctly
educate the patient. At least three encounters per year
have been suggested for the first year of care.44
This only
seems practical from chronic kidney disease stage 4
onwards. 24 h urine collections allow the monitoring of
daily protein intake (via urea measurements) and sodium
intake and should be done twice yearly to control and
implement the diet. If intentional weight loss is planned,
the benefit should be balanced against the risk of protein
wasting, particularly in the late stages of chronic kidney
disease.
On dialysis, additional catabolism takes place in
response to chronic inflammation, surgical interventions,
recurrent sepsis, and nutrient losses during the dialysis
procedure. Whereas caloric intake can be maintained
above 30 kcal/kg per day, protein intake should be
increased to more than 1·0 g/kg per day of protein in
patients on haemodialysis and 1·2 g/kg per day in
peritoneal dialysis (table 2).45
In stable patients, protein intake can be calculated as
protein catabolic rate from pre-dialysis and post-dialysis
urea measurements. Fluid intake is an indicator of
nutritional status. In patients with anuria, interdialytic
weight gain was positively associated with protein catabolic
rate.46
Reduced weight gain, or a low serum urea or
creatinine pre-dialysis are negative signs and should prompt
rapid dietary intervention.47
Nevertheless, fluid intake that is
too high and interdialytic weight gain increase intradialytic
ultrafiltration rate, which should be avoided because of its
association with mortality in patients on dialysis.48
Dietary phosphate intake should be controlled without
causing a reduction in protein intake, because a reduction
in protein intake is associated with worse survival.49
Foods
and beverages rich in phosphate, such as preserves,
processed meat, frozen foods, dairy products, and soft
drinks should be discouraged. Potassium intake that is too
high should also be avoided (table 2). For more detailed
dietary advice see appendix.
Pharmacological treatment
Chronic kidney disease is a challenging disease with
few treatment options to prevent progression. In
clinical practice, measurements of albumin-to-
creatinine ratio and eGFR allow risk classification for
chronic kidney disease progression and cardiovascular
mortality. Additional biomarkers might be helpful in
refining these strategies, but their use needs further
validation.
Chronic
kidney
disease
stages 3–5,
not on
dialysis
Receiving
haemodialysis
Receiving
peritoneal
dialysis
Had
transplant
<3 months
ago
Had
transplant
≥3 months
ago
Diet Low protein Standard
protein
Standard
protein
High protein Low protein
Target protein intake
(g/kg per day)
0·6–0·8
(or less with
keto-
analogues)
1·0–1·2 1·2 1·4 0·6–0·8
Target energy intake
(kcal/kg per day)
30–35 30–35 30–35 30–35 30–35
Salt intake (mg per
day)*
<6000 <5000 <5000 <6000 <6000
Potassium intake
(mg per day)
2500 2500 2500 Free Free†
Phosphorus intake
(mg per day)
<800 <1000 <1000 Free <800
Wasting risk ++ +++ +++ +++ +
Overweight risk ++ + + ++ +++
*Except in case of salt-losing nephropathy. †Except in case of hyperkalaemia (>5·5 mmol/L). Risk of being overweight
indicated as mild (+), moderate (++), and severe (+++).
Table 2: Integrated optimum nutrition of patients with chronic kidney disease
8 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4
Review
The mainstays of non-specific prevention of chronic
kidney disease progression, irrespective of cause, include
blood pressure control and proteinuria-directed strategies
to preserve residual kidney function, with special emphasis
on angiotensin-converting enzyme (ACE) inhibitors or
angiotensin-receptor blockers.50
In diabetic nephropathy,
strategies that focus on strict glycaemic control slow down
progression.51
Although hyperlipidaemia could play a role
in progression, there is no consensus regarding statin
treatment for nephroprotection.52,53
In a randomised controlled trial54
of bicarbonate
supplementation with a target serum bicarbonate
concentration greater than 23 mmol/L, progression of
chronic kidney disease was slowed and nutritional status
improved. In another randomised controlled trial,55
reduction of uric acid by allopurinol treatment was
nephroprotective. In a trial56
in patients with diabetic
nephropathy, pentoxifylline reduced the rate of decline in
eGFR and proteinuria. The findings of these three
studies should, however, be confirmed in larger
controlled trials.
Uraemia is a strong, independent risk factor for
cardiovascular disease, therefore, patients with chronic
kidney disease need close cardiovascular follow-up as
part of their management strategy to prevent chronic
kidney disease progression.
Approaches targeting inflammation (eg, anti-
inflammatory drugs or statins) did not provide health
benefits for patients with chronic kidney disease. In the
AURORA trial,57
rosuvastatin lowered serum C-reactive
protein concentrations by 27%, but failed to reduce
mortality or the risk of cardiovascular events. Resistance
to interventions targeting inflammation could depend
on the severity of inflammation in end-stage kidney
disease (in the AURORA trial,57
median C-reactive
protein in the treatment group remained four times
above upper normal concentrations), and on its
multifactorial origin. Interference of proinflammatory
and oxidative mechanisms—by the reduction of NF-κB
activation and activation of nuclear factor-erythroid-2-
related factor 2—seemed an attractive option to reduce
the enhanced risk of all-cause and cardiovascular death
in advanced chronic kidney disease. However, a trial58
testing a NF-κB blocker (bardoxolone) in patients with
chronic kidney disease and type 2 diabetes was
prematurely terminated because of excessive risk of
cardiovascular events in the bardoxolone group.
In pre-dialysis chronic kidney disease, the well
established cardiovascular benefit of antihypertensive
treatment has led to specific guidelines recommending
target blood pressures of 140/90 mm Hg or less in
chronic kidney disease (130/80 mm Hg or less in the
presence of proteinuria).59
The relation between pre-
dialysis or post-dialysis blood pressure and mortality in
patients on dialysis is inverse or U-shaped, which is a
classical example of reverse causality. However, dialysis-
related blood pressure values hardly reflect true blood
pressure burden in patients on haemodialysis.60
Out-of-
dialysis systolic blood pressure, in fact, predicts a linear
increase in the risk of death from 110 mm Hg or higher,61
as in the general population.
Antihypertensive treatment reduces the incidence of
death and cardiovascular events at all risk levels and in
absolute terms most of all in patients with a higher
baseline risk.62
In the SPRINT trial,63
patients with
hypertension randomised to target systolic blood
pressures of less than 120 mm Hg had fewer
cardiovascular events than in those targeted to less than
140 mm Hg, in general and in the subgroup with chronic
kidney disease. In patients with end-stage kidney disease,
including patients with heart failure, the use of
antihypertensive drugs reduces mortality,64
but treatment
should be titrated to tolerable levels—ie, to minimise the
risk of hypotension by autonomic dysfunction or arterial
stiffness,65
which enhance the risk for ischaemic events.
Optimisation of volume control at constant ultrafiltration
rate (eg, by more frequent or extended dialysis), and
judicious use of antihypertensive drugs accounting for
comorbidities and pharmacokinetic profile66
could reduce
cardiovascular risk in patients on dialysis. In a single
centre-based observational study,67
extended haemo-
dialysis (24 h per week) in an older population resulted in
good blood pressure control without antihypertensive
drugs.
Controlling hyperglycaemia in patients with renal
failure and diabetes is difficult because of the higher risk
of hypoglycaemia as compared with the diabetic
population without chronic kidney disease.68
Target
HbA1c levels should account for additional risk factors,
with the aim to achieve strict control in those with low
risk, but more leniency in those with comorbidities and
high risk for hypoglycaemia.69
Patients with diabetes,
chronic kidney disease, and heart failure, ischaemic
heart disease, or hypertension, can be treated with renin–
angiotensin system blockers at a maximally tolerated
dose. However, combinations of ACE inhibitors and
angiotensin II receptor blockers should be used carefully
in patients with chronic kidney disease not yet on
dialysis69
to avoid hyperkalaemia, kidney impairment,
and hypotensive symptoms,50
although in a meta-
analysis,70
a positive effect on kidney function
preservation was suggested with this combination in
people with diabetes.
Empagliflozin, a sodium-glucose co-transporter
inhibitor, significantly reduced a composite of cardio-
vascular events and death in patients with diabetes type 2
and eGFR more than 30 mL/min per 1·73 m² at high
cardiovascular risk (26% with chronic kidney disease
stage 3).71
Because of malnutrition and inflammation, hyper-
cholesterolaemia often regresses as chronic kidney
disease advances. Identification of dyslipidaemia (high
total or LDL cholesterol, low HDL cholesterol, high
triglycerides) in patients with end-stage kidney disease is
www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 9
Review
deemed useful by existing Kidney Disease Improving
Global Outcomes (KDIGO) guidelines to assess overall
cardiovascular risk,72
but follow-up of lipid concentrations
is not recommended because of the absence of evidence
that this approach improves clinical outcomes.
Findings from the SHARP trial73
and subsequent
meta-analysis53
showed the usefulness of an absolute
reduction of LDL cholesterol in patients with chronic
kidney disease to decrease the associated cardiovascular
risk. Cholesterol-lowering therapy is especially
recommended by KDIGO in all patients with chronic
kidney disease at stage 3–5, who are more than 50 years
of age and not on dialysis.72
However, the benefit of
statins was less prominent in end-stage kidney
disease,57,74
which discouraged their use in end-stage
kidney disease, irrespective of inflammation or
malnutrition. In patients already treated, however, this
therapy can be continued.69
Results from a large end-stage kidney disease cohort in
the USA showed no survival benefit of lowering
phosphate concentrations with calcium carbonate or
acetate.75
Findings from another observational study76
showed a reduction in death risk in patients who were
given any type of phosphate binder. Results from a meta-
analysis77
of seven trials in haemodialysis patients and
one in moderate chronic kidney disease showed lower
mortality in patients given the non-calcium-based
phosphate binder sevelamer. However, this apparent
benefit (heterogeneity I²=89%) is potentially skewed by a
study in patients on haemodialysis in which sevelamer
reduced the death risk by 91%, hence, the issue remains
unresolved. Serum phosphate concentrations between
0·68 and 1·93 mmol/L do not associate with an excessive
death risk in dialysis patients.78
The decision to treat hyperphosphataemia should take
into account serum calcium, PTH, and serum vitamin D
concentrations from the early stages of chronic kidney
disease-associated mineral bone disorder.78
Secondary
hyperparathyroidism has been implicated in the
cardiovascular risk of chronic kidney disease. However,
improvement of mortality and cardiovascular outcome
was inconclusive in a large trial79
of calcium receptor
agonist cinacalcet in secondary hyperparathyroidism, and
this finding was confirmed by meta-analysis.80
Many patients with chronic kidney disease have low
serum concentrations of 25-hydroxyvitamin D or
calcitriol, but a randomised, placebo-controlled trial
testing their supplementation on hard endpoints is not
yet available. However, these supplements might help
control hyperparathyroidism. The effect of modulation of
uraemic retention product concentrations on metabolic
bone disease and cardiovascular outcomes needs to be
investigated because several of these retention products
are associated with both complications.81
Malnutrition should be diagnosed using the
International Society for Renal Nutrition and Metabolism
protein energy wasting criteria,82
which include clinical,
laboratory, and body composition measurements.
Wasting scores can predict survival and should be used
more frequently to help identify patients at risk of death
from wasting. An increase in nutrient intake is the key
intervention and has been shown to be effective even in
the context of chronic inflammation. One or two units of
oral supplements taken separately from regular meals to
avoid further reductions in spontaneous intake should be
used as a primary measure. The FINE study,83
which
compared oral nutritional supplements with or without
parenteral nutrition in dialysis patients with protein
energy wasting, was negative for its primary endpoints
(eg, mortality). Oral supplements were well tolerated and
increase total nutrient intake, serum albumin, and
serum prealbumin.83
Results of a multivariate analysis
showed that mortality was reduced if serum prealbumin
increased above 30 mg/L in the first 3 months of
supplementation.83
Therefore, in ambulatory patients, if
nutritional status does not improve within 4 weeks,
enteral feeding should be considered. For hospital
inpatients, intensive nutrition through a nasogastric tube
or percutaneous gastrostomy can be considered if oral
energy intake is less than 20 kcal/kg per day for more
than 10 days.
Few randomised controlled trials have been done that
investigate the effect of the restoration of intestinal
symbiosis in chronic kidney disease by giving patients
prebiotics (selectively fermentable ingredients),
probiotics (live biotherapeutics), or synbiotics (com-
bination of prebiotics and probiotics) to reduce
circulating concentrations of uraemic retention products,
inflammation, oxidative stress, and progression of
chronic kidney disease. Specific prebiotics decrease
serum indoxyl sulfate and urea nitrogen.84,85
Results from
a study86
in patients receiving peritoneal dialysis showed
a significant reduction in concentrations of pro-
inflammatory cytokines, an increase in concentrations of
anti-inflammatory cytokines, and better preservation of
residual kidney function in the group receiving
probiotics. Synbiotic therapy reduces the concentration
of p-cresol.87
In several of the above trials, however, the
intervention was coupled to a low-protein diet, which
could itself decrease uremic toxin concentration and
progression of kidney failure. The actual effect of
restoring intestinal symbiosis on the intestinal
microbiota profile or on hard outcomes was not assessed
in any of the above trials.
Reduction of indoxyl sulfate by intestinal sorbents, such
as AST-120, stopped progression of chronic kidney
disease in small Japanese randomised trials.88,89
This
benefit was not confirmed in a large European–American
randomised controlled trial.90
Interventions improving
symbiosis need further study before their efficacy can be
accepted.
The conventional approaches for iron repletion in
chronic kidney disease are oral ferrous salts or intravenous
colloidal compounds. High concentrations of hepcidin,
10 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4
Review
however, make intestinal iron uptake ineffective.
Additionally, oral iron intake further contributes to the
pill burden of patients with chronic kidney disease and
causes severe gastrointestinal side-effects. Therefore,
intravenous iron has gained popularity in the past 5 years.
However, experimental and observational data suggest
unwanted immunological, cardiovascular, and renal
effects of such treatment,91,92
which might differ between
classic preparations and new, more stable formulations.91
Long-term prospective studies are, however, unavailable.
The introduction of recombinant human erythropoietin
was one of the major steps forward in the treatment of
renal anaemia. Both short-acting and long-acting
erythropoiesis stimulating agents are in use at present.
According to the existing guidelines, the decision of
whether and when to start treatment with an
erythropoiesis stimulating agent should be individualised
and take into account related risks and anaemic
symptoms.34
The target values for erythropoiesis
stimulating agent therapy are 10·0–12·0 g/dL.34
These
drugs should be used with care in patients at high risk of
stroke or with active or past malignancy.34
Blood transfusions might be considered, especially in
patients with haemoglobin concentration of less than
7·0 g/dL, who are resistant to erythropoiesis-stimulating
agents, or at potential risk of complications with
erythropoiesis stimulating agent therapy.93
However, the
risk of developing panel reactive antibodies should be
taken into account in transplant candidates.
Few data are available on the effect of treatment for
chronic kidney disease on risk of stroke and other
neurological disorders in patients with this disease,
particularly in those with end-stage kidney disease.
Patients with chronic kidney disease have been under-
represented in the cardiovascular trials that prove net
benefit of commonly used preventive treatments (eg,
antihypertensive drugs, low-dose aspirin, carotid revascu-
larisation, and thromboprophylaxis for atrial fibrillation),
and safety and efficacy of many of these treatments in
chronic kidney disease remains uncertain. Conflicting
results have been reported on the effect of statins in
patients with chronic kidney disease. In a Cochrane
review,53
the authors concluded that statins did not
necessarily have effects on stroke in patients with chronic
kidney disease who did not need dialysis, by contrast
with another meta-analysis94
in which statins were
associated with a decrease in cardiovascular disease in
chronic kidney disease, including stroke. Correcting
anaemia might not prevent stroke and could even
increase its risk in chronic kidney disease.95
Moreover,
the effect of anaemia treatment on cognitive disorders
remains a matter of debate.
Oxidative stress, inflammation, or uraemic retention
products could contribute to neurological disorders in
patients with chronic kidney disease. Thus, treatments
modulating these factors might improve neurological
outcomes. To the best of our knowledge, no direct
interventional trials have targeted these abnormalities.
However, improvement of intra-dialytic haemodynamic
stability by cooling dialysate has been shown to protect
against brain injury.65
Renal replacement therapy
Replacement of kidney function requires trans-
plantation or dialysis, and haemodialysis and peritoneal
dialysis are the main dialysis modalities (for more on
different dialysis strategies see appendix). Haemodialysis
is usually done three times weekly, whereas peritoneal
dialysis clears retention products with lower efficiency
but does so continuously. The high efficiency solute
removal provided by a single haemodialysis session over
4 h should be extrapolated to the full interdialytic interval
of 48–72 h, by comparison with kidney removal capacity.
Thus, renal replacement therapy through dialysis allows
the anuric patient to have a kidney-equivalent filtration
rate of only 5–10 mL/min for small molecules that are
the size of urea or creatinine, which results in higher
uraemic solute concentrations than is normal. In
general, dialysis removes retention products of higher
molecular weight or those bound to protein less
efficiently than the normal kidney. Oral phosphate
binders are frequently needed to maintain serum
phosphate concentrations that are close to normal. The
standard dialysis clearance of small proteins such as
cytokines, FGF-23, and β2-microglobulin is negligible,
as compared with normal kidneys. Clearance of
β2-microglobulin (11·8 kDa), deemed representative of
middle-sized molecules, was estimated at 3–19 mL/min
for a conventional haemodialysis session (ie,
0·17–1·0 mL/min when averaged over 72 h), as compared
with 65 mL/min in a normally functioning kidney.96
Conventional haemodialysis relies on diffusion for
solute removal, whereas haemodiafiltration also
depends on convective transport. This additional
mechanism enhances the removal of middle-sized
molecules, such as β2-microglobulin and FGF-23, but
has little effect on many protein-bound retention
products.97,98
However, β2-microglobulin clearance per
single haemodiafiltration session is still far from
normal kidney removal rates when averaged over
48–72 h (3·6 mL/min higher than conventional
haemodialysis).99
Despite this poor efficiency, findings
from a randomised controlled trial100
published in 2013,
suggested that haemodiafiltration could improve hard
outcomes versus standard dialysis, but a meta-analysis
of all studies showed improvement only for cardio-
vascular mortality, but not overall mortality.
Additionally, the reliability of potential benefits found
in this meta-analysis was deemed debatable because
the studies included in the analysis had several
methodological limitations.101
Dialyser design changes might further improve removal
of middle molecules.102
However, for each additional
20 mg of β2-microglobulin removed in a single session
www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 11
Review
(less than 10% increase), 1·24 g of albumin were lost (40%
increase).102
Researchers who aim to increase protein-
bound solute removal are exploring how to increase the
release of uraemic retention products from their protein-
bound states,103
for example by including adsorbents, but
length of dialysis using these technologies is a limiting
factor.97
Extension of haemodialysis time, in daily or
extended regimens, either at home or in centre, can
improve weekly solute clearance.97,98
Transplanted kidneys develop hypertrophy and mean
eGFR is 60–70 mL/min per 1·73 m² 1 year after
transplantation.104
However, in a cross-sectional study of
graft recipients 5 years after their transplantation, more
than 70% of patients had measured GFR of less than
60 mL/min per 1·73 m², and thus, had limited removal
capacity.105
Serum β2-microglobulin concentrations of
3·0 mg/L or more were observed in 58% of kidney
transplant recipients at discharge.106
Kidney graft
handling of certain uraemic retention products might be
abnormal. For example, phosphate leakage is frequent
early after transplantation, especially in patients with
severe hyperparathyroidism.105
Over time, kidney grafts
lose function—the half-life of a cadaveric kidney graft is
9 years and 12 years for a living-donor graft.
Conclusion
Patients with chronic kidney disease have complex
pathophysiology for which the underlying mechanisms
intertwine (appendix). Inflammation and disturbed
bone homoeostasis in particular lead to complications
and high and accelerated mortality. Management
(appendix) cannot always be based on high-level
evidence, because of difficulties in the recruitment of
patients with sufficiently homogeneous background of
primary disease, metabolic features, and response to the
uraemic syndrome. Specific therapeutic recom-
mendations are, therefore, based on an amalgam of
high-level and lower-level evidence and uraemia-related
pathophysiological reasoning. Therapeutic approaches
cannot always be extrapolated from the general
population, because beneficial interventions in the
general population often have a different effect in
populations with chronic kidney disease. Treatment of
one aspect of the uraemic syndrome might exacerbate
other deleterious elements. In future studies, a more
holistic therapeutic approach to cope with the high
mortality of this disease could be more useful rather
than the pursuit of one single factor as is usual practice
in randomised controlled trials. Therapeutic approaches
should focus on additional outcomes beyond mortality,
especially standardised quality of life. Consultation with
patients to understand what is important to them might
be useful for the definition of patient-related outcomes.
Contributors
All authors wrote the first draft of one or more sections of the Review,
and all contributed to the editing of subsequent versions.
RV coordinated the initiative, merged the texts, and did general editing.
Declaration of interests
RV has received travel grants and honoraria from Nipro, Bayer, and
Fresenius Medical Care, and has acted as a consultant for Fresenius
Medical Care and Debiotech. DF has acted as a consultant for Fresenius
Kabi. GHH has received study grants from Pharmacosmos and
Fresenius Medical Care. ZAM has received speaker’s honoraria and
research grants from Amgen, Genzyme, Fresenius Medical Care, and
Shire. AO has received speaker’s honoraria and research grants from
Amgen, Genzyme, Fresenius Medical Care, Servier, and Shire. PR has
received honoraria from Baxter-Gambro, Fresenius, and Relypsa. AW has
received speaker’s and consulting honoraria from Amgen, Fresenius
Medical Care, Astellas, Roche, GlaxoSmithKline, Pharmacosmos, and
Teva. GG, MK, FM, CZ, and GML declare no competing interests.
Acknowledgments
The European Renal and Cardiovascular Medicine (EURECA-m)
working group is one of the working groups of the European Renal
Association—European Dialysis and Transplant Association
(ERA-EDTA) and is composed of members whose careers have been
devoted to unravelling the pathophysiology of cardiovascular disease in
chronic kidney disease and to defining accurate therapeutic approaches
to address this problem.
References
1 Almeras C, Argilés A. The general picture of uremia. Semin Dial
2009; 22: 329–33.
2 Meyer TW, Hostetter TH. Uremia. N Engl J Med 2007; 357: 1316–25.
3 Roberts MA, Polkinghorne KR, McDonald SP, Ierino FL.
Secular trends in cardiovascular mortality rates of patients receiving
dialysis compared with the general population. Am J Kidney Dis
2011; 58: 64–72.
4 Chapman JR. What are the key challenges we face in kidney
transplantation today? Transplant Res 2013; 2 (suppl 1): S1.
5 Matsushita K, van der Velde M, Astor BC, et al, for the Chronic
Kidney Disease Prognosis Consortium. Association of estimated
glomerular filtration rate and albuminuria with all-cause and
cardiovascular mortality in general population cohorts:
a collaborative meta-analysis. Lancet 2010; 375: 2073–81.
6 Vanholder R, Massy Z, Argiles A, et al. Chronic kidney disease as
cause of cardiovascular morbidity and mortality.
Nephrol Dial Transplant 2005; 20: 1048–56.
7 Jun M, Lv J, Perkovic V, Jardine MJ. Managing cardiovascular risk in
people with chronic kidney disease: a review of the evidence from
randomized controlled trials. Ther Adv Chronic Dis 2011; 2: 265–78.
8 Ortiz A, Covic A, Fliser D, et al, for the Board of the EURECA-m
Working Group of ERA-EDTA. Epidemiology, contributors to, and
clinical trials of mortality risk in chronic kidney failure. Lancet 2014;
383: 1831–43.
9 de Jager DJ, Grootendorst DC, Jager KJ, et al. Cardiovascular and
noncardiovascular mortality among patients starting dialysis. JAMA
2009; 302: 1782–89.
Search strategy and selection criteria
We searched PubMed, Embase,Webof Science, andthe
Cochrane Library for articles published in Englishupto Dec 1,
2015. Searchterms relatedtothe maintopicsofthis Review
wereused—eg, “metabolic bonedisease”or “inflammation”
and “chronic kidneydisease”, “chronic renal failure”or “chronic
kidneydisease” and “physiopathology”, “treatment”, “therapy”,
“outcomes”, “morbidity”,or “mortality”. Randomised
controlledtrialsor largeobservational studieson hard
outcomeswere retained by preference. Reference listsofthe
resulting articleswere searched forother relevant papers.
Review articles are citedto provide readerswith moredetails
and more referencesthan space permits inthis narrative
review, andwere selected fortheir comprehensiveness and
compactness.
12 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4
Review
10 Morton RL, Schlackow I, Mihaylova B, Staplin ND, Gray A, Cass A.
The impact of social disadvantage in moderate-to-severe chronic
kidney disease: an equity-focused systematic review.
Nephrol Dial Transplant 2016; 31: 46–56.
11 Weiner DE, Tighiouart H, Elsayed EF, et al. The Framingham
predictive instrument in chronic kidney disease. J Am Coll Cardiol
2007; 50: 217–24.
12 Matsushita K, Coresh J, Sang Y, et al. Estimated glomerular filtration
rate and albuminuria for prediction of cardiovascular outcomes: a
collaborative meta-analysis of individual participant data.
Lancet Diabetes Endocrinol 2015; 3: 514–25.
13 Vanholder R, De Smet R, Glorieux G, et al, for the European
Uremic Toxin Work Group (EUTox). Review on uremic toxins:
classification, concentration, and interindividual variability.
Kidney Int 2003; 63: 1934–43.
14 Duranton F, Cohen G, De Smet R, et al, for the European Uremic
Toxin Work Group. Normal and pathologic concentrations of
uremic toxins. J Am Soc Nephrol 2012; 23: 1258–70.
15 Vanholder R, Baurmeister U, Brunet P, Cohen G, Glorieux G,
Jankowski J, for the European Uremic Toxin Work Group. A bench
to bedside view of uremic toxins. J Am Soc Nephrol 2008; 19: 863–70.
16 Tripepi G, Kollerits B, Leonardis D, et al. Competitive interaction
between fibroblast growth factor 23 and asymmetric dimethylarginine
in patients with CKD. J Am Soc Nephrol 2015; 26: 935–44.
17 Inker LA, Tonelli M, Hemmelgarn BR, Levitan EB, Muntner P.
Comparison of concurrent complications of CKD by 2 risk
categorization systems. Am J Kidney Dis 2012; 59: 372–81.
18 Wühl E, Schaefer F. Managing kidney disease with blood-pressure
control. Nat Rev Nephrol 2011; 7: 434–44.
19 Gordon JW, Shaw JA, Kirshenbaum LA. Multiple facets of NF-κB in
the heart: to be or not to NF-κB. Circ Res 2011; 108: 1122–32.
20 Ruiz S, Pergola PE, Zager RA, Vaziri ND. Targeting the transcription
factor Nrf2 to ameliorate oxidative stress and inflammation in
chronic kidney disease. Kidney Int 2013; 83: 1029–41.
21 Kooman JP, Kotanko P, Schols AM, Shiels PG, Stenvinkel P.
Chronic kidney disease and premature ageing. Nat Rev Nephrol
2014; 10: 732–42.
22 Briet M, Boutouyrie P, Laurent S, London GM. Arterial stiffness
and pulse pressure in CKD and ESRD. Kidney Int 2012; 82: 388–400.
23 Minamino T, Komuro I. Vascular cell senescence: contribution to
atherosclerosis. Circ Res 2007; 100: 15–26.
24 Boström KI, Rajamannan NM, Towler DA. The regulation of
valvular and vascular sclerosis by osteogenic morphogens. Circ Res
2011; 109: 564–77.
25 Lim K, Lu TS, Molostvov G, et al. Vascular Klotho deficiency
potentiates the development of human artery calcification and
mediates resistance to fibroblast growth factor 23. Circulation 2012;
125: 2243–55.
26 London GM, Marchais SJ, Guérin AP, de Vernejoul MC.
Ankle-brachial index and bone turnover in patients on dialysis.
J Am Soc Nephrol 2015; 26: 476–83.
27 Nitta K, Okada K, Yanai M, Takahashi S. Aging and chronic kidney
disease. Kidney Blood Press Res 2013; 38: 109–20.
28 Raschenberger J, Kollerits B, Titze S, et al. Association of relative
telomere length with cardiovascular disease in a large chronic kidney
disease cohort: the GCKD study. Atherosclerosis 2015; 242: 529–34.
29 Zawada AM, Rogacev KS, Heine GH. Clinical relevance of
epigenetic dysregulation in chronic kidney disease-associated
cardiovascular disease. Nephrol Dial Transplant 2013; 28: 1663–71.
30 Zawada AM, Rogacev KS, Hummel B, et al. SuperTAG
methylation-specific digital karyotyping reveals uremia-induced
epigenetic dysregulation of atherosclerosis-related genes.
Circ Cardiovasc Genet 2012; 5: 611–20.
31 Kalantar-Zadeh K, Block G, McAllister CJ, Humphreys MH,
Kopple JD. Appetite and inflammation, nutrition, anemia, and
clinical outcome in hemodialysis patients. Am J Clin Nutr 2004;
80: 299–307.
32 Wong J, Piceno YM, Desantis TZ, Pahl M, Andersen GL, Vaziri ND.
Expansion of urease- and uricase-containing, indole- and
p-cresol-forming and contraction of short-chain fatty acid-producing
intestinal microbiota in ESRD. Am J Nephrol 2014; 39: 230–37.
33 Anders HJ, Andersen K, Stecher B. The intestinal microbiota,
a leaky gut, and abnormal immunity in kidney disease. Kidney Int
2013; 83: 1010–16.
34 Locatelli F, Barany P, Covic A, et al. Kidney Disease: Improving
Global Outcomes guidelines on anaemia management in chronic
kidney disease: a European Renal Best Practice position statement.
Nephrol Dial Transplant 2013; 28: 1346–59.
35 Bugnicourt JM, Godefroy O, Chillon JM, Choukroun G, Massy ZA.
Cognitive disorders and dementia in CKD: the neglected
kidney-brain axis. J Am Soc Nephrol 2013; 24: 353–63.
36 Chen YC, Su YC, Lee CC, Huang YS, Hwang SJ. Chronic kidney
disease itself is a causal risk factor for stroke beyond traditional
cardiovascular risk factors: a nationwide cohort study in Taiwan.
PLoS One 2012; 7: e36332.
37 Smith LK, He Y, Park JS, et al. β2-microglobulin is a systemic
pro-aging factor that impairs cognitive function and neurogenesis.
Nat Med 2015; 21: 932–37.
38 Heiwe S, Jacobson SH. Exercise training in adults with CKD:
a systematic review and meta-analysis. Am J Kidney Dis 2014;
64: 383–93.
39 Yacoub R, Habib H, Lahdo A, et al. Association between smoking
and chronic kidney disease: a case control study.
BMC Public Health 2010; 10: 731.
40 Babayev R, Whaley-Connell A, Kshirsagar A, et al. Association of
race and body mass index with ESRD and mortality in CKD stages
3-4: results from the Kidney Early Evaluation Program (KEEP).
Am J Kidney Dis 2013; 61: 404–12.
41 Lu JL, Molnar MZ, Naseer A, Mikkelsen MK, Kalantar-Zadeh K,
Kovesdy CP. Association of age and BMI with kidney function and
mortality: a cohort study. Lancet Diabetes Endocrinol 2015; 3: 704–14.
42 Park J, Ahmadi SF, Streja E, et al. Obesity paradox in end-stage
kidney disease patients. Prog Cardiovasc Dis 2014; 56: 415–25.
43 Fouque D, Aparicio M. Eleven reasons to control the protein intake
of patients with chronic kidney disease. Nat Clin Pract Nephrol
2007; 3: 383–92.
44 Laville M, Fouque D. Nutritional aspects in hemodialysis.
Kidney Int Suppl 2000; 58 (suppl 76): S133–39.
45 Fouque D, Vennegoor M, ter Wee P, et al. EBPG guideline on
nutrition. Nephrol Dial Transplant 2007; 22 (suppl 2): ii45–87.
46 Testa A, Plou A. Clinical determinants of interdialytic weight gain.
J Ren Nutr 2001; 11: 155–60.
47 Moreau-Gaudry X, Jean G, Genet L, et al. A simple protein-energy
wasting score predicts survival in maintenance hemodialysis
patients. J Ren Nutr 2014; 24: 395–400.
48 Saran R, Bragg-Gresham JL, Levin NW, et al. Longer treatment
time and slower ultrafiltration in hemodialysis: associations with
reduced mortality in the DOPPS. Kidney Int 2006; 69: 1222–28.
49 Shinaberger CS, Greenland S, Kopple JD, et al. Is controlling
phosphorus by decreasing dietary protein intake beneficial or
harmful in persons with chronic kidney disease? Am J Clin Nutr
2008; 88: 1511–18.
50 Yusuf S, Diener HC, Sacco RL, et al, for the PRoFESS Study Group.
Telmisartan to prevent recurrent stroke and cardiovascular events.
N Engl J Med 2008; 359: 1225–37.
51 Writing Team for the Diabetes Control and Complications
Trial/Epidemiology of Diabetes Interventions and Complications
Research Group. Sustained effect of intensive treatment of type 1
diabetes mellitus on development and progression of diabetic
nephropathy: the Epidemiology of Diabetes Interventions and
Complications (EDIC) study. JAMA 2003; 290: 2159–67.
52 Haynes R, Lewis D, Emberson J, et al, and the SHARP
Collaborative Group, and the SHARP Collaborative Group.
Effects of lowering LDL cholesterol on progression of kidney
disease. J Am Soc Nephrol 2014; 25: 1825–33.
53 Palmer SC, Navaneethan SD, Craig JC, et al. HMG CoA reductase
inhibitors (statins) for people with chronic kidney disease not
requiring dialysis. Cochrane Database Syst Rev 2014; 5: CD007784.
54 de Brito-Ashurst I, Varagunam M, Raftery MJ, Yaqoob MM.
Bicarbonate supplementation slows progression of CKD and
improves nutritional status. J Am Soc Nephrol 2009; 20: 2075–84.
55 Goicoechea M, de Vinuesa SG, Verdalles U, et al. Effect of
allopurinol in chronic kidney disease progression and
cardiovascular risk. Clin J Am Soc Nephrol 2010; 5: 1388–93.
56 Navarro-González JF, Mora-Fernández C, Muros de Fuentes M,
et al. Effect of pentoxifylline on renal function and urinary albumin
excretion in patients with diabetic kidney disease: the PREDIAN
trial. J Am Soc Nephrol 2015; 26: 220–29.
www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 13
Review
57 Fellström BC, Jardine AG, Schmieder RE, et al, for the AURORA
Study Group. Rosuvastatin and cardiovascular events in patients
undergoing hemodialysis. N Engl J Med 2009; 360: 1395–407.
58 de Zeeuw D, Akizawa T, Audhya P, et al, and the BEACON Trial
Investigators. Bardoxolone methyl in type 2 diabetes and stage 4
chronic kidney disease. N Engl J Med 2013; 369: 2492–503.
59 Verbeke F, Lindley E, Van Bortel L, et al. A European Renal Best
Practice (ERBP) position statement on the Kidney Disease:
Improving Global Outcomes (KDIGO) clinical practice guideline
for the management of blood pressure in non-dialysis-dependent
chronic kidney disease: an endorsement with some caveats for
real-life application. Nephrol Dial Transplant 2014; 29: 490–96.
60 Agarwal R, Peixoto AJ, Santos SF, Zoccali C. Pre- and postdialysis
blood pressures are imprecise estimates of interdialytic ambulatory
blood pressure. Clin J Am Soc Nephrol 2006; 1: 389–98.
61 Bansal N, McCulloch CE, Rahman M, et al, and the CRIC Study
Investigators. Blood pressure and risk of all-cause mortality in
advanced chronic kidney disease and hemodialysis: the chronic
renal insufficiency cohort study. Hypertension 2015; 65: 93–100.
62 The Blood Pressure Lowering Treatment Trialists’ Collaboration.
Blood pressure-lowering treatment based on cardiovascular risk:
a meta-analysis of individual patient data. Lancet 2014;
384: 591–98.
63 The SPRINT Research Group. A randomized trial of intensive
versus standard blood-pressure control. N Engl J Med 2015;
373: 2103–16.
64 Heerspink HJ, Ninomiya T, Zoungas S, et al. Effect of lowering
blood pressure on cardiovascular events and mortality in patients
on dialysis: a systematic review and meta-analysis of randomised
controlled trials. Lancet 2009; 373: 1009–15.
65 Eldehni MT, Odudu A, McIntyre CW. Randomized clinical trial of
dialysate cooling and effects on brain white matter.
J Am Soc Nephrol 2015; 26: 957–65.
66 Levin NW, Kotanko P, Eckardt KU, et al. Blood pressure in chronic
kidney disease stage 5D-report from a Kidney Disease: Improving
Global Outcomes controversies conference. Kidney Int 2010;
77: 273–84.
67 Charra B, Calemard E, Ruffet M, et al. Survival as an index of
adequacy of dialysis. Kidney Int 1992; 41: 1286–91.
68 Moen MF, Zhan M, Hsu VD, et al. Frequency of hypoglycemia and
its significance in chronic kidney disease. Clin J Am Soc Nephrol
2009; 4: 1121–27.
69 Guideline development Group. Clinical practice guideline on
management of patients with diabetes and chronic kidney disease
stage 3b or higher (eGFR <45 mL/min). Nephrol Dial Transplant
2015; 30 (suppl 2): ii1–142.
70 Palmer SC, Mavridis D, Navarese E, et al. Comparative efficacy and
safety of blood pressure-lowering agents in adults with diabetes and
kidney disease: a network meta-analysis. Lancet 2015; 385: 2047–56.
71 Zinman B, Wanner C, Lachin JM, et al. Empagliflozin,
cardiovascular outcomes, and mortality in type 2 diabetes.
N Engl J Med 2015; 373: 2117–28.
72 Tonelli M, Wanner C, for the Kidney Disease: Improving Global
Outcomes Lipid Guideline Development Work Group Members.
Lipid management in chronic kidney disease: synopsis of the
Kidney Disease: Improving Global Outcomes 2013 clinical practice
guideline. Ann Intern Med 2014; 160: 182–89.
73 Baigent C, Landray MJ, Reith C, et al. The effects of lowering LDL
cholesterol with simvastatin plus ezetimibe in patients with chronic
kidney disease (Study of Heart and Renal Protection): a randomised
placebo-controlled trial. Lancet 2011; 377: 2181–92.
74 Massy ZA, de Zeeuw D. LDL cholesterol in CKD—to treat or not to
treat? Kidney Int 2013; 84: 451–56.
75 Winkelmayer WC, Liu J, Kestenbaum B. Comparative effectiveness
of calcium-containing phosphate binders in incident U.S. dialysis
patients. Clin J Am Soc Nephrol 2011; 6: 175–83.
76 Isakova T, Gutiérrez OM, Chang Y, et al. Phosphorus binders and
survival on hemodialysis. J Am Soc Nephrol 2009; 20: 388–96.
77 Zoccali C, Bolignano D, D’Arrigo G, et al, for the European Renal
and Cardiovascular Medicine (EURECA-m) Working Group of the
European Renal Association–European Dialysis Transplantation
Association (ERA-EDTA). Validity of vascular calcification as a
screening tool and as a surrogate end point in clinical research.
Hypertension 2015; 66: 3–9.
78 Tentori F, Blayney MJ, Albert JM, et al. Mortality risk for dialysis
patients with different levels of serum calcium, phosphorus, and
PTH: the Dialysis Outcomes and Practice Patterns Study (DOPPS).
Am J Kidney Dis 2008; 52: 519–30.
79 Chertow GM, Block GA, Correa-Rotter R, et al, and the EVOLVE
Trial Investigators. Effect of cinacalcet on cardiovascular disease in
patients undergoing dialysis. N Engl J Med 2012; 367: 2482–94.
80 Palmer SC, Nistor I, Craig JC, et al. Cinacalcet in patients with
chronic kidney disease: a cumulative meta-analysis of randomized
controlled trials. PLoS Med 2013; 10: e1001436.
81 Liabeuf S, Neirynck N, Drüeke TB, Vanholder R, Massy ZA.
Clinical studies and chronic kidney disease: what did we learn
recently? Semin Nephrol 2014; 34: 164–79.
82 Fouque D, Kalantar-Zadeh K, Kopple J, et al. A proposed
nomenclature and diagnostic criteria for protein-energy wasting in
acute and chronic kidney disease. Kidney Int 2008; 73: 391–98.
83 Cano NJ, Fouque D, Roth H, et al, for the French Study Group for
Nutrition in Dialysis. Intradialytic parenteral nutrition does not
improve survival in malnourished hemodialysis patients: a 2-year
multicenter, prospective, randomized study. J Am Soc Nephrol 2007;
18: 2583–91.
84 Sirich TL, Plummer NS, Gardner CD, Hostetter TH, Meyer TW.
Effect of increasing dietary fiber on plasma levels of colon-derived
solutes in hemodialysis patients. Clin J Am Soc Nephrol 2014;
9: 1603–10.
85 Bliss DZ, Stein TP, Schleifer CR, Settle RG. Supplementation
with gum arabic fiber increases fecal nitrogen excretion and
lowers serum urea nitrogen concentration in chronic renal failure
patients consuming a low-protein diet. Am J Clin Nutr 1996;
63: 392–98.
86 Wang IK, Wu YY, Yang YF, et al. The effect of probiotics on serum
levels of cytokine and endotoxin in peritoneal dialysis patients:
a randomised, double-blind, placebo-controlled trial. Benef Microbes
2015; 6: 423–30.
87 Guida B, Germanò R, Trio R, et al. Effect of short-term synbiotic
treatment on plasma p-cresol levels in patients with chronic renal
failure: a randomized clinical trial. Nutr Metab Cardiovasc Dis 2014;
24: 1043–49.
88 Akizawa T, Asano Y, Morita S, et al. Effect of a carbonaceous oral
adsorbent on the progression of CKD: a multicenter, randomized,
controlled trial. Am J Kidney Dis 2009; 54: 459–67.
89 Ueda H, Shibahara N, Takagi S, Inoue T, Katsuoka Y. AST-120, an
oral adsorbent, delays the initiation of dialysis in patients with
chronic kidney diseases. Ther Apher Dial 2007; 11: 189–95.
90 Schulman G, Berl T, Beck GJ, et al. Randomized placebo-controlled
EPPIC Trials of AST-120 in CKD. J Am Soc Nephrol 2015; 26: 1732–46.
91 Fell LH, Zawada AM, Rogacev KS, Seiler S, Fliser D, Heine GH.
Distinct immunologic effects of different intravenous iron
preparations on monocytes. Nephrol Dial Transplant 2014; 29: 809–22.
92 Bailie GR, Larkina M, Goodkin DA, et al. Data from the Dialysis
Outcomes and Practice Patterns Study validate an association
between high intravenous iron doses and mortality. Kidney Int 2015;
87: 162–68.
93 Macdougall IC, Obrador GT. How important is transfusion
avoidance in 2013? Nephrol Dial Transplant 2013; 28: 1092–99.
94 Zhang X, Xiang C, Zhou YH, Jiang A, Qin YY, He J. Effect of statins
on cardiovascular events in patients with mild to moderate chronic
kidney disease: a systematic review and meta-analysis of randomized
clinical trials. BMC Cardiovasc Disord 2014; 14: 19.
95 Seliger SL, Zhang AD, Weir MR, et al. Erythropoiesis-stimulating
agents increase the risk of acute stroke in patients with chronic
kidney disease. Kidney Int 2011; 80: 288–94.
96 Floege J, Bartsch A, Schulze M, Shaldon S, Koch KM, Smeby LC.
Clearance and synthesis rates of beta 2-microglobulin in patients
undergoing hemodialysis and in normal subjects. J Lab Clin Med
1991; 118: 153–65.
97 Cornelis T, van der Sande FM, Eloot S, et al. Acute hemodynamic
response and uremic toxin removal in conventional and extended
hemodialysis and hemodiafiltration: a randomized crossover study.
Am J Kidney Dis 2014; 64: 247–56.
98 Cornelis T, Eloot S, Vanholder R, et al. Protein-bound uraemic
toxins, dicarbonyl stress and advanced glycation end products in
conventional and extended haemodialysis and haemodiafiltration.
Nephrol Dial Transplant 2015; 30: 1395–402.
14 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4
Review
99 Susantitaphong P, Siribamrungwong M, Jaber BL.
Convective therapies versus low-flux hemodialysis for chronic
kidney failure: a meta-analysis of randomized controlled trials.
Nephrol Dial Transplant 2013; 28: 2859–74.
100 Maduell F, Moreso F, Pons M, et al, for the ESHOL Study Group.
High-efficiency postdilution online hemodiafiltration reduces
all-cause mortality in hemodialysis patients. J Am Soc Nephrol 2013;
24: 487–97.
101 Nistor I, Palmer SC, Craig JC, et al. Convective versus diffusive dialysis
therapies for chronic kidney failure: an updated systematic review of
randomized controlled trials. Am J Kidney Dis 2014; 63: 954–67.
102 Maduell F, Arias-Guillen M, Fontseré N, et al. Elimination of large
uremic toxins by a dialyzer specifically designed for high-volume
convective therapies. Blood Purif 2014; 37: 125–30.
103 Böhringer F, Jankowski V, Gajjala PR, Zidek W, Jankowski J.
Release of uremic retention solutes from protein binding by
hypertonic predilution hemodiafiltration. ASAIO J 2015; 61: 55–60.
104 Budde K, Becker T, Arns W, et al. Everolimus-based,
calcineurin-inhibitor-free regimen in recipients of de-novo kidney
transplants: an open-label, randomised, controlled trial. Lancet 2011;
377: 837–47.
105 Perrin P, Caillard S, Javier RM, et al. Persistent hyperparathyroidism
is a major risk factor for fractures in the five years after kidney
transplantation. Am J Transplant 2013; 13: 2653–63.
106 Astor BC, Muth B, Kaufman DB, Pirsch JD, Michael Hofmann R,
Djamali A. Serum β2-microglobulin at discharge predicts mortality
and graft loss following kidney transplantation. Kidney Int 2013;
84: 810–17.

More Related Content

What's hot

Post transplant erythrocytosis
Post transplant erythrocytosisPost transplant erythrocytosis
Post transplant erythrocytosis
Patricia Khashayar
 
Features of Inflammatory Markers in Patients With Coronary Heart Disease
Features of Inflammatory Markers in Patients With Coronary Heart DiseaseFeatures of Inflammatory Markers in Patients With Coronary Heart Disease
Features of Inflammatory Markers in Patients With Coronary Heart Disease
Healthcare and Medical Sciences
 
SaudiJKidneyDisTranspl2661307-7143059_015903
SaudiJKidneyDisTranspl2661307-7143059_015903SaudiJKidneyDisTranspl2661307-7143059_015903
SaudiJKidneyDisTranspl2661307-7143059_015903
kifayat ullah
 

What's hot (20)

WCIM poster 2016, Bali
WCIM poster 2016, BaliWCIM poster 2016, Bali
WCIM poster 2016, Bali
 
ERC, diagnóstico y manejo.pdf
ERC, diagnóstico y manejo.pdfERC, diagnóstico y manejo.pdf
ERC, diagnóstico y manejo.pdf
 
Post transplant erythrocytosis
Post transplant erythrocytosisPost transplant erythrocytosis
Post transplant erythrocytosis
 
Is serum urate causally associated with incident cardiovascular disease?
Is serum urate causally associated with incident cardiovascular disease?Is serum urate causally associated with incident cardiovascular disease?
Is serum urate causally associated with incident cardiovascular disease?
 
Features of Inflammatory Markers in Patients With Coronary Heart Disease
Features of Inflammatory Markers in Patients With Coronary Heart DiseaseFeatures of Inflammatory Markers in Patients With Coronary Heart Disease
Features of Inflammatory Markers in Patients With Coronary Heart Disease
 
Acute kidney injury
Acute kidney injuryAcute kidney injury
Acute kidney injury
 
SaudiJKidneyDisTranspl2661307-7143059_015903
SaudiJKidneyDisTranspl2661307-7143059_015903SaudiJKidneyDisTranspl2661307-7143059_015903
SaudiJKidneyDisTranspl2661307-7143059_015903
 
Nia (1)
Nia (1)Nia (1)
Nia (1)
 
Intra-operative determinants of postoperative AKI
Intra-operative determinants of postoperative AKIIntra-operative determinants of postoperative AKI
Intra-operative determinants of postoperative AKI
 
Evaluation of plasma Creatine Kinase activity and Inorganic phosphate among S...
Evaluation of plasma Creatine Kinase activity and Inorganic phosphate among S...Evaluation of plasma Creatine Kinase activity and Inorganic phosphate among S...
Evaluation of plasma Creatine Kinase activity and Inorganic phosphate among S...
 
Evaluation of Plasma Creatine Kinase Activity and Inorganic Phosphate among S...
Evaluation of Plasma Creatine Kinase Activity and Inorganic Phosphate among S...Evaluation of Plasma Creatine Kinase Activity and Inorganic Phosphate among S...
Evaluation of Plasma Creatine Kinase Activity and Inorganic Phosphate among S...
 
Evaluation of Oxidative status of gout patients in a Cameroonian urban hospit...
Evaluation of Oxidative status of gout patients in a Cameroonian urban hospit...Evaluation of Oxidative status of gout patients in a Cameroonian urban hospit...
Evaluation of Oxidative status of gout patients in a Cameroonian urban hospit...
 
Nejm Effects of Aspirin for Primary Prevention in Persons with Diabetes Mellitus
Nejm Effects of Aspirin for Primary Prevention in Persons with Diabetes MellitusNejm Effects of Aspirin for Primary Prevention in Persons with Diabetes Mellitus
Nejm Effects of Aspirin for Primary Prevention in Persons with Diabetes Mellitus
 
Emerging MRA-Based Treatments for End-Stage Renal Disease (ESRD) Patients on ...
Emerging MRA-Based Treatments for End-Stage Renal Disease (ESRD) Patients on ...Emerging MRA-Based Treatments for End-Stage Renal Disease (ESRD) Patients on ...
Emerging MRA-Based Treatments for End-Stage Renal Disease (ESRD) Patients on ...
 
Yin, l., . (2013). the surgical treatment for portal hypertension
Yin, l., . (2013). the surgical treatment for portal hypertensionYin, l., . (2013). the surgical treatment for portal hypertension
Yin, l., . (2013). the surgical treatment for portal hypertension
 
Nephrology & Therapeutics
Nephrology & TherapeuticsNephrology & Therapeutics
Nephrology & Therapeutics
 
International Journal of Nephrology & Therapeutics
International Journal of Nephrology & TherapeuticsInternational Journal of Nephrology & Therapeutics
International Journal of Nephrology & Therapeutics
 
Hepatorenal Syndrome
Hepatorenal Syndrome Hepatorenal Syndrome
Hepatorenal Syndrome
 
Evaluation Of Aetiology Of Upper Gastro Intestinal Bleeding &Management In Th...
Evaluation Of Aetiology Of Upper Gastro Intestinal Bleeding &Management In Th...Evaluation Of Aetiology Of Upper Gastro Intestinal Bleeding &Management In Th...
Evaluation Of Aetiology Of Upper Gastro Intestinal Bleeding &Management In Th...
 
Role of the Biochemistry Labs in Promoting the Health Care Services for the I...
Role of the Biochemistry Labs in Promoting the Health Care Services for the I...Role of the Biochemistry Labs in Promoting the Health Care Services for the I...
Role of the Biochemistry Labs in Promoting the Health Care Services for the I...
 

Similar to Clinical_management_erc

The Spectrum of Histopathological Patterns in Diabetic Kidney Disease in East...
The Spectrum of Histopathological Patterns in Diabetic Kidney Disease in East...The Spectrum of Histopathological Patterns in Diabetic Kidney Disease in East...
The Spectrum of Histopathological Patterns in Diabetic Kidney Disease in East...
ANALYTICAL AND QUANTITATIVE CYTOPATHOLOGY AND HISTOPATHOLOGY
 
Hepatic Encephalopathy - Medicina Interna II
Hepatic Encephalopathy - Medicina Interna IIHepatic Encephalopathy - Medicina Interna II
Hepatic Encephalopathy - Medicina Interna II
Matias Fernandez Viña
 
Acute+kidney+injury+ +final+version+(08+march+2011)+
Acute+kidney+injury+ +final+version+(08+march+2011)+Acute+kidney+injury+ +final+version+(08+march+2011)+
Acute+kidney+injury+ +final+version+(08+march+2011)+
centurionyegros
 
Acg guideline acute_pancreatitis_september_2013
Acg guideline acute_pancreatitis_september_2013Acg guideline acute_pancreatitis_september_2013
Acg guideline acute_pancreatitis_september_2013
iberzamz
 
Liver Fibrosis: Difficulties in Diagnostic and Treatment: A Review-Crimson Pu...
Liver Fibrosis: Difficulties in Diagnostic and Treatment: A Review-Crimson Pu...Liver Fibrosis: Difficulties in Diagnostic and Treatment: A Review-Crimson Pu...
Liver Fibrosis: Difficulties in Diagnostic and Treatment: A Review-Crimson Pu...
CrimsonGastroenterology
 
Liver Cirrhosis and Neurological Disorder Associated with Liver Disease
Liver Cirrhosis and Neurological Disorder Associated with Liver DiseaseLiver Cirrhosis and Neurological Disorder Associated with Liver Disease
Liver Cirrhosis and Neurological Disorder Associated with Liver Disease
ijtsrd
 
Acg guideline acute_pancreatitis_september_2013 - copia
Acg guideline acute_pancreatitis_september_2013 - copiaAcg guideline acute_pancreatitis_september_2013 - copia
Acg guideline acute_pancreatitis_september_2013 - copia
hgvilla
 

Similar to Clinical_management_erc (20)

The Spectrum of Histopathological Patterns in Diabetic Kidney Disease in East...
The Spectrum of Histopathological Patterns in Diabetic Kidney Disease in East...The Spectrum of Histopathological Patterns in Diabetic Kidney Disease in East...
The Spectrum of Histopathological Patterns in Diabetic Kidney Disease in East...
 
480309
480309480309
480309
 
Endocrine Hypertension.pdf
Endocrine Hypertension.pdfEndocrine Hypertension.pdf
Endocrine Hypertension.pdf
 
pancreatitis aguda .pdf
pancreatitis aguda .pdfpancreatitis aguda .pdf
pancreatitis aguda .pdf
 
DIRI with commercial available agents ,,,.pdf
DIRI with commercial available agents ,,,.pdfDIRI with commercial available agents ,,,.pdf
DIRI with commercial available agents ,,,.pdf
 
New Onset diabetes after kidney transplantation.
New Onset diabetes after kidney transplantation.New Onset diabetes after kidney transplantation.
New Onset diabetes after kidney transplantation.
 
F0562030032
F0562030032F0562030032
F0562030032
 
Hepatic Encephalopathy - Medicina Interna II
Hepatic Encephalopathy - Medicina Interna IIHepatic Encephalopathy - Medicina Interna II
Hepatic Encephalopathy - Medicina Interna II
 
Acute+kidney+injury+ +final+version+(08+march+2011)+
Acute+kidney+injury+ +final+version+(08+march+2011)+Acute+kidney+injury+ +final+version+(08+march+2011)+
Acute+kidney+injury+ +final+version+(08+march+2011)+
 
Curcumin
CurcuminCurcumin
Curcumin
 
Acg guideline acute_pancreatitis_september_2013
Acg guideline acute_pancreatitis_september_2013Acg guideline acute_pancreatitis_september_2013
Acg guideline acute_pancreatitis_september_2013
 
90124843 study-1
90124843 study-190124843 study-1
90124843 study-1
 
Liver Fibrosis: Difficulties in Diagnostic and Treatment: A Review-Crimson Pu...
Liver Fibrosis: Difficulties in Diagnostic and Treatment: A Review-Crimson Pu...Liver Fibrosis: Difficulties in Diagnostic and Treatment: A Review-Crimson Pu...
Liver Fibrosis: Difficulties in Diagnostic and Treatment: A Review-Crimson Pu...
 
Critical care nephrology
Critical care nephrologyCritical care nephrology
Critical care nephrology
 
The Indian Consensus Document on Cardiac Biomarker
The Indian Consensus Document on Cardiac BiomarkerThe Indian Consensus Document on Cardiac Biomarker
The Indian Consensus Document on Cardiac Biomarker
 
Metabolic abnormalities observed in osteoarthritis of knee: A single center e...
Metabolic abnormalities observed in osteoarthritis of knee: A single center e...Metabolic abnormalities observed in osteoarthritis of knee: A single center e...
Metabolic abnormalities observed in osteoarthritis of knee: A single center e...
 
Liver Cirrhosis and Neurological Disorder Associated with Liver Disease
Liver Cirrhosis and Neurological Disorder Associated with Liver DiseaseLiver Cirrhosis and Neurological Disorder Associated with Liver Disease
Liver Cirrhosis and Neurological Disorder Associated with Liver Disease
 
Future Considerations of Biological Disparities in Drug Development for NAFLD...
Future Considerations of Biological Disparities in Drug Development for NAFLD...Future Considerations of Biological Disparities in Drug Development for NAFLD...
Future Considerations of Biological Disparities in Drug Development for NAFLD...
 
Acg guideline acute_pancreatitis_september_2013
Acg guideline acute_pancreatitis_september_2013Acg guideline acute_pancreatitis_september_2013
Acg guideline acute_pancreatitis_september_2013
 
Acg guideline acute_pancreatitis_september_2013 - copia
Acg guideline acute_pancreatitis_september_2013 - copiaAcg guideline acute_pancreatitis_september_2013 - copia
Acg guideline acute_pancreatitis_september_2013 - copia
 

Recently uploaded

Cell structure slideshare.pptx Unlocking the Secrets of Cells: Structure, Fun...
Cell structure slideshare.pptx Unlocking the Secrets of Cells: Structure, Fun...Cell structure slideshare.pptx Unlocking the Secrets of Cells: Structure, Fun...
Cell structure slideshare.pptx Unlocking the Secrets of Cells: Structure, Fun...
ananyagirishbabu1
 
BOWEL ELIMINATION BY ANUSHRI SRIVASTAVA.pptx
BOWEL ELIMINATION BY ANUSHRI SRIVASTAVA.pptxBOWEL ELIMINATION BY ANUSHRI SRIVASTAVA.pptx
BOWEL ELIMINATION BY ANUSHRI SRIVASTAVA.pptx
AnushriSrivastav
 
ASSISTING WITH THE USE OF BED PAN BY ANUSHRI SRIVASTAVA.pptx
ASSISTING WITH THE USE OF BED PAN BY ANUSHRI SRIVASTAVA.pptxASSISTING WITH THE USE OF BED PAN BY ANUSHRI SRIVASTAVA.pptx
ASSISTING WITH THE USE OF BED PAN BY ANUSHRI SRIVASTAVA.pptx
AnushriSrivastav
 
Medical Technology Tackles New Health Care Demand - Research Report - March 2...
Medical Technology Tackles New Health Care Demand - Research Report - March 2...Medical Technology Tackles New Health Care Demand - Research Report - March 2...
Medical Technology Tackles New Health Care Demand - Research Report - March 2...
pchutichetpong
 
CHAPTER 1 SEMESTER V - ROLE OF PEADIATRIC NURSE.pdf
CHAPTER 1 SEMESTER V - ROLE OF PEADIATRIC NURSE.pdfCHAPTER 1 SEMESTER V - ROLE OF PEADIATRIC NURSE.pdf
CHAPTER 1 SEMESTER V - ROLE OF PEADIATRIC NURSE.pdf
Sachin Sharma
 
Demystifying-Gene-Editing-The-Promise-and-Peril-of-CRISPR.pdf
Demystifying-Gene-Editing-The-Promise-and-Peril-of-CRISPR.pdfDemystifying-Gene-Editing-The-Promise-and-Peril-of-CRISPR.pdf
Demystifying-Gene-Editing-The-Promise-and-Peril-of-CRISPR.pdf
SasikiranMarri
 

Recently uploaded (20)

Enhancing-Patient-Centric-Clinical-Trials.pdf
Enhancing-Patient-Centric-Clinical-Trials.pdfEnhancing-Patient-Centric-Clinical-Trials.pdf
Enhancing-Patient-Centric-Clinical-Trials.pdf
 
Myopia Management & Control Strategies.pptx
Myopia Management & Control Strategies.pptxMyopia Management & Control Strategies.pptx
Myopia Management & Control Strategies.pptx
 
Navigating Healthcare with Telemedicine
Navigating Healthcare with  TelemedicineNavigating Healthcare with  Telemedicine
Navigating Healthcare with Telemedicine
 
Jesse Jhaj: Building Relationships with Patients as a Doctor or Healthcare Wo...
Jesse Jhaj: Building Relationships with Patients as a Doctor or Healthcare Wo...Jesse Jhaj: Building Relationships with Patients as a Doctor or Healthcare Wo...
Jesse Jhaj: Building Relationships with Patients as a Doctor or Healthcare Wo...
 
Cell structure slideshare.pptx Unlocking the Secrets of Cells: Structure, Fun...
Cell structure slideshare.pptx Unlocking the Secrets of Cells: Structure, Fun...Cell structure slideshare.pptx Unlocking the Secrets of Cells: Structure, Fun...
Cell structure slideshare.pptx Unlocking the Secrets of Cells: Structure, Fun...
 
BOWEL ELIMINATION BY ANUSHRI SRIVASTAVA.pptx
BOWEL ELIMINATION BY ANUSHRI SRIVASTAVA.pptxBOWEL ELIMINATION BY ANUSHRI SRIVASTAVA.pptx
BOWEL ELIMINATION BY ANUSHRI SRIVASTAVA.pptx
 
Global launch of the Healthy Ageing and Prevention Index 2nd wave – alongside...
Global launch of the Healthy Ageing and Prevention Index 2nd wave – alongside...Global launch of the Healthy Ageing and Prevention Index 2nd wave – alongside...
Global launch of the Healthy Ageing and Prevention Index 2nd wave – alongside...
 
Unlocking the Benefits of Cognitive Behavioural Therapy (CBT) with Renewed Edge
Unlocking the Benefits of Cognitive Behavioural Therapy (CBT) with Renewed EdgeUnlocking the Benefits of Cognitive Behavioural Therapy (CBT) with Renewed Edge
Unlocking the Benefits of Cognitive Behavioural Therapy (CBT) with Renewed Edge
 
ASSISTING WITH THE USE OF BED PAN BY ANUSHRI SRIVASTAVA.pptx
ASSISTING WITH THE USE OF BED PAN BY ANUSHRI SRIVASTAVA.pptxASSISTING WITH THE USE OF BED PAN BY ANUSHRI SRIVASTAVA.pptx
ASSISTING WITH THE USE OF BED PAN BY ANUSHRI SRIVASTAVA.pptx
 
Enhance Your Wellbeing with Natural Women's Health Supplements
Enhance Your Wellbeing with Natural Women's Health SupplementsEnhance Your Wellbeing with Natural Women's Health Supplements
Enhance Your Wellbeing with Natural Women's Health Supplements
 
Artificial Intelligence to Optimize Cardiovascular Therapy
Artificial Intelligence to Optimize Cardiovascular TherapyArtificial Intelligence to Optimize Cardiovascular Therapy
Artificial Intelligence to Optimize Cardiovascular Therapy
 
Medical Technology Tackles New Health Care Demand - Research Report - March 2...
Medical Technology Tackles New Health Care Demand - Research Report - March 2...Medical Technology Tackles New Health Care Demand - Research Report - March 2...
Medical Technology Tackles New Health Care Demand - Research Report - March 2...
 
PT MANAGEMENT OF URINARY INCONTINENCE.pptx
PT MANAGEMENT OF URINARY INCONTINENCE.pptxPT MANAGEMENT OF URINARY INCONTINENCE.pptx
PT MANAGEMENT OF URINARY INCONTINENCE.pptx
 
VVIP Dehradun Girls 9719300533 Heat-bake { Dehradun } Genteel ℂall Serviℂe By...
VVIP Dehradun Girls 9719300533 Heat-bake { Dehradun } Genteel ℂall Serviℂe By...VVIP Dehradun Girls 9719300533 Heat-bake { Dehradun } Genteel ℂall Serviℂe By...
VVIP Dehradun Girls 9719300533 Heat-bake { Dehradun } Genteel ℂall Serviℂe By...
 
QA Paediatric dentistry department, Hospital Melaka 2020
QA Paediatric dentistry department, Hospital Melaka 2020QA Paediatric dentistry department, Hospital Melaka 2020
QA Paediatric dentistry department, Hospital Melaka 2020
 
A Community health , health for prisoners
A Community health  , health for prisonersA Community health  , health for prisoners
A Community health , health for prisoners
 
Dehradun ❤CALL Girls 8901183002 ❤ℂall Girls IN Dehradun ESCORT SERVICE❤
Dehradun ❤CALL Girls  8901183002 ❤ℂall  Girls IN Dehradun ESCORT SERVICE❤Dehradun ❤CALL Girls  8901183002 ❤ℂall  Girls IN Dehradun ESCORT SERVICE❤
Dehradun ❤CALL Girls 8901183002 ❤ℂall Girls IN Dehradun ESCORT SERVICE❤
 
CHAPTER 1 SEMESTER V - ROLE OF PEADIATRIC NURSE.pdf
CHAPTER 1 SEMESTER V - ROLE OF PEADIATRIC NURSE.pdfCHAPTER 1 SEMESTER V - ROLE OF PEADIATRIC NURSE.pdf
CHAPTER 1 SEMESTER V - ROLE OF PEADIATRIC NURSE.pdf
 
Demystifying-Gene-Editing-The-Promise-and-Peril-of-CRISPR.pdf
Demystifying-Gene-Editing-The-Promise-and-Peril-of-CRISPR.pdfDemystifying-Gene-Editing-The-Promise-and-Peril-of-CRISPR.pdf
Demystifying-Gene-Editing-The-Promise-and-Peril-of-CRISPR.pdf
 
Overcome Your Phobias with Hypnotherapy.pptx
Overcome Your Phobias with Hypnotherapy.pptxOvercome Your Phobias with Hypnotherapy.pptx
Overcome Your Phobias with Hypnotherapy.pptx
 

Clinical_management_erc

  • 1. www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 1 Review Lancet Diabetes Endocrinol 2016 Published Online March 2, 2016 http://dx.doi.org/10.1016/ S2213-8587(16)00033-4 Nephrology Section, Department of Internal Medicine, Ghent University Hospital, Ghent, Belgium (Prof RVanholder MD, Prof G Glorieux PhD); Department of Nephrology- Nutrition-Dialysis, Centre Hospitalier Lyon Sud, Carmen-CENS, Université Claude Bernard Lyon 1, Lyon, France; F-CRIN-INI-CRCT, France (Prof D Fouque MD); Department of Internal Medicine IV, Saarland University Medical Centre, Homburg, Germany (Prof G H Heine MD); Department of Medicine, Division of Nephrology, Koc University School of Medicine, Istanbul,Turkey (Prof M Kanbay MD); Nephrology, Dialysis and Transplantation Unit, and CNR-IFC Clinical Epidemiology and Pathophysiology of Renal Diseases and Hypertension, Ospedali Riuniti, Reggio Calabria, Italy (Prof F Mallamaci MD, Prof C Zoccali MD); Division of Nephrology, Ambroise Paré University Hospital (APHP), University of Paris Ouest, Versailles-Saint-Quentin-en- Yvelines (UVSQ), Boulogne-Billancourt, Paris, France (Prof Z A Massy MD); INSERM U1018, Research Centre in Epidemiology and Population Health (CESP), UVSQ,Villejuif, France (Prof Z A Massy); Division of Nephrology, IIS-Fundacion Jimenez Diaz, Madrid, Spain (Prof A Ortiz MD); INSERM Centre d’Investigations Cliniques (CIC)-1433, and INSERM U1116, Nancy, France (Prof P Rossignol MD); Institut Lorrain du Cœur et des Vaisseaux, CHU Nancy, Vandoeuvre lès Nancy, France (Prof P Rossignol); Université de Clinical management of the uraemic syndrome in chronic kidney disease RaymondVanholder, Denis Fouque, Griet Glorieux, Gunnar H Heine, Mehmet Kanbay, Francesca Mallamaci, Ziad A Massy, Alberto Ortiz, Patrick Rossignol, AndrzejWiecek, Carmine Zoccali, Gérard Michel London, for the European Renal Association European Dialysis and Transplant Association (ERA-EDTA) European Renal and Cardiovascular Medicine (EURECA-m) working group The clinical picture of the uraemic syndrome is a complex amalgam of accelerated ageing and organ dysfunction, which progress in parallel to chronic kidney disease. The uraemic syndrome is associated with cardiovascular disease, metabolic bone disease, inflammation, protein energy wasting, intestinal dysbiosis, anaemia, and neurological and endocrine dysfunction. In this Review, we summarise specific, modern management options for the uraemic syndrome in chronic kidney disease. Although large randomised controlled trials are scarce, based on data from randomised controlled trials and observational studies, as well as pathophysiological reasoning, a therapeutic algorithm can be developed for this complex and multifactorial condition, with interventions targeting several modifiable factors simultaneously. Introduction Kidney failure is associated with deterioration of body functions. The clinical picture as a whole—the uraemic syndrome—is named after urea, the most abundant metabolite retained in kidney failure and the first uraemic retention product identified. The uraemic syndrome can be caused by chronic kidney disease or acute kidney injury, and affects almost every organ system (panel 1).1 The syndrome results from the biological effects of metabolites that are not excreted or metabolised by the kidneys and are retained within the body.2 Such metabolites are named uraemic retention products, or uraemic toxins if they exert biological or toxic effects. The deterioration of renal endocrine function (production of erythropoietin, active vitamin D, or renin), the deregulation of kidney electrolyte homoeostasis, and functional alterations resulting from chronic kidney disease and its causes (eg, diabetes, autoimmune disorders) also contribute to the syndrome. The clinical picture worsens with kidney failure, with coma and death (end-stage kidney disease; table 1) the ultimate result if the patient is left untreated. However, since the 1940s, renal replacement therapies (dialysis or transplantation) have extended the life expectancy of patients with this potentially fatal condition. Although dialysis and transplantation extend the life expectancy of patients with uraemia, mortality remains substantially higher than in age-matched populations with normal kidney function;3,4 general and cardiovascular mortality tend to rise even before patients need dialysis.5,6 In this Review, we discuss several therapeutic options to treat the consequences of the uraemic syndrome in chronic kidney disease, based on the pathophysiology of the uraemic syndrome and taking into account newly detected, pathological pathways. Although we have followed the principles of evidence- based medicine as much as possible in this Review, much of the data cited are from observation studies. Randomised controlled trials in kidney disease are scarce,7 and many studies have had negative results.8 This fact is largely attributable to the complex and multifactorial nature of the disease, which makes it difficult to recruit large patient groups with uniform pathophysiological backgrounds. Additionally, because of multi-layered pathophysiology, the effect of therapeutic options that correct one aspect of disease (eg, hypercholesterolaemia) can be masked by the effect of other factors (eg, hypertension, fluid overload) on outcome measures. Traditional and non-traditional risk factors Cardiovascular and non-cardiovascular mortality contri- bute equally to the high mortality seen in people with chronic kidney disease.9 Socioeconomic and geographical factors, including access to therapy, explain the variable mortality in chronic kidney disease and end-stage kidney disease populations.10 Cardio- vascular disease in patients with chronic kidney disease is characterised by immunity-driven inflammatory changes that cause vessel wall stiffening, arteriopathy, and cardiomyopathy leading to heart failure, arrhythmia, and cardiac arrest. Risk factors span from traditional (Framingham) factors to an expanding list of non-traditional risk factors (panel 2).8 Among non- cardiovascular causes of death, infection, cancer, cachexia, suicide, and refusal of treatment account for the largest share of fatalities.9 The gap between the predictive value of traditional risk factors and real cardiovascular mortality in chronic kidney disease11 is to a large extent shown by indicators of kidney dysfunction, such as estimated glomerular filtration rate (eGFR) and albuminuria.12 Hence, factors related to a decline in kidney function, such as subclinical volume expansion and uraemic solute retention, might play a part in this process. Many factors that are affected by the uraemic status have been associated with causes of cardiovascular damage, such as inflammation, oxidative stress, macrophage infiltration, endothelial dysfunction, thrombogenesis, arterial calcification, or osteodystrophy.
  • 2. 2 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 Review Lorraine, Nancy, France (Prof P Rossignol); Association Lorraine pour leTraitement de l’Insuffisance Rénale, Vandoeuvre lès Nancy, France (Prof P Rossignol); Department of Nephrology,Transplantation and Internal Medicine, Medical University of Silesia, Katowice, Poland (Prof AWiecek MD); and INSERM U970, Hôpital Européen Georges Pompidou, Paris (Prof G M London MD) Correspondence to: Prof RaymondVanholder, Nephrology Section, University Hospital, De Pintelaan 185, B9000 Ghent, Belgium raymond.vanholder@ugent.be See Online for appendix Uraemic retention products Information about uraemic retention products has increased continuously in the past few decades. At least 150 uraemic retention products have been described so far,13,14 and with developments in metabolomics and proteomics in the past decade, each new study has the potential to add dozens of new substances to this list. The idea that removal of one single solute would be sufficient to solve the problem of uraemic toxicity has long since been abandoned. By contrast, the notion that all these molecules interact is developing.15,16 In the past 30 years, based on the physicochemical characteristics that affect their elimination during dialysis (the main removal strategy until now), uraemic retention products have been divided into three major categories: small, water-soluble compounds; protein- bound compounds; and larger, middle molecules.13 The characteristics of these classes and some of the main compounds in the groups are summarised in the appendix. Pathophysiology Scope of Review Since the pathophysiology of the uraemic syndrome affects the function of almost every organ (panel 1), this Review is restricted to elements for which relevant information is available and that imply specific therapeutic approaches. Definition of which pathophysiological events are directly caused by uraemic retention products and which, as a whole or in part, are caused by other aspects of kidney dysfunction is not always easy. We will summarise which elements are linked to uraemic retention and which ones to kidney dysfunction. Since the links are sometimes very complex, we can only focus on what we deem the most important aspects. Progression of kidney failure Progression of chronic kidney disease varies depending on the underlying cause, disease-specific pathology, and predisposing risk factors (figure 1).17 Independent of the initial cause, any loss of functional kidney parenchyma leads to compensatory hyperfiltration and intraglomerular hypertension of the remaining nephrons, which causes fibrosis and progressive decline in kidney function. Furthermore, uraemia-specific and non-specific nephro- toxins, inflammation, tissue ischaemia, and pro-coagulant mechanisms damage glomeruli and tubules. Proteinuria increases the risk of chronic kidney disease progression by causing tubular injury that leads to inflammatory macrophage infiltration and tubulointerstitial fibrosis. Angiotensin II release increases intraglomerular pressure and oxidative stress, which alters podocyte function and promotes synthesis of chemokines and cytokines.18 Several uraemic retention products have been linked to progression of kidney disease such as indoxyl sulfate, p-cresyl sulfate, asymmetrical dimethylarginine (ADMA), and several cytokines. Factors not linked to uraemic toxins are exacerbations of primary kidney disease, acute kidney injury, proteinuria, hyperglycaemia, external nephrotoxic agents (radiocontrast, non-steroidals), compensatory hyperfiltration, and ischaemia. Hypertension is a partly retention-independent factor—ie, it can be caused by uraemic toxins or by other factors (figure 1). Inflammation, metabolic bone disease, and cardiovascular disease Non-traditional risk factors, such as low-grade inflammation and oxidative stress play a fundamental part in chronic kidney disease-associated cardiovascular complications, and to a larger extent than in the general population.19 A key element in this proinflammatory status is the activation of the redox-sensitive nuclear transcription factor kappa B (NF-κB) (figure 2), in response to several factors including oxidative stress (reactive oxygen species [ROS]), mitochondrial dysfunction, Panel 1: Consequences of the uraemic syndrome Cardiovascular Hypertension, fluid overload, cardiac decompensation, vascular damage and stiffness, cardiovascular events, pericarditis Haematological Anaemia, erythrocyte fragility, immune dysfunction (susceptibility to infections, low response to vaccination), inflammation, hypercoagulability, bleeding tendency Endocrine Hyperparathyroidism, insulin resistance, impotence, infertility, thyroid dysfunction, hyperaldosteronism, growth disturbance, adipokine dysbalance, klotho deficiency and FGF-23 excess, active vitamin D deficiency Osteoarticular problems Osteomalacia, osteodystrophy, adynamic bone disease, β2-microglobulin amyloidosis, muscle weakness, fractures, bone pain, calciphylaxis and cardiovascular calcification Neurological Polyneuropathy, coordination disturbances, tremor, cognitive dysfunction, decreased attention span, coma Gastrointestinal Anorexia, gastroparesis, nausea, vomiting Dermatological Skin atrophy, pruritus, calciphylaxis Stomatological Periodontitis, stomatitis Nephrological Renal tubular damage, progression of kidney failure Other Malnutrition, changes in drug protein binding, changes in metabolism, hyperkalaemia, metabolic acidosis
  • 3. www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 3 Review several uraemic retention products, infection, vasoactive substances, dyslipidaemia, and inflammation. Activation of NF-κB is associated with the release of proinflammatory cytokines (tumour necrosis factor α, interleukin 1β, interleukin 6, soluble tumour necrosis factor-like weak inducer of apoptosis) and activation of monocyte chemoattractant protein-1 and profibrotic transforming growth factor β1. Inflammation and oxidative stress are tightly interconnected. NF-κB activation initiates production of ROS and vice versa, but under normal conditions, nuclear factor-erythroid-2-related factor 2 (a regulator of resistance to oxidants via expression of antioxidants and cytoprotective agents) is also activated. This balance might be disrupted in chronic kidney disease.20 A substantial number of uraemic toxins (eg, indoxyl sulfate, p-cresyl sulfate, the cytokines, advanced glycation end products, trimethylamine N-oxide) are pro- inflammatory, as are compounds that until 5 years ago were thought to be inert, like urea (for a more comprehensive list, see appendix). Furthermore, several other factors that are inflammatory are partly or entirely unrelated from uraemic retention products: infection, dialysis fluid contamination, dialysis bioincompatibility, concomitant diseases (eg, autoimmune disorders), and vasoactive agents (figure 2). Cardiac and arterial complications are the principal complications of the uraemic syndrome. The cardiomyopathy in chronic kidney disease is characterised by left ventricular hypertrophy, capillary rarefaction, and interstitial fibrosis, with predominant diastolic dys- function. Arrhythmias, sudden death, and heart failure are the main consequences of these cardiac com- plications. NF-κB activation by vasoactive substances affects cardiomyocyte growth, fibrosis, and apoptosis, inducing hypertrophy.19 In addition to the proinflammatory factors mentioned above, some uraemic toxins and several factors that are independent of uraemic toxins are damaging for heart and vessels: indoxyl sulfate, ADMA, endothelin, oxalate (uraemic retention products), hypertension, sodium and fluid retention, primary diseases (eg, amyloidosis), infections (endocarditis). Compared with the general population, arterial disease in populations with chronic kidney disease is premature, and characterised by endothelial dysfunction, arterial remodelling with arterial calcifications (figure 3A, 3B), vascular stiffness, and the transition of the vascular smooth muscle cells to an osseous phenotype.21 Vascular stiffness, which is closely linked to endothelial dysfunction, precedes arterial calcifications that further exacerbate stiffening.22 Arterial calcifications are due to an GFR* Definition Therapeutic approaches Pathophysiological changes Mortality Stage 1 >90 Normal kidney function, but urine abnormalities (eg, haematuriaor albuminuria†), proven structural kidneydamage, or genetictrait Observation, blood pressure and cardiovascular risk factor control, preventionof cardiovasculardisease Klothodeficiency Increasedoverall and cardiovascular mortality + Stage 2 60–89 Mildly reduced kidney function associatedwith other findings (as in stage 1) Observation, blood pressure and cardiovascular risk factor control, preventionof cardiovasculardisease Klothodeficiency, elevated FGF-23 Increasedoverall and cardiovascular mortality + Stage 3A Stage 3B 45–59; 30–44 Moderately reduced kidney function Observation, blood pressure and cardiovascular risk factor control, preventionof cardiovasculardisease, treatmentof anaemia and metabolic bone disease, preventionof chronic kidney disease progression Klothodeficiency, elevated FGF-23 and PTH, malnutrition, hypertension, left ventricular hypertrophy, anaemia Increasedoverall and cardiovascular mortality ++ Stage 4 15–29 Severely reduced kidney function Planning for renal replacementtherapy, blood pressure and cardiovascular risk factor control, preventionof cardiovascular disease,treatmentof anaemia and metabolic bonedisease, preventionof chronic kidneydisease progression Klothodeficiency, elevated FGF-23 and PTH, malnutrition, hypertension, left ventricular hypertrophy, anaemia, hypertriglyceridaemia, hyperphosphataemia, metabolic acidosis, hyperkalaemia Increasedoverall and cardiovascular mortality +++ Stage 5 <15 Very severe kidney failureor end-stage kidneydisease Renal replacementtherapyor conservative approach, blood pressure and cardiovascular risk factor control, preventionof cardiovasculardisease, treatmentof anaemia and metabolic bone disease Klothodeficiency, elevated FGF-23 and PTH, malnutrition, hypertension, left ventricular hypertrophy, anaemia, hypertriglyceridaemia, hyperphosphataemia, metabolic acidosis, hyperkalaemia, need for renal replacementtherapy Increasedoverall and cardiovascular mortality ++++ FGF-23=fibroblast growth factor-23. GFR=glomerular filtration rate (mL/min per 1·73 m²). PTH=parathyroid hormone. *GFR can be directly measured (eg, clearance of inulin, iothalamate, or ethylene diamino acetic acid), or calculated based on serum markers, such as creatinine, cystatin C, or both and anthropometric parameters (estimated GFR– eGFR). †Albuminuria is classified as moderately increased (30–300 mg/g or 3–30 mg/mmol creatinine) and severely increased (>300 mg/g or >30 mg/mmol) and increases risk by one level unless the highest risk category has been reached. Severity of mortality is indicated as mild (+), mild-moderate (++), moderate (+++), and severe (++++). Table 1: Chronic kidney disease stages and their implications
  • 4. 4 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 Review imbalance of calcification inducers and inhibitors, and development of a senescence-associated secretory phenotype (SASP) in vascular smooth muscle cells.23 Inducers of arterial calcifications and SASP are all linked to secretion of proinflammatory cytokines. Inflammation activates the BMP2:BMP4, Msx2, and Wnt pathways, which promote increased transcription of genes associated with conversion of vascular smooth muscle cells to the osteochondrogenic phenotype (Runx2, osterix, Sox9).24 Several chronic kidney disease-linked and biochemical changes related to mineral bone disorder (figure 3C), such as hyperphosphataemia, are associated with arterial calcifications. Hyperphosphataemia also stimulates the production of ROS and thereby activation of NF-κB. The resultant inflammation decreases the concentration of calcification inhibitors and reduces klotho expression, which induces resistance to the phosphaturic effect of fibroblast growth factor 23 (FGF-23),25 bone resistance to parathyroid hormone (PTH), secondary hyperpara- thyroidism,26 and accelerated ageing.27 Telomere shortening, which is typical for ageing, is associated with cardiovascular disease manifestations.28 Inflammatory epigenetic changes, which, in part, involve inhibited gene methylation of s-adenosinemethionine, also contribute to inflammation and cardiovascular lesions.29,30 Uraemic bone disease is mainly triggered by increased concentrations of the uraemic toxin phosphate, which results in the enhanced generation of several compensatory factors to maintain serum phosphate con- centration, such as FGF-23 and PTH, at the cost of loss of bone structure and function. Independent of metabolite retention, ageing, inflammation, bone ischaemia, and inadequate renal production of active vitamin D metabolites further enhance bone degradation. Some patients develop adynamic bone disease, whereby low PTH hinders calcium deposition in the bone, favouring vascular calcification. In rare cases, patients develop calciphylaxis—a severe syndrome consisting of vascular calcification, thrombosis, and skin necrosis. In summary, inflammation, oxidative stress, and bone disease show specific features linked to chronic kidney disease and uraemia resulting in accelerated vascular damage and ageing. Malnutrition and dysbiosis Malnutrition and dysbiosis are two features of the uraemic syndrome that relate to digestive function (figure 4). Some uraemic retention products (such as cytokines leptin, ghrelin, and neuropeptide Y) can be implicated in reduced appetite and nutrient intake. p-cresyl sulfate causes insulin resistance, disturbances of lipid metabolism, and aberrant distribution of fat cells throughout the body. Inflammation—a common epi- phenomenon of chronic kidney disease—is associated with reduced appetite, at all chronic kidney disease stages, including chronic kidney disease stage 5 on dialysis.31 Other factors implicated in reduced appetite, independent of solute retention, are changes in taste and smell perception, reduced intestinal motility and absorption capacity, depression, and loss of aminoacids and protein via dialysis (figure 4). Consequently, patients might have a spontaneous reduction in energy intake, which can induce protein energy wasting even before starting dialysis (figure 3D). Although decreased protein intake could reduce generation of uraemic retention products, Panel 2: Non-traditional risk factors in the uraemic syndrome5 • Anaemia • Volume overload • Metabolic bone disorder and related mediators • Hyperphosphataemia • High levels of FGF-23 • Low expression of the anti-ageing factor α-klotho • Secondary hyperparathyroidism • Active vitamin D deficiency • Low grade inflammation • Oxidative stress • Uraemic retention products* • Post-translational protein modifications • Accumulation of atherogenic remnant lipoproteins • Endogenous nitric oxide synthase (NOS) inhibitors • High sympathetic activity • Insulin resistance • Exposure to bioincompatible dialysis conditions • Immune response activating membranes • Contaminated haemodialysate • Peritoneal dialysate containing glycation products • Haemodynamic instability during haemodialysis • Infections • General • Vascular access: arteriovenous fistula, catheters, and arteriovenous grafts • Dyskalaemia (hypokalaemia and hyperkalaemia) *For details, see appendix. Figure 1: Role of progression of kidney failure in defining the uraemic syndrome ↑ Angiotensin II ↑ Catecholamines ↑ Endothelin Glomerular hypertension and hyperfiltration Podocyte injury and proteinuria Glomerular and tubulo-interstitial fibrosis Progressive kidney dysfunction Inflammation Uraemic retention products Chronic kidney disease
  • 5. www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 5 Review muscle protein catabolism, indirectly caused by acidosis and inflammation, might enhance the accumulation of toxic metabolites. Thus, a diet containing the appropriate nutrients in the appropriate quantity and balance is crucial for wellbeing and to reduce metabolic poisoning in patients with chronic kidney disease. The intestinal microbiota plays an important part in regulating the host’s nutritional status, metabolism, and different aspects of immunity. Dysbiosis has been linked to several chronic diseases. The few studies of dysbiosis in patients with chronic kidney disease suggest changes in the abundance and metabolic characteristics of intestinal microbiota as compared with healthy individuals.32 These changes induce the preferential and abundant generation of uraemic retention products with toxic effects. Additionally, chronic kidney disease and uraemic retention cause structural and functional changes of the intestinal epithelial barrier, inducing inflammation via different pathways.33 Although progression of kidney disease is clearly linked to changes in intestinal microbiota, whether or not uraemic retention products play a part in these changes is not clear. Other conditions that are associated with kidney disease could also affect intestinal microbiota—eg, ageing, diabetes, obesity, changes in nutrient intake and diet, and antibiotic intake. Anaemia Kidney dysfunction is associated with decreased concentrations of haemoglobin in blood, mainly due to deficiency of and resistance to erythropoietin and absolute and relative iron deficiency (figure 5). The kidneys Figure 2: Key factors in the microinflammatory condition of chronic kidney disease All elements lead to the activation of NF-κB and the expression of several proinflammatory factors. ROS=reactive oxygen species. AGEs=advanced glycation end products. ADMA=asymmetrical dimethylarginine. IS=indoxyl sulfate. pCS=p-cresyl sulfate.TMAO=trimethylamine-N-oxide. LPS=lipopolysaccharide. NF-κB=nuclear factor kappa B. OxLDL=oxidised low density lipoprotein. MCP-1=monocyte chemoattractant protein-1.TGF-β=transforming growth factor β. ROS Uraemic retention products AGEs, ADMA, IS, pCS, TMAO, phosphate Infectious agents Infection, endotoxin (LPS), bacterial DNA, peptidoglycan Vasoactive agents Angiotensin II, noradrenaline, endothelin I, aldosterone Cytokines and chemokines Proinflammatory (eg, interleukin 6, MCP-1), profibrotic (eg,TGF-β) ↑NF-κB Mitochondrial dysfunction Dialysis-related factors Bioincompatibility, dialysis fluid impurities Dyslipidaemia OxLDL Figure 3:Typical clinical problems related to the uraemic syndrome Vascular calcifications (A, B); rugger-jersey spine: alternating osteosclerosis and osteopenia (C); uraemic malnutrition (D); central lesions: silent infarctions (E), microbleeds (F); white matter lesions (G). A B C E F G D
  • 6. 6 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 Review respond to low tissue oxygen by increasing erythropoietin production, which induces erythropoiesis. Decline in kidney function impedes this mechanism. Iron deficiency is primarily attributable to ineffective intestinal iron absorption, because inflammation associated with chronic kidney disease elevates hepcidin, an inhibitor of iron export from enterocytes to plasma. Insufficient oral iron intake and blood loss from occult gastrointestinal or urogenital bleeding, repetitive blood sampling, and dialysis further drive iron deficiency. Finally, vitamin B12 and folate deficiency, shortened erythrocyte lifespan, and frequent infections are also contributing factors.34 Erythropoietin and iron deficiencies are the main drivers of renal anaemia, and these effects are largely independent of uraemic retention; however, uraemic toxins can affect erythrocyte production, solidity, and survival (figure 5). Neurological dysfunction High incidence of cerebrovascular diseases (such as stroke, white matter lesions, and intracerebral microbleeds; figure 3E–G) and cognitive disorders (linked to vascular abnormalities, neurodegenerative abnormalities, or both; figure 6) have been reported in chronic kidney disease.35 Cerebrovascular disorders could be linked to traditional and non-traditional cardiovascular risk factors, such as oxidative stress, chronic inflammation, endothelial dysfunction, vascular calcification, anaemia, dialysis-related haemodynamic instability, and uraemic retention products.36 Uraemic toxins could affect cerebrovascular diseases or cognition either by direct functional effects or by modulation of other factors (such as inflammation and oxidative stress). β2-microglobulin—a typical uraemic toxin— was shown to impair cognitive function and act as a systemic, pro-ageing factor.37 Other uraemic retention products frequently associated with neurological dysfunction are guanidines. Phenols and indoles have also been linked to neurological disturbances. Further contributing factors that are partly linked to retention are inflammation, vascular disease, and hypertension. Factors that are more or less independent of solute retention are depression, enhanced activity of drugs that treat psychiatric disorders or have psychological effects (eg, sleeping pills), and blood pressure changes related or not to dialysis (figure 6). Management The studies, guidelines, position statements, and reviews referred to in this section are summarised in the appendix. Treatment aimed at modifying risk factors will be discussed in broad terms independently of underlying intermediate mechanisms. Of note, positive interventions in the general population, could counterintuitively cause or amplify complications in chronic kidney disease. This section on management includes 56 original studies (appendix), of which only 35 (63%) are randomised controlled trials or meta-analyses. Only six (11%) of these randomised controlled trials or meta-analyses have an unambiguously positive intervention in a study taking into account more than 1000 patients (all but one of these six are meta-analyses). Lifestyle Emphasis is often laid on pharmacological drug interventions in cardiovascular and kidney protection in chronic kidney disease, of which the benefit is not necessarily unequivocal. Several general lifestyle measures could also be beneficial, and need negligible societal investment, except for educational initiatives. In a large meta-analysis, regular exercise training was associated with improved outcomes in chronic kidney disease. However, risk of bias of the included studies was high.38 In a single-centre, observational study, heavy smoking was associated with increased risk of progression of chronic kidney disease, especially in patients with hypertensive and diabetic nephropathy.39 In a large, cross-sectional follow-up study, no significant Figure 4: Role of malnutrition and dysbiosis and chronic kidney disease in defining the uraemic syndrome Figure 5: Role of renal anaemia and chronic kidney disease in defining the uraemic syndrome ↓ Nutritional status ↑ InflammationUraemic retention products Chronic kidney disease Protein energy wasting Intestinal modifications and translocation ↑ Hepcidin production ↓ Erythropoietin generation Iron deficiency Inflammation, infection Deficient iron intake, blood loss Anaemia Deficiency of folic acid and vitamin B12, erythrocyte fragility Chronic kidney disease Figure 6: Role of neurological dysfunction and chronic kidney disease in defining the uraemic syndrome Cognitive dysfunction Neurological damage Oxidative stress, inflammation, endothelial dysfunction, vascular calcification, dialysis-related haemodynamic instability Uraemic retention products Chronic kidney disease Cerebrovascular disease
  • 7. www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 7 Review association was recorded between pre-dialysis obesity and progression of chronic kidney disease.40 Another cohort study,41 however, showed a clear association between BMI more than 30 kg/m² and progression of kidney disease, especially from the age of 40 years on. The relative survival advantage in patients who are overweight and on dialysis (obesity paradox) is remarkable, but can be attributed to poor outcomes of their malnourished counterparts,42 partly related to the negative effects of inflammation. Unfortunately, focused and conclusive studies on lifestyle in chronic kidney disease are scarce. Diet A key factor of the dietary management of the uraemic syndrome is to supply enough nutrition, but not too much, because the body needs fuel, but cannot handle well enough the end products of cellular metabolism. This notion is particularly true of proteins because they cannot be stored to adjust for intake fluctuations. Accumulation of uraemic retention products and metabolic poisoning in chronic kidney disease can be ameliorated by reduced protein intake, which, rather than being left to spontaneous evolution, should be prescribed as a well balanced, carefully controlled, low- protein diet. Thus, patients with stages 3–5 chronic kidney disease who are not yet on dialysis should be educated to pursue an equilibrium between sufficient energy intake (at least 30 kcal/kg per day) and reduced protein intake (0·6 to 0·8 g/kg per day, 50% of which should be high value proteins of animal origin containing essential aminoacids (table 2).43 This approach reportedly reduces serum urea by 30%, and improves insulin resistance, phosphate and parathyroid metabolism, blood pressure, and anaemia.43 Reduction of phosphorus intake allows better control of mineral bone disease and reduces the need for oral phosphate binders. Sodium intake should be targeted to a maximum of 6 g sodium chloride per day and, if possible, lower (table 2), except in the case of salt-losing nephropathy, in which intake can be higher. Specialised dietitians should regularly be involved in the implementation of advice to correctly educate the patient. At least three encounters per year have been suggested for the first year of care.44 This only seems practical from chronic kidney disease stage 4 onwards. 24 h urine collections allow the monitoring of daily protein intake (via urea measurements) and sodium intake and should be done twice yearly to control and implement the diet. If intentional weight loss is planned, the benefit should be balanced against the risk of protein wasting, particularly in the late stages of chronic kidney disease. On dialysis, additional catabolism takes place in response to chronic inflammation, surgical interventions, recurrent sepsis, and nutrient losses during the dialysis procedure. Whereas caloric intake can be maintained above 30 kcal/kg per day, protein intake should be increased to more than 1·0 g/kg per day of protein in patients on haemodialysis and 1·2 g/kg per day in peritoneal dialysis (table 2).45 In stable patients, protein intake can be calculated as protein catabolic rate from pre-dialysis and post-dialysis urea measurements. Fluid intake is an indicator of nutritional status. In patients with anuria, interdialytic weight gain was positively associated with protein catabolic rate.46 Reduced weight gain, or a low serum urea or creatinine pre-dialysis are negative signs and should prompt rapid dietary intervention.47 Nevertheless, fluid intake that is too high and interdialytic weight gain increase intradialytic ultrafiltration rate, which should be avoided because of its association with mortality in patients on dialysis.48 Dietary phosphate intake should be controlled without causing a reduction in protein intake, because a reduction in protein intake is associated with worse survival.49 Foods and beverages rich in phosphate, such as preserves, processed meat, frozen foods, dairy products, and soft drinks should be discouraged. Potassium intake that is too high should also be avoided (table 2). For more detailed dietary advice see appendix. Pharmacological treatment Chronic kidney disease is a challenging disease with few treatment options to prevent progression. In clinical practice, measurements of albumin-to- creatinine ratio and eGFR allow risk classification for chronic kidney disease progression and cardiovascular mortality. Additional biomarkers might be helpful in refining these strategies, but their use needs further validation. Chronic kidney disease stages 3–5, not on dialysis Receiving haemodialysis Receiving peritoneal dialysis Had transplant <3 months ago Had transplant ≥3 months ago Diet Low protein Standard protein Standard protein High protein Low protein Target protein intake (g/kg per day) 0·6–0·8 (or less with keto- analogues) 1·0–1·2 1·2 1·4 0·6–0·8 Target energy intake (kcal/kg per day) 30–35 30–35 30–35 30–35 30–35 Salt intake (mg per day)* <6000 <5000 <5000 <6000 <6000 Potassium intake (mg per day) 2500 2500 2500 Free Free† Phosphorus intake (mg per day) <800 <1000 <1000 Free <800 Wasting risk ++ +++ +++ +++ + Overweight risk ++ + + ++ +++ *Except in case of salt-losing nephropathy. †Except in case of hyperkalaemia (>5·5 mmol/L). Risk of being overweight indicated as mild (+), moderate (++), and severe (+++). Table 2: Integrated optimum nutrition of patients with chronic kidney disease
  • 8. 8 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 Review The mainstays of non-specific prevention of chronic kidney disease progression, irrespective of cause, include blood pressure control and proteinuria-directed strategies to preserve residual kidney function, with special emphasis on angiotensin-converting enzyme (ACE) inhibitors or angiotensin-receptor blockers.50 In diabetic nephropathy, strategies that focus on strict glycaemic control slow down progression.51 Although hyperlipidaemia could play a role in progression, there is no consensus regarding statin treatment for nephroprotection.52,53 In a randomised controlled trial54 of bicarbonate supplementation with a target serum bicarbonate concentration greater than 23 mmol/L, progression of chronic kidney disease was slowed and nutritional status improved. In another randomised controlled trial,55 reduction of uric acid by allopurinol treatment was nephroprotective. In a trial56 in patients with diabetic nephropathy, pentoxifylline reduced the rate of decline in eGFR and proteinuria. The findings of these three studies should, however, be confirmed in larger controlled trials. Uraemia is a strong, independent risk factor for cardiovascular disease, therefore, patients with chronic kidney disease need close cardiovascular follow-up as part of their management strategy to prevent chronic kidney disease progression. Approaches targeting inflammation (eg, anti- inflammatory drugs or statins) did not provide health benefits for patients with chronic kidney disease. In the AURORA trial,57 rosuvastatin lowered serum C-reactive protein concentrations by 27%, but failed to reduce mortality or the risk of cardiovascular events. Resistance to interventions targeting inflammation could depend on the severity of inflammation in end-stage kidney disease (in the AURORA trial,57 median C-reactive protein in the treatment group remained four times above upper normal concentrations), and on its multifactorial origin. Interference of proinflammatory and oxidative mechanisms—by the reduction of NF-κB activation and activation of nuclear factor-erythroid-2- related factor 2—seemed an attractive option to reduce the enhanced risk of all-cause and cardiovascular death in advanced chronic kidney disease. However, a trial58 testing a NF-κB blocker (bardoxolone) in patients with chronic kidney disease and type 2 diabetes was prematurely terminated because of excessive risk of cardiovascular events in the bardoxolone group. In pre-dialysis chronic kidney disease, the well established cardiovascular benefit of antihypertensive treatment has led to specific guidelines recommending target blood pressures of 140/90 mm Hg or less in chronic kidney disease (130/80 mm Hg or less in the presence of proteinuria).59 The relation between pre- dialysis or post-dialysis blood pressure and mortality in patients on dialysis is inverse or U-shaped, which is a classical example of reverse causality. However, dialysis- related blood pressure values hardly reflect true blood pressure burden in patients on haemodialysis.60 Out-of- dialysis systolic blood pressure, in fact, predicts a linear increase in the risk of death from 110 mm Hg or higher,61 as in the general population. Antihypertensive treatment reduces the incidence of death and cardiovascular events at all risk levels and in absolute terms most of all in patients with a higher baseline risk.62 In the SPRINT trial,63 patients with hypertension randomised to target systolic blood pressures of less than 120 mm Hg had fewer cardiovascular events than in those targeted to less than 140 mm Hg, in general and in the subgroup with chronic kidney disease. In patients with end-stage kidney disease, including patients with heart failure, the use of antihypertensive drugs reduces mortality,64 but treatment should be titrated to tolerable levels—ie, to minimise the risk of hypotension by autonomic dysfunction or arterial stiffness,65 which enhance the risk for ischaemic events. Optimisation of volume control at constant ultrafiltration rate (eg, by more frequent or extended dialysis), and judicious use of antihypertensive drugs accounting for comorbidities and pharmacokinetic profile66 could reduce cardiovascular risk in patients on dialysis. In a single centre-based observational study,67 extended haemo- dialysis (24 h per week) in an older population resulted in good blood pressure control without antihypertensive drugs. Controlling hyperglycaemia in patients with renal failure and diabetes is difficult because of the higher risk of hypoglycaemia as compared with the diabetic population without chronic kidney disease.68 Target HbA1c levels should account for additional risk factors, with the aim to achieve strict control in those with low risk, but more leniency in those with comorbidities and high risk for hypoglycaemia.69 Patients with diabetes, chronic kidney disease, and heart failure, ischaemic heart disease, or hypertension, can be treated with renin– angiotensin system blockers at a maximally tolerated dose. However, combinations of ACE inhibitors and angiotensin II receptor blockers should be used carefully in patients with chronic kidney disease not yet on dialysis69 to avoid hyperkalaemia, kidney impairment, and hypotensive symptoms,50 although in a meta- analysis,70 a positive effect on kidney function preservation was suggested with this combination in people with diabetes. Empagliflozin, a sodium-glucose co-transporter inhibitor, significantly reduced a composite of cardio- vascular events and death in patients with diabetes type 2 and eGFR more than 30 mL/min per 1·73 m² at high cardiovascular risk (26% with chronic kidney disease stage 3).71 Because of malnutrition and inflammation, hyper- cholesterolaemia often regresses as chronic kidney disease advances. Identification of dyslipidaemia (high total or LDL cholesterol, low HDL cholesterol, high triglycerides) in patients with end-stage kidney disease is
  • 9. www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 9 Review deemed useful by existing Kidney Disease Improving Global Outcomes (KDIGO) guidelines to assess overall cardiovascular risk,72 but follow-up of lipid concentrations is not recommended because of the absence of evidence that this approach improves clinical outcomes. Findings from the SHARP trial73 and subsequent meta-analysis53 showed the usefulness of an absolute reduction of LDL cholesterol in patients with chronic kidney disease to decrease the associated cardiovascular risk. Cholesterol-lowering therapy is especially recommended by KDIGO in all patients with chronic kidney disease at stage 3–5, who are more than 50 years of age and not on dialysis.72 However, the benefit of statins was less prominent in end-stage kidney disease,57,74 which discouraged their use in end-stage kidney disease, irrespective of inflammation or malnutrition. In patients already treated, however, this therapy can be continued.69 Results from a large end-stage kidney disease cohort in the USA showed no survival benefit of lowering phosphate concentrations with calcium carbonate or acetate.75 Findings from another observational study76 showed a reduction in death risk in patients who were given any type of phosphate binder. Results from a meta- analysis77 of seven trials in haemodialysis patients and one in moderate chronic kidney disease showed lower mortality in patients given the non-calcium-based phosphate binder sevelamer. However, this apparent benefit (heterogeneity I²=89%) is potentially skewed by a study in patients on haemodialysis in which sevelamer reduced the death risk by 91%, hence, the issue remains unresolved. Serum phosphate concentrations between 0·68 and 1·93 mmol/L do not associate with an excessive death risk in dialysis patients.78 The decision to treat hyperphosphataemia should take into account serum calcium, PTH, and serum vitamin D concentrations from the early stages of chronic kidney disease-associated mineral bone disorder.78 Secondary hyperparathyroidism has been implicated in the cardiovascular risk of chronic kidney disease. However, improvement of mortality and cardiovascular outcome was inconclusive in a large trial79 of calcium receptor agonist cinacalcet in secondary hyperparathyroidism, and this finding was confirmed by meta-analysis.80 Many patients with chronic kidney disease have low serum concentrations of 25-hydroxyvitamin D or calcitriol, but a randomised, placebo-controlled trial testing their supplementation on hard endpoints is not yet available. However, these supplements might help control hyperparathyroidism. The effect of modulation of uraemic retention product concentrations on metabolic bone disease and cardiovascular outcomes needs to be investigated because several of these retention products are associated with both complications.81 Malnutrition should be diagnosed using the International Society for Renal Nutrition and Metabolism protein energy wasting criteria,82 which include clinical, laboratory, and body composition measurements. Wasting scores can predict survival and should be used more frequently to help identify patients at risk of death from wasting. An increase in nutrient intake is the key intervention and has been shown to be effective even in the context of chronic inflammation. One or two units of oral supplements taken separately from regular meals to avoid further reductions in spontaneous intake should be used as a primary measure. The FINE study,83 which compared oral nutritional supplements with or without parenteral nutrition in dialysis patients with protein energy wasting, was negative for its primary endpoints (eg, mortality). Oral supplements were well tolerated and increase total nutrient intake, serum albumin, and serum prealbumin.83 Results of a multivariate analysis showed that mortality was reduced if serum prealbumin increased above 30 mg/L in the first 3 months of supplementation.83 Therefore, in ambulatory patients, if nutritional status does not improve within 4 weeks, enteral feeding should be considered. For hospital inpatients, intensive nutrition through a nasogastric tube or percutaneous gastrostomy can be considered if oral energy intake is less than 20 kcal/kg per day for more than 10 days. Few randomised controlled trials have been done that investigate the effect of the restoration of intestinal symbiosis in chronic kidney disease by giving patients prebiotics (selectively fermentable ingredients), probiotics (live biotherapeutics), or synbiotics (com- bination of prebiotics and probiotics) to reduce circulating concentrations of uraemic retention products, inflammation, oxidative stress, and progression of chronic kidney disease. Specific prebiotics decrease serum indoxyl sulfate and urea nitrogen.84,85 Results from a study86 in patients receiving peritoneal dialysis showed a significant reduction in concentrations of pro- inflammatory cytokines, an increase in concentrations of anti-inflammatory cytokines, and better preservation of residual kidney function in the group receiving probiotics. Synbiotic therapy reduces the concentration of p-cresol.87 In several of the above trials, however, the intervention was coupled to a low-protein diet, which could itself decrease uremic toxin concentration and progression of kidney failure. The actual effect of restoring intestinal symbiosis on the intestinal microbiota profile or on hard outcomes was not assessed in any of the above trials. Reduction of indoxyl sulfate by intestinal sorbents, such as AST-120, stopped progression of chronic kidney disease in small Japanese randomised trials.88,89 This benefit was not confirmed in a large European–American randomised controlled trial.90 Interventions improving symbiosis need further study before their efficacy can be accepted. The conventional approaches for iron repletion in chronic kidney disease are oral ferrous salts or intravenous colloidal compounds. High concentrations of hepcidin,
  • 10. 10 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 Review however, make intestinal iron uptake ineffective. Additionally, oral iron intake further contributes to the pill burden of patients with chronic kidney disease and causes severe gastrointestinal side-effects. Therefore, intravenous iron has gained popularity in the past 5 years. However, experimental and observational data suggest unwanted immunological, cardiovascular, and renal effects of such treatment,91,92 which might differ between classic preparations and new, more stable formulations.91 Long-term prospective studies are, however, unavailable. The introduction of recombinant human erythropoietin was one of the major steps forward in the treatment of renal anaemia. Both short-acting and long-acting erythropoiesis stimulating agents are in use at present. According to the existing guidelines, the decision of whether and when to start treatment with an erythropoiesis stimulating agent should be individualised and take into account related risks and anaemic symptoms.34 The target values for erythropoiesis stimulating agent therapy are 10·0–12·0 g/dL.34 These drugs should be used with care in patients at high risk of stroke or with active or past malignancy.34 Blood transfusions might be considered, especially in patients with haemoglobin concentration of less than 7·0 g/dL, who are resistant to erythropoiesis-stimulating agents, or at potential risk of complications with erythropoiesis stimulating agent therapy.93 However, the risk of developing panel reactive antibodies should be taken into account in transplant candidates. Few data are available on the effect of treatment for chronic kidney disease on risk of stroke and other neurological disorders in patients with this disease, particularly in those with end-stage kidney disease. Patients with chronic kidney disease have been under- represented in the cardiovascular trials that prove net benefit of commonly used preventive treatments (eg, antihypertensive drugs, low-dose aspirin, carotid revascu- larisation, and thromboprophylaxis for atrial fibrillation), and safety and efficacy of many of these treatments in chronic kidney disease remains uncertain. Conflicting results have been reported on the effect of statins in patients with chronic kidney disease. In a Cochrane review,53 the authors concluded that statins did not necessarily have effects on stroke in patients with chronic kidney disease who did not need dialysis, by contrast with another meta-analysis94 in which statins were associated with a decrease in cardiovascular disease in chronic kidney disease, including stroke. Correcting anaemia might not prevent stroke and could even increase its risk in chronic kidney disease.95 Moreover, the effect of anaemia treatment on cognitive disorders remains a matter of debate. Oxidative stress, inflammation, or uraemic retention products could contribute to neurological disorders in patients with chronic kidney disease. Thus, treatments modulating these factors might improve neurological outcomes. To the best of our knowledge, no direct interventional trials have targeted these abnormalities. However, improvement of intra-dialytic haemodynamic stability by cooling dialysate has been shown to protect against brain injury.65 Renal replacement therapy Replacement of kidney function requires trans- plantation or dialysis, and haemodialysis and peritoneal dialysis are the main dialysis modalities (for more on different dialysis strategies see appendix). Haemodialysis is usually done three times weekly, whereas peritoneal dialysis clears retention products with lower efficiency but does so continuously. The high efficiency solute removal provided by a single haemodialysis session over 4 h should be extrapolated to the full interdialytic interval of 48–72 h, by comparison with kidney removal capacity. Thus, renal replacement therapy through dialysis allows the anuric patient to have a kidney-equivalent filtration rate of only 5–10 mL/min for small molecules that are the size of urea or creatinine, which results in higher uraemic solute concentrations than is normal. In general, dialysis removes retention products of higher molecular weight or those bound to protein less efficiently than the normal kidney. Oral phosphate binders are frequently needed to maintain serum phosphate concentrations that are close to normal. The standard dialysis clearance of small proteins such as cytokines, FGF-23, and β2-microglobulin is negligible, as compared with normal kidneys. Clearance of β2-microglobulin (11·8 kDa), deemed representative of middle-sized molecules, was estimated at 3–19 mL/min for a conventional haemodialysis session (ie, 0·17–1·0 mL/min when averaged over 72 h), as compared with 65 mL/min in a normally functioning kidney.96 Conventional haemodialysis relies on diffusion for solute removal, whereas haemodiafiltration also depends on convective transport. This additional mechanism enhances the removal of middle-sized molecules, such as β2-microglobulin and FGF-23, but has little effect on many protein-bound retention products.97,98 However, β2-microglobulin clearance per single haemodiafiltration session is still far from normal kidney removal rates when averaged over 48–72 h (3·6 mL/min higher than conventional haemodialysis).99 Despite this poor efficiency, findings from a randomised controlled trial100 published in 2013, suggested that haemodiafiltration could improve hard outcomes versus standard dialysis, but a meta-analysis of all studies showed improvement only for cardio- vascular mortality, but not overall mortality. Additionally, the reliability of potential benefits found in this meta-analysis was deemed debatable because the studies included in the analysis had several methodological limitations.101 Dialyser design changes might further improve removal of middle molecules.102 However, for each additional 20 mg of β2-microglobulin removed in a single session
  • 11. www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 11 Review (less than 10% increase), 1·24 g of albumin were lost (40% increase).102 Researchers who aim to increase protein- bound solute removal are exploring how to increase the release of uraemic retention products from their protein- bound states,103 for example by including adsorbents, but length of dialysis using these technologies is a limiting factor.97 Extension of haemodialysis time, in daily or extended regimens, either at home or in centre, can improve weekly solute clearance.97,98 Transplanted kidneys develop hypertrophy and mean eGFR is 60–70 mL/min per 1·73 m² 1 year after transplantation.104 However, in a cross-sectional study of graft recipients 5 years after their transplantation, more than 70% of patients had measured GFR of less than 60 mL/min per 1·73 m², and thus, had limited removal capacity.105 Serum β2-microglobulin concentrations of 3·0 mg/L or more were observed in 58% of kidney transplant recipients at discharge.106 Kidney graft handling of certain uraemic retention products might be abnormal. For example, phosphate leakage is frequent early after transplantation, especially in patients with severe hyperparathyroidism.105 Over time, kidney grafts lose function—the half-life of a cadaveric kidney graft is 9 years and 12 years for a living-donor graft. Conclusion Patients with chronic kidney disease have complex pathophysiology for which the underlying mechanisms intertwine (appendix). Inflammation and disturbed bone homoeostasis in particular lead to complications and high and accelerated mortality. Management (appendix) cannot always be based on high-level evidence, because of difficulties in the recruitment of patients with sufficiently homogeneous background of primary disease, metabolic features, and response to the uraemic syndrome. Specific therapeutic recom- mendations are, therefore, based on an amalgam of high-level and lower-level evidence and uraemia-related pathophysiological reasoning. Therapeutic approaches cannot always be extrapolated from the general population, because beneficial interventions in the general population often have a different effect in populations with chronic kidney disease. Treatment of one aspect of the uraemic syndrome might exacerbate other deleterious elements. In future studies, a more holistic therapeutic approach to cope with the high mortality of this disease could be more useful rather than the pursuit of one single factor as is usual practice in randomised controlled trials. Therapeutic approaches should focus on additional outcomes beyond mortality, especially standardised quality of life. Consultation with patients to understand what is important to them might be useful for the definition of patient-related outcomes. Contributors All authors wrote the first draft of one or more sections of the Review, and all contributed to the editing of subsequent versions. RV coordinated the initiative, merged the texts, and did general editing. Declaration of interests RV has received travel grants and honoraria from Nipro, Bayer, and Fresenius Medical Care, and has acted as a consultant for Fresenius Medical Care and Debiotech. DF has acted as a consultant for Fresenius Kabi. GHH has received study grants from Pharmacosmos and Fresenius Medical Care. ZAM has received speaker’s honoraria and research grants from Amgen, Genzyme, Fresenius Medical Care, and Shire. AO has received speaker’s honoraria and research grants from Amgen, Genzyme, Fresenius Medical Care, Servier, and Shire. PR has received honoraria from Baxter-Gambro, Fresenius, and Relypsa. AW has received speaker’s and consulting honoraria from Amgen, Fresenius Medical Care, Astellas, Roche, GlaxoSmithKline, Pharmacosmos, and Teva. GG, MK, FM, CZ, and GML declare no competing interests. Acknowledgments The European Renal and Cardiovascular Medicine (EURECA-m) working group is one of the working groups of the European Renal Association—European Dialysis and Transplant Association (ERA-EDTA) and is composed of members whose careers have been devoted to unravelling the pathophysiology of cardiovascular disease in chronic kidney disease and to defining accurate therapeutic approaches to address this problem. References 1 Almeras C, Argilés A. The general picture of uremia. Semin Dial 2009; 22: 329–33. 2 Meyer TW, Hostetter TH. Uremia. N Engl J Med 2007; 357: 1316–25. 3 Roberts MA, Polkinghorne KR, McDonald SP, Ierino FL. Secular trends in cardiovascular mortality rates of patients receiving dialysis compared with the general population. Am J Kidney Dis 2011; 58: 64–72. 4 Chapman JR. What are the key challenges we face in kidney transplantation today? Transplant Res 2013; 2 (suppl 1): S1. 5 Matsushita K, van der Velde M, Astor BC, et al, for the Chronic Kidney Disease Prognosis Consortium. Association of estimated glomerular filtration rate and albuminuria with all-cause and cardiovascular mortality in general population cohorts: a collaborative meta-analysis. Lancet 2010; 375: 2073–81. 6 Vanholder R, Massy Z, Argiles A, et al. Chronic kidney disease as cause of cardiovascular morbidity and mortality. Nephrol Dial Transplant 2005; 20: 1048–56. 7 Jun M, Lv J, Perkovic V, Jardine MJ. Managing cardiovascular risk in people with chronic kidney disease: a review of the evidence from randomized controlled trials. Ther Adv Chronic Dis 2011; 2: 265–78. 8 Ortiz A, Covic A, Fliser D, et al, for the Board of the EURECA-m Working Group of ERA-EDTA. Epidemiology, contributors to, and clinical trials of mortality risk in chronic kidney failure. Lancet 2014; 383: 1831–43. 9 de Jager DJ, Grootendorst DC, Jager KJ, et al. Cardiovascular and noncardiovascular mortality among patients starting dialysis. JAMA 2009; 302: 1782–89. Search strategy and selection criteria We searched PubMed, Embase,Webof Science, andthe Cochrane Library for articles published in Englishupto Dec 1, 2015. Searchterms relatedtothe maintopicsofthis Review wereused—eg, “metabolic bonedisease”or “inflammation” and “chronic kidneydisease”, “chronic renal failure”or “chronic kidneydisease” and “physiopathology”, “treatment”, “therapy”, “outcomes”, “morbidity”,or “mortality”. Randomised controlledtrialsor largeobservational studieson hard outcomeswere retained by preference. Reference listsofthe resulting articleswere searched forother relevant papers. Review articles are citedto provide readerswith moredetails and more referencesthan space permits inthis narrative review, andwere selected fortheir comprehensiveness and compactness.
  • 12. 12 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 Review 10 Morton RL, Schlackow I, Mihaylova B, Staplin ND, Gray A, Cass A. The impact of social disadvantage in moderate-to-severe chronic kidney disease: an equity-focused systematic review. Nephrol Dial Transplant 2016; 31: 46–56. 11 Weiner DE, Tighiouart H, Elsayed EF, et al. The Framingham predictive instrument in chronic kidney disease. J Am Coll Cardiol 2007; 50: 217–24. 12 Matsushita K, Coresh J, Sang Y, et al. Estimated glomerular filtration rate and albuminuria for prediction of cardiovascular outcomes: a collaborative meta-analysis of individual participant data. Lancet Diabetes Endocrinol 2015; 3: 514–25. 13 Vanholder R, De Smet R, Glorieux G, et al, for the European Uremic Toxin Work Group (EUTox). Review on uremic toxins: classification, concentration, and interindividual variability. Kidney Int 2003; 63: 1934–43. 14 Duranton F, Cohen G, De Smet R, et al, for the European Uremic Toxin Work Group. Normal and pathologic concentrations of uremic toxins. J Am Soc Nephrol 2012; 23: 1258–70. 15 Vanholder R, Baurmeister U, Brunet P, Cohen G, Glorieux G, Jankowski J, for the European Uremic Toxin Work Group. A bench to bedside view of uremic toxins. J Am Soc Nephrol 2008; 19: 863–70. 16 Tripepi G, Kollerits B, Leonardis D, et al. Competitive interaction between fibroblast growth factor 23 and asymmetric dimethylarginine in patients with CKD. J Am Soc Nephrol 2015; 26: 935–44. 17 Inker LA, Tonelli M, Hemmelgarn BR, Levitan EB, Muntner P. Comparison of concurrent complications of CKD by 2 risk categorization systems. Am J Kidney Dis 2012; 59: 372–81. 18 Wühl E, Schaefer F. Managing kidney disease with blood-pressure control. Nat Rev Nephrol 2011; 7: 434–44. 19 Gordon JW, Shaw JA, Kirshenbaum LA. Multiple facets of NF-κB in the heart: to be or not to NF-κB. Circ Res 2011; 108: 1122–32. 20 Ruiz S, Pergola PE, Zager RA, Vaziri ND. Targeting the transcription factor Nrf2 to ameliorate oxidative stress and inflammation in chronic kidney disease. Kidney Int 2013; 83: 1029–41. 21 Kooman JP, Kotanko P, Schols AM, Shiels PG, Stenvinkel P. Chronic kidney disease and premature ageing. Nat Rev Nephrol 2014; 10: 732–42. 22 Briet M, Boutouyrie P, Laurent S, London GM. Arterial stiffness and pulse pressure in CKD and ESRD. Kidney Int 2012; 82: 388–400. 23 Minamino T, Komuro I. Vascular cell senescence: contribution to atherosclerosis. Circ Res 2007; 100: 15–26. 24 Boström KI, Rajamannan NM, Towler DA. The regulation of valvular and vascular sclerosis by osteogenic morphogens. Circ Res 2011; 109: 564–77. 25 Lim K, Lu TS, Molostvov G, et al. Vascular Klotho deficiency potentiates the development of human artery calcification and mediates resistance to fibroblast growth factor 23. Circulation 2012; 125: 2243–55. 26 London GM, Marchais SJ, Guérin AP, de Vernejoul MC. Ankle-brachial index and bone turnover in patients on dialysis. J Am Soc Nephrol 2015; 26: 476–83. 27 Nitta K, Okada K, Yanai M, Takahashi S. Aging and chronic kidney disease. Kidney Blood Press Res 2013; 38: 109–20. 28 Raschenberger J, Kollerits B, Titze S, et al. Association of relative telomere length with cardiovascular disease in a large chronic kidney disease cohort: the GCKD study. Atherosclerosis 2015; 242: 529–34. 29 Zawada AM, Rogacev KS, Heine GH. Clinical relevance of epigenetic dysregulation in chronic kidney disease-associated cardiovascular disease. Nephrol Dial Transplant 2013; 28: 1663–71. 30 Zawada AM, Rogacev KS, Hummel B, et al. SuperTAG methylation-specific digital karyotyping reveals uremia-induced epigenetic dysregulation of atherosclerosis-related genes. Circ Cardiovasc Genet 2012; 5: 611–20. 31 Kalantar-Zadeh K, Block G, McAllister CJ, Humphreys MH, Kopple JD. Appetite and inflammation, nutrition, anemia, and clinical outcome in hemodialysis patients. Am J Clin Nutr 2004; 80: 299–307. 32 Wong J, Piceno YM, Desantis TZ, Pahl M, Andersen GL, Vaziri ND. Expansion of urease- and uricase-containing, indole- and p-cresol-forming and contraction of short-chain fatty acid-producing intestinal microbiota in ESRD. Am J Nephrol 2014; 39: 230–37. 33 Anders HJ, Andersen K, Stecher B. The intestinal microbiota, a leaky gut, and abnormal immunity in kidney disease. Kidney Int 2013; 83: 1010–16. 34 Locatelli F, Barany P, Covic A, et al. Kidney Disease: Improving Global Outcomes guidelines on anaemia management in chronic kidney disease: a European Renal Best Practice position statement. Nephrol Dial Transplant 2013; 28: 1346–59. 35 Bugnicourt JM, Godefroy O, Chillon JM, Choukroun G, Massy ZA. Cognitive disorders and dementia in CKD: the neglected kidney-brain axis. J Am Soc Nephrol 2013; 24: 353–63. 36 Chen YC, Su YC, Lee CC, Huang YS, Hwang SJ. Chronic kidney disease itself is a causal risk factor for stroke beyond traditional cardiovascular risk factors: a nationwide cohort study in Taiwan. PLoS One 2012; 7: e36332. 37 Smith LK, He Y, Park JS, et al. β2-microglobulin is a systemic pro-aging factor that impairs cognitive function and neurogenesis. Nat Med 2015; 21: 932–37. 38 Heiwe S, Jacobson SH. Exercise training in adults with CKD: a systematic review and meta-analysis. Am J Kidney Dis 2014; 64: 383–93. 39 Yacoub R, Habib H, Lahdo A, et al. Association between smoking and chronic kidney disease: a case control study. BMC Public Health 2010; 10: 731. 40 Babayev R, Whaley-Connell A, Kshirsagar A, et al. Association of race and body mass index with ESRD and mortality in CKD stages 3-4: results from the Kidney Early Evaluation Program (KEEP). Am J Kidney Dis 2013; 61: 404–12. 41 Lu JL, Molnar MZ, Naseer A, Mikkelsen MK, Kalantar-Zadeh K, Kovesdy CP. Association of age and BMI with kidney function and mortality: a cohort study. Lancet Diabetes Endocrinol 2015; 3: 704–14. 42 Park J, Ahmadi SF, Streja E, et al. Obesity paradox in end-stage kidney disease patients. Prog Cardiovasc Dis 2014; 56: 415–25. 43 Fouque D, Aparicio M. Eleven reasons to control the protein intake of patients with chronic kidney disease. Nat Clin Pract Nephrol 2007; 3: 383–92. 44 Laville M, Fouque D. Nutritional aspects in hemodialysis. Kidney Int Suppl 2000; 58 (suppl 76): S133–39. 45 Fouque D, Vennegoor M, ter Wee P, et al. EBPG guideline on nutrition. Nephrol Dial Transplant 2007; 22 (suppl 2): ii45–87. 46 Testa A, Plou A. Clinical determinants of interdialytic weight gain. J Ren Nutr 2001; 11: 155–60. 47 Moreau-Gaudry X, Jean G, Genet L, et al. A simple protein-energy wasting score predicts survival in maintenance hemodialysis patients. J Ren Nutr 2014; 24: 395–400. 48 Saran R, Bragg-Gresham JL, Levin NW, et al. Longer treatment time and slower ultrafiltration in hemodialysis: associations with reduced mortality in the DOPPS. Kidney Int 2006; 69: 1222–28. 49 Shinaberger CS, Greenland S, Kopple JD, et al. Is controlling phosphorus by decreasing dietary protein intake beneficial or harmful in persons with chronic kidney disease? Am J Clin Nutr 2008; 88: 1511–18. 50 Yusuf S, Diener HC, Sacco RL, et al, for the PRoFESS Study Group. Telmisartan to prevent recurrent stroke and cardiovascular events. N Engl J Med 2008; 359: 1225–37. 51 Writing Team for the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications Research Group. Sustained effect of intensive treatment of type 1 diabetes mellitus on development and progression of diabetic nephropathy: the Epidemiology of Diabetes Interventions and Complications (EDIC) study. JAMA 2003; 290: 2159–67. 52 Haynes R, Lewis D, Emberson J, et al, and the SHARP Collaborative Group, and the SHARP Collaborative Group. Effects of lowering LDL cholesterol on progression of kidney disease. J Am Soc Nephrol 2014; 25: 1825–33. 53 Palmer SC, Navaneethan SD, Craig JC, et al. HMG CoA reductase inhibitors (statins) for people with chronic kidney disease not requiring dialysis. Cochrane Database Syst Rev 2014; 5: CD007784. 54 de Brito-Ashurst I, Varagunam M, Raftery MJ, Yaqoob MM. Bicarbonate supplementation slows progression of CKD and improves nutritional status. J Am Soc Nephrol 2009; 20: 2075–84. 55 Goicoechea M, de Vinuesa SG, Verdalles U, et al. Effect of allopurinol in chronic kidney disease progression and cardiovascular risk. Clin J Am Soc Nephrol 2010; 5: 1388–93. 56 Navarro-González JF, Mora-Fernández C, Muros de Fuentes M, et al. Effect of pentoxifylline on renal function and urinary albumin excretion in patients with diabetic kidney disease: the PREDIAN trial. J Am Soc Nephrol 2015; 26: 220–29.
  • 13. www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 13 Review 57 Fellström BC, Jardine AG, Schmieder RE, et al, for the AURORA Study Group. Rosuvastatin and cardiovascular events in patients undergoing hemodialysis. N Engl J Med 2009; 360: 1395–407. 58 de Zeeuw D, Akizawa T, Audhya P, et al, and the BEACON Trial Investigators. Bardoxolone methyl in type 2 diabetes and stage 4 chronic kidney disease. N Engl J Med 2013; 369: 2492–503. 59 Verbeke F, Lindley E, Van Bortel L, et al. A European Renal Best Practice (ERBP) position statement on the Kidney Disease: Improving Global Outcomes (KDIGO) clinical practice guideline for the management of blood pressure in non-dialysis-dependent chronic kidney disease: an endorsement with some caveats for real-life application. Nephrol Dial Transplant 2014; 29: 490–96. 60 Agarwal R, Peixoto AJ, Santos SF, Zoccali C. Pre- and postdialysis blood pressures are imprecise estimates of interdialytic ambulatory blood pressure. Clin J Am Soc Nephrol 2006; 1: 389–98. 61 Bansal N, McCulloch CE, Rahman M, et al, and the CRIC Study Investigators. Blood pressure and risk of all-cause mortality in advanced chronic kidney disease and hemodialysis: the chronic renal insufficiency cohort study. Hypertension 2015; 65: 93–100. 62 The Blood Pressure Lowering Treatment Trialists’ Collaboration. Blood pressure-lowering treatment based on cardiovascular risk: a meta-analysis of individual patient data. Lancet 2014; 384: 591–98. 63 The SPRINT Research Group. A randomized trial of intensive versus standard blood-pressure control. N Engl J Med 2015; 373: 2103–16. 64 Heerspink HJ, Ninomiya T, Zoungas S, et al. Effect of lowering blood pressure on cardiovascular events and mortality in patients on dialysis: a systematic review and meta-analysis of randomised controlled trials. Lancet 2009; 373: 1009–15. 65 Eldehni MT, Odudu A, McIntyre CW. Randomized clinical trial of dialysate cooling and effects on brain white matter. J Am Soc Nephrol 2015; 26: 957–65. 66 Levin NW, Kotanko P, Eckardt KU, et al. Blood pressure in chronic kidney disease stage 5D-report from a Kidney Disease: Improving Global Outcomes controversies conference. Kidney Int 2010; 77: 273–84. 67 Charra B, Calemard E, Ruffet M, et al. Survival as an index of adequacy of dialysis. Kidney Int 1992; 41: 1286–91. 68 Moen MF, Zhan M, Hsu VD, et al. Frequency of hypoglycemia and its significance in chronic kidney disease. Clin J Am Soc Nephrol 2009; 4: 1121–27. 69 Guideline development Group. Clinical practice guideline on management of patients with diabetes and chronic kidney disease stage 3b or higher (eGFR <45 mL/min). Nephrol Dial Transplant 2015; 30 (suppl 2): ii1–142. 70 Palmer SC, Mavridis D, Navarese E, et al. Comparative efficacy and safety of blood pressure-lowering agents in adults with diabetes and kidney disease: a network meta-analysis. Lancet 2015; 385: 2047–56. 71 Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med 2015; 373: 2117–28. 72 Tonelli M, Wanner C, for the Kidney Disease: Improving Global Outcomes Lipid Guideline Development Work Group Members. Lipid management in chronic kidney disease: synopsis of the Kidney Disease: Improving Global Outcomes 2013 clinical practice guideline. Ann Intern Med 2014; 160: 182–89. 73 Baigent C, Landray MJ, Reith C, et al. The effects of lowering LDL cholesterol with simvastatin plus ezetimibe in patients with chronic kidney disease (Study of Heart and Renal Protection): a randomised placebo-controlled trial. Lancet 2011; 377: 2181–92. 74 Massy ZA, de Zeeuw D. LDL cholesterol in CKD—to treat or not to treat? Kidney Int 2013; 84: 451–56. 75 Winkelmayer WC, Liu J, Kestenbaum B. Comparative effectiveness of calcium-containing phosphate binders in incident U.S. dialysis patients. Clin J Am Soc Nephrol 2011; 6: 175–83. 76 Isakova T, Gutiérrez OM, Chang Y, et al. Phosphorus binders and survival on hemodialysis. J Am Soc Nephrol 2009; 20: 388–96. 77 Zoccali C, Bolignano D, D’Arrigo G, et al, for the European Renal and Cardiovascular Medicine (EURECA-m) Working Group of the European Renal Association–European Dialysis Transplantation Association (ERA-EDTA). Validity of vascular calcification as a screening tool and as a surrogate end point in clinical research. Hypertension 2015; 66: 3–9. 78 Tentori F, Blayney MJ, Albert JM, et al. Mortality risk for dialysis patients with different levels of serum calcium, phosphorus, and PTH: the Dialysis Outcomes and Practice Patterns Study (DOPPS). Am J Kidney Dis 2008; 52: 519–30. 79 Chertow GM, Block GA, Correa-Rotter R, et al, and the EVOLVE Trial Investigators. Effect of cinacalcet on cardiovascular disease in patients undergoing dialysis. N Engl J Med 2012; 367: 2482–94. 80 Palmer SC, Nistor I, Craig JC, et al. Cinacalcet in patients with chronic kidney disease: a cumulative meta-analysis of randomized controlled trials. PLoS Med 2013; 10: e1001436. 81 Liabeuf S, Neirynck N, Drüeke TB, Vanholder R, Massy ZA. Clinical studies and chronic kidney disease: what did we learn recently? Semin Nephrol 2014; 34: 164–79. 82 Fouque D, Kalantar-Zadeh K, Kopple J, et al. A proposed nomenclature and diagnostic criteria for protein-energy wasting in acute and chronic kidney disease. Kidney Int 2008; 73: 391–98. 83 Cano NJ, Fouque D, Roth H, et al, for the French Study Group for Nutrition in Dialysis. Intradialytic parenteral nutrition does not improve survival in malnourished hemodialysis patients: a 2-year multicenter, prospective, randomized study. J Am Soc Nephrol 2007; 18: 2583–91. 84 Sirich TL, Plummer NS, Gardner CD, Hostetter TH, Meyer TW. Effect of increasing dietary fiber on plasma levels of colon-derived solutes in hemodialysis patients. Clin J Am Soc Nephrol 2014; 9: 1603–10. 85 Bliss DZ, Stein TP, Schleifer CR, Settle RG. Supplementation with gum arabic fiber increases fecal nitrogen excretion and lowers serum urea nitrogen concentration in chronic renal failure patients consuming a low-protein diet. Am J Clin Nutr 1996; 63: 392–98. 86 Wang IK, Wu YY, Yang YF, et al. The effect of probiotics on serum levels of cytokine and endotoxin in peritoneal dialysis patients: a randomised, double-blind, placebo-controlled trial. Benef Microbes 2015; 6: 423–30. 87 Guida B, Germanò R, Trio R, et al. Effect of short-term synbiotic treatment on plasma p-cresol levels in patients with chronic renal failure: a randomized clinical trial. Nutr Metab Cardiovasc Dis 2014; 24: 1043–49. 88 Akizawa T, Asano Y, Morita S, et al. Effect of a carbonaceous oral adsorbent on the progression of CKD: a multicenter, randomized, controlled trial. Am J Kidney Dis 2009; 54: 459–67. 89 Ueda H, Shibahara N, Takagi S, Inoue T, Katsuoka Y. AST-120, an oral adsorbent, delays the initiation of dialysis in patients with chronic kidney diseases. Ther Apher Dial 2007; 11: 189–95. 90 Schulman G, Berl T, Beck GJ, et al. Randomized placebo-controlled EPPIC Trials of AST-120 in CKD. J Am Soc Nephrol 2015; 26: 1732–46. 91 Fell LH, Zawada AM, Rogacev KS, Seiler S, Fliser D, Heine GH. Distinct immunologic effects of different intravenous iron preparations on monocytes. Nephrol Dial Transplant 2014; 29: 809–22. 92 Bailie GR, Larkina M, Goodkin DA, et al. Data from the Dialysis Outcomes and Practice Patterns Study validate an association between high intravenous iron doses and mortality. Kidney Int 2015; 87: 162–68. 93 Macdougall IC, Obrador GT. How important is transfusion avoidance in 2013? Nephrol Dial Transplant 2013; 28: 1092–99. 94 Zhang X, Xiang C, Zhou YH, Jiang A, Qin YY, He J. Effect of statins on cardiovascular events in patients with mild to moderate chronic kidney disease: a systematic review and meta-analysis of randomized clinical trials. BMC Cardiovasc Disord 2014; 14: 19. 95 Seliger SL, Zhang AD, Weir MR, et al. Erythropoiesis-stimulating agents increase the risk of acute stroke in patients with chronic kidney disease. Kidney Int 2011; 80: 288–94. 96 Floege J, Bartsch A, Schulze M, Shaldon S, Koch KM, Smeby LC. Clearance and synthesis rates of beta 2-microglobulin in patients undergoing hemodialysis and in normal subjects. J Lab Clin Med 1991; 118: 153–65. 97 Cornelis T, van der Sande FM, Eloot S, et al. Acute hemodynamic response and uremic toxin removal in conventional and extended hemodialysis and hemodiafiltration: a randomized crossover study. Am J Kidney Dis 2014; 64: 247–56. 98 Cornelis T, Eloot S, Vanholder R, et al. Protein-bound uraemic toxins, dicarbonyl stress and advanced glycation end products in conventional and extended haemodialysis and haemodiafiltration. Nephrol Dial Transplant 2015; 30: 1395–402.
  • 14. 14 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 Review 99 Susantitaphong P, Siribamrungwong M, Jaber BL. Convective therapies versus low-flux hemodialysis for chronic kidney failure: a meta-analysis of randomized controlled trials. Nephrol Dial Transplant 2013; 28: 2859–74. 100 Maduell F, Moreso F, Pons M, et al, for the ESHOL Study Group. High-efficiency postdilution online hemodiafiltration reduces all-cause mortality in hemodialysis patients. J Am Soc Nephrol 2013; 24: 487–97. 101 Nistor I, Palmer SC, Craig JC, et al. Convective versus diffusive dialysis therapies for chronic kidney failure: an updated systematic review of randomized controlled trials. Am J Kidney Dis 2014; 63: 954–67. 102 Maduell F, Arias-Guillen M, Fontseré N, et al. Elimination of large uremic toxins by a dialyzer specifically designed for high-volume convective therapies. Blood Purif 2014; 37: 125–30. 103 Böhringer F, Jankowski V, Gajjala PR, Zidek W, Jankowski J. Release of uremic retention solutes from protein binding by hypertonic predilution hemodiafiltration. ASAIO J 2015; 61: 55–60. 104 Budde K, Becker T, Arns W, et al. Everolimus-based, calcineurin-inhibitor-free regimen in recipients of de-novo kidney transplants: an open-label, randomised, controlled trial. Lancet 2011; 377: 837–47. 105 Perrin P, Caillard S, Javier RM, et al. Persistent hyperparathyroidism is a major risk factor for fractures in the five years after kidney transplantation. Am J Transplant 2013; 13: 2653–63. 106 Astor BC, Muth B, Kaufman DB, Pirsch JD, Michael Hofmann R, Djamali A. Serum β2-microglobulin at discharge predicts mortality and graft loss following kidney transplantation. Kidney Int 2013; 84: 810–17.