Vous êtes sur la page 1sur 14

Review

Clinical management of the uraemic syndrome in chronic


kidney disease
Raymond Vanholder, Denis Fouque, Griet Glorieux, Gunnar H Heine, Mehmet Kanbay, Francesca Mallamaci, Ziad A Massy, Alberto Ortiz,
Patrick Rossignol, Andrzej Wiecek, 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, Lancet Diabetes Endocrinol 2016
which progress in parallel to chronic kidney disease. The uraemic syndrome is associated with cardiovascular disease, Published Online
metabolic bone disease, inflammation, protein energy wasting, intestinal dysbiosis, anaemia, and neurological and March 2, 2016
http://dx.doi.org/10.1016/
endocrine dysfunction. In this Review, we summarise specific, modern management options for the uraemic
S2213-8587(16)00033-4
syndrome in chronic kidney disease. Although large randomised controlled trials are scarce, based on data from
Nephrology Section,
randomised controlled trials and observational studies, as well as pathophysiological reasoning, a therapeutic Department of Internal
algorithm can be developed for this complex and multifactorial condition, with interventions targeting several Medicine, Ghent University
modifiable factors simultaneously. Hospital, Ghent, Belgium
(Prof R Vanholder MD,
Prof G Glorieux PhD);
Introduction This fact is largely attributable to the complex and Department of Nephrology-
Kidney failure is associated with deterioration of body multifactorial nature of the disease, which makes it Nutrition-Dialysis, Centre
functions. The clinical picture as a whole—the uraemic difficult to recruit large patient groups with uniform Hospitalier Lyon Sud,
Carmen-CENS, Université
syndrome—is named after urea, the most abundant pathophysiological backgrounds. Additionally, because
Claude Bernard Lyon 1, Lyon,
metabolite retained in kidney failure and the first uraemic of multi-layered pathophysiology, the effect of France; F-CRIN-INI-CRCT,
retention product identified. The uraemic syndrome can therapeutic options that correct one aspect of disease France (Prof D Fouque MD);
be caused by chronic kidney disease or acute kidney (eg, hypercholesterolaemia) can be masked by the effect Department of Internal
Medicine IV, Saarland
injury, and affects almost every organ system (panel 1).1 of other factors (eg, hypertension, fluid overload) on
University Medical Centre,
The syndrome results from the biological effects of outcome measures. Homburg, Germany
metabolites that are not excreted or metabolised by the (Prof G H Heine MD);
kidneys and are retained within the body.2 Such Traditional and non-traditional risk factors Department of Medicine,
Division of Nephrology, Koc
metabolites are named uraemic retention products, or Cardiovascular and non-cardiovascular mortality contri- University School of Medicine,
uraemic toxins if they exert biological or toxic effects. The bute equally to the high mortality seen in people with Istanbul, Turkey
deterioration of renal endocrine function (production of chronic kidney disease.9 Socioeconomic and (Prof M Kanbay MD);
erythropoietin, active vitamin D, or renin), the geographical factors, including access to therapy, Nephrology, Dialysis and
Transplantation Unit, and
deregulation of kidney electrolyte homoeostasis, and explain the variable mortality in chronic kidney disease CNR-IFC Clinical Epidemiology
functional alterations resulting from chronic kidney and end-stage kidney disease populations.10 Cardio- and Pathophysiology of Renal
disease and its causes (eg, diabetes, autoimmune vascular disease in patients with chronic kidney disease Diseases and Hypertension,
disorders) also contribute to the syndrome. The clinical is characterised by immunity-driven inflammatory Ospedali Riuniti, Reggio
Calabria, Italy
picture worsens with kidney failure, with coma and death changes that cause vessel wall stiffening, arteriopathy, (Prof F Mallamaci MD,
(end-stage kidney disease; table 1) the ultimate result if the and cardiomyopathy leading to heart failure, Prof C Zoccali MD); Division of
patient is left untreated. However, since the 1940s, renal arrhythmia, and cardiac arrest. Risk factors span from Nephrology, Ambroise Paré
replacement therapies (dialysis or transplantation) have traditional (Framingham) factors to an expanding list of University Hospital (APHP),
University of Paris Ouest,
extended the life expectancy of patients with this non-traditional risk factors (panel 2).8 Among non- Versailles-Saint-Quentin-en-
potentially fatal condition. cardiovascular causes of death, infection, cancer, Yvelines (UVSQ),
Although dialysis and transplantation extend the life cachexia, suicide, and refusal of treatment account for Boulogne-Billancourt, Paris,
expectancy of patients with uraemia, mortality remains the largest share of fatalities.9 France (Prof Z A Massy MD);
INSERM U1018, Research
substantially higher than in age-matched populations The gap between the predictive value of traditional Centre in Epidemiology and
with normal kidney function;3,4 general and cardiovascular risk factors and real cardiovascular mortality in chronic Population Health (CESP),
mortality tend to rise even before patients need dialysis.5,6 kidney disease11 is to a large extent shown by indicators UVSQ, Villejuif, France
(Prof Z A Massy); Division of
In this Review, we discuss several therapeutic options to of kidney dysfunction, such as estimated glomerular
Nephrology, IIS-Fundacion
treat the consequences of the uraemic syndrome in filtration rate (eGFR) and albuminuria.12 Hence, factors Jimenez Diaz, Madrid, Spain
chronic kidney disease, based on the pathophysiology of related to a decline in kidney function, such as (Prof A Ortiz MD); INSERM
the uraemic syndrome and taking into account newly subclinical volume expansion and uraemic solute Centre d’Investigations
Cliniques (CIC)-1433, and
detected, pathological pathways. retention, might play a part in this process. Many factors
INSERM U1116, Nancy, France
Although we have followed the principles of evidence- that are affected by the uraemic status have been (Prof P Rossignol MD); Institut
based medicine as much as possible in this Review, associated with causes of cardiovascular damage, such Lorrain du Cœur et des
much of the data cited are from observation studies. as inflammation, oxidative stress, macrophage Vaisseaux, CHU Nancy,
Vandoeuvre lès Nancy, France
Randomised controlled trials in kidney disease are infiltration, endothelial dysfunction, thrombogenesis,
(Prof P Rossignol); Université de
scarce,7 and many studies have had negative results.8 arterial calcification, or osteodystrophy.

www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 1


Review

Lorraine, Nancy, France characteristics of these classes and some of the main
(Prof P Rossignol); Association Panel 1: Consequences of the uraemic syndrome compounds in the groups are summarised in the appendix.
Lorraine pour le Traitement de
l’Insuffisance Rénale, Cardiovascular
Vandoeuvre lès Nancy, France Hypertension, fluid overload, cardiac decompensation, Pathophysiology
(Prof P Rossignol); Department vascular damage and stiffness, cardiovascular events, Scope of Review
of Nephrology, Transplantation Since the pathophysiology of the uraemic syndrome
and Internal Medicine, Medical
pericarditis
University of Silesia, Katowice,
affects the function of almost every organ (panel 1), this
Haematological Review is restricted to elements for which relevant
Poland (Prof A Wiecek MD); and
INSERM U970, Hôpital Anaemia, erythrocyte fragility, immune dysfunction information is available and that imply specific therapeutic
Européen Georges Pompidou, (susceptibility to infections, low response to vaccination), approaches. Definition of which pathophysiological events
Paris (Prof G M London MD) inflammation, hypercoagulability, bleeding tendency are directly caused by uraemic retention products and
Correspondence to:
Prof Raymond Vanholder, Endocrine which, as a whole or in part, are caused by other aspects of
Nephrology Section, University Hyperparathyroidism, insulin resistance, impotence, kidney dysfunction is not always easy. We will summarise
Hospital, De Pintelaan infertility, thyroid dysfunction, hyperaldosteronism, growth which elements are linked to uraemic retention and which
185, B9000 Ghent, Belgium ones to kidney dysfunction. Since the links are sometimes
raymond.vanholder@ugent.be
disturbance, adipokine dysbalance, klotho deficiency and
FGF-23 excess, active vitamin D deficiency very complex, we can only focus on what we deem the
See Online for appendix most important aspects.
Osteoarticular problems
Osteomalacia, osteodystrophy, adynamic bone disease, Progression of kidney failure
β2-microglobulin amyloidosis, muscle weakness, fractures, Progression of chronic kidney disease varies depending
bone pain, calciphylaxis and cardiovascular calcification on the underlying cause, disease-specific pathology, and
Neurological predisposing risk factors (figure 1).17 Independent of the
Polyneuropathy, coordination disturbances, tremor, cognitive initial cause, any loss of functional kidney parenchyma
dysfunction, decreased attention span, coma leads to compensatory hyperfiltration and intraglomerular
hypertension of the remaining nephrons, which causes
Gastrointestinal fibrosis and progressive decline in kidney function.
Anorexia, gastroparesis, nausea, vomiting Furthermore, uraemia-specific and non-specific nephro-
Dermatological toxins, inflammation, tissue ischaemia, and pro-coagulant
Skin atrophy, pruritus, calciphylaxis mechanisms damage glomeruli and tubules. Proteinuria
increases the risk of chronic kidney disease progression
Stomatological by causing tubular injury that leads to inflammatory
Periodontitis, stomatitis macrophage infiltration and tubulointerstitial fibrosis.
Nephrological Angiotensin II release increases intraglomerular pressure
Renal tubular damage, progression of kidney failure and oxidative stress, which alters podocyte function and
promotes synthesis of chemokines and cytokines.18
Other Several uraemic retention products have been linked to
Malnutrition, changes in drug protein binding, changes in progression of kidney disease such as indoxyl sulfate,
metabolism, hyperkalaemia, metabolic acidosis p-cresyl sulfate, asymmetrical dimethylarginine (ADMA),
and several cytokines. Factors not linked to uraemic toxins
are exacerbations of primary kidney disease, acute kidney
Uraemic retention products injury, proteinuria, hyperglycaemia, external nephrotoxic
Information about uraemic retention products has agents (radiocontrast, non-steroidals), compensatory
increased continuously in the past few decades. At least hyperfiltration, and ischaemia. Hypertension is a partly
150 uraemic retention products have been described so retention-independent factor—ie, it can be caused by
far,13,14 and with developments in metabolomics and uraemic toxins or by other factors (figure 1).
proteomics in the past decade, each new study has the
potential to add dozens of new substances to this list. Inflammation, metabolic bone disease, and
The idea that removal of one single solute would be cardiovascular disease
sufficient to solve the problem of uraemic toxicity has Non-traditional risk factors, such as low-grade
long since been abandoned. By contrast, the notion that inflammation and oxidative stress play a fundamental part
all these molecules interact is developing.15,16 in chronic kidney disease-associated cardiovascular
In the past 30 years, based on the physicochemical complications, and to a larger extent than in the general
characteristics that affect their elimination during population.19 A key element in this proinflammatory
dialysis (the main removal strategy until now), uraemic status is the activation of the redox-sensitive nuclear
retention products have been divided into three major transcription factor kappa B (NF-κB) (figure 2), in response
categories: small, water-soluble compounds; protein- to several factors including oxidative stress (reactive
bound compounds; and larger, middle molecules.13 The oxygen species [ROS]), mitochondrial dysfunction,

2 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4


Review

GFR* Definition Therapeutic approaches Pathophysiological changes Mortality


Stage 1 >90 Normal kidney function, but Observation, blood pressure and Klotho deficiency Increased overall and
urine abnormalities (eg, cardiovascular risk factor control, cardiovascular
haematuria or prevention of cardiovascular disease mortality +
albuminuria†), proven
structural kidney damage,
or genetic trait
Stage 2 60–89 Mildly reduced kidney Observation, blood pressure and Klotho deficiency, elevated FGF-23 Increased overall and
function associated with cardiovascular risk factor control, cardiovascular
other findings (as in stage 1) prevention of cardiovascular disease mortality +
Stage 3A 45–59; Moderately reduced kidney Observation, blood pressure and Klotho deficiency, elevated FGF-23 Increased overall and
Stage 3B 30–44 function cardiovascular risk factor control, and PTH, malnutrition, hypertension, cardiovascular
prevention of cardiovascular disease, left ventricular hypertrophy, anaemia mortality ++
treatment of anaemia and metabolic bone
disease, prevention of chronic kidney
disease progression
Stage 4 15–29 Severely reduced kidney Planning for renal replacement therapy, Klotho deficiency, elevated FGF-23 Increased overall and
function blood pressure and cardiovascular risk and PTH, malnutrition, hypertension, cardiovascular
factor control, prevention of cardiovascular left ventricular hypertrophy, anaemia, mortality +++
disease, treatment of anaemia and hypertriglyceridaemia,
metabolic bone disease, prevention of hyperphosphataemia, metabolic
chronic kidney disease progression acidosis, hyperkalaemia
Stage 5 <15 Very severe kidney failure or Renal replacement therapy or conservative Klotho deficiency, elevated FGF-23 Increased overall and
end-stage kidney disease approach, blood pressure and and PTH, malnutrition, hypertension, cardiovascular
cardiovascular risk factor control, left ventricular hypertrophy, anaemia, mortality ++++
prevention of cardiovascular disease, hypertriglyceridaemia,
treatment of anaemia and metabolic bone hyperphosphataemia, metabolic
disease acidosis, hyperkalaemia, need for
renal replacement therapy

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

several uraemic retention products, infection, vasoactive Cardiac and arterial complications are the principal
substances, dyslipidaemia, and inflammation. Activation complications of the uraemic syndrome. The
of NF-κB is associated with the release of proinflammatory cardiomyopathy in chronic kidney disease is characterised
cytokines (tumour necrosis factor α, interleukin 1β, by left ventricular hypertrophy, capillary rarefaction, and
interleukin 6, soluble tumour necrosis factor-like weak interstitial fibrosis, with predominant diastolic dys-
inducer of apoptosis) and activation of monocyte function. Arrhythmias, sudden death, and heart failure
chemoattractant protein-1 and profibrotic transforming are the main consequences of these cardiac com-
growth factor β1. plications. NF-κB activation by vasoactive substances
Inflammation and oxidative stress are tightly affects cardiomyocyte growth, fibrosis, and apoptosis,
interconnected. NF-κB activation initiates production of inducing hypertrophy.19
ROS and vice versa, but under normal conditions, In addition to the proinflammatory factors mentioned
nuclear factor-erythroid-2-related factor 2 (a regulator of above, some uraemic toxins and several factors that are
resistance to oxidants via expression of antioxidants independent of uraemic toxins are damaging for heart
and cytoprotective agents) is also activated. This balance and vessels: indoxyl sulfate, ADMA, endothelin, oxalate
might be disrupted in chronic kidney disease.20 (uraemic retention products), hypertension, sodium and
A substantial number of uraemic toxins (eg, indoxyl fluid retention, primary diseases (eg, amyloidosis),
sulfate, p-cresyl sulfate, the cytokines, advanced glycation infections (endocarditis).
end products, trimethylamine N-oxide) are pro- Compared with the general population, arterial disease
inflammatory, as are compounds that until 5 years ago in populations with chronic kidney disease is premature,
were thought to be inert, like urea (for a more and characterised by endothelial dysfunction, arterial
comprehensive list, see appendix). Furthermore, several remodelling with arterial calcifications (figure 3A, 3B),
other factors that are inflammatory are partly or entirely vascular stiffness, and the transition of the vascular
unrelated from uraemic retention products: infection, smooth muscle cells to an osseous phenotype.21 Vascular
dialysis fluid contamination, dialysis bioincompatibility, stiffness, which is closely linked to endothelial
concomitant diseases (eg, autoimmune disorders), and dysfunction, precedes arterial calcifications that further
vasoactive agents (figure 2). exacerbate stiffening.22 Arterial calcifications are due to an

www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 3


Review

osteochondrogenic phenotype (Runx2, osterix, Sox9).24


Panel 2: Non-traditional risk factors in the uraemic syndrome5 Several chronic kidney disease-linked and biochemical
• Anaemia changes related to mineral bone disorder (figure 3C), such
• Volume overload as hyperphosphataemia, are associated with arterial
• Metabolic bone disorder and related mediators calcifications. Hyperphosphataemia also stimulates the
• Hyperphosphataemia production of ROS and thereby activation of NF-κB. The
• High levels of FGF-23 resultant inflammation decreases the concentration of
• Low expression of the anti-ageing factor α-klotho calcification inhibitors and reduces klotho expression,
• Secondary hyperparathyroidism which induces resistance to the phosphaturic effect
• Active vitamin D deficiency of fibroblast growth factor 23 (FGF-23),25 bone resistance
• Low grade inflammation to parathyroid hormone (PTH), secondary hyperpara-
• Oxidative stress thyroidism,26 and accelerated ageing.27 Telomere
• Uraemic retention products* shortening, which is typical for ageing, is associated with
• Post-translational protein modifications cardiovascular disease manifestations.28 Inflammatory
• Accumulation of atherogenic remnant lipoproteins epigenetic changes, which, in part, involve inhibited gene
• Endogenous nitric oxide synthase (NOS) inhibitors methylation of s-adenosinemethionine, also contribute to
• High sympathetic activity inflammation and cardiovascular lesions.29,30
• Insulin resistance Uraemic bone disease is mainly triggered by increased
• Exposure to bioincompatible dialysis conditions concentrations of the uraemic toxin phosphate, which
• Immune response activating membranes results in the enhanced generation of several
• Contaminated haemodialysate compensatory factors to maintain serum phosphate con-
• Peritoneal dialysate containing glycation products centration, such as FGF-23 and PTH, at the cost of loss of
• Haemodynamic instability during haemodialysis bone structure and function. Independent of metabolite
• Infections retention, ageing, inflammation, bone ischaemia, and
• General inadequate renal production of active vitamin D
• Vascular access: arteriovenous fistula, catheters, and arteriovenous grafts metabolites further enhance bone degradation. Some
• Dyskalaemia (hypokalaemia and hyperkalaemia) patients develop adynamic bone disease, whereby low
PTH hinders calcium deposition in the bone, favouring
*For details, see appendix.
vascular calcification. In rare cases, patients develop
calciphylaxis—a severe syndrome consisting of vascular
calcification, thrombosis, and skin necrosis.
↑ Angiotensin II In summary, inflammation, oxidative stress, and bone
↑ Catecholamines
↑ Endothelin
disease show specific features linked to chronic kidney
disease and uraemia resulting in accelerated vascular
damage and ageing.
Glomerular hypertension
and hyperfiltration
Malnutrition and dysbiosis
Malnutrition and dysbiosis are two features of the
Chronic Podocyte injury and Glomerular and Progressive kidney uraemic syndrome that relate to digestive function
kidney proteinuria tubulo-interstitial dysfunction
disease fibrosis
(figure 4). Some uraemic retention products (such as
cytokines leptin, ghrelin, and neuropeptide Y) can be
implicated in reduced appetite and nutrient intake.
Inflammation p-cresyl sulfate causes insulin resistance, disturbances of
lipid metabolism, and aberrant distribution of fat cells
Uraemic retention products throughout the body. Inflammation—a common epi-
phenomenon of chronic kidney disease—is associated
Figure 1: Role of progression of kidney failure in defining the uraemic syndrome 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
imbalance of calcification inducers and inhibitors, and of solute retention, are changes in taste and smell
development of a senescence-associated secretory perception, reduced intestinal motility and absorption
phenotype (SASP) in vascular smooth muscle cells.23 capacity, depression, and loss of aminoacids and protein
Inducers of arterial calcifications and SASP are all linked via dialysis (figure 4). Consequently, patients might have a
to secretion of proinflammatory cytokines. Inflammation spontaneous reduction in energy intake, which can
activates the BMP2:BMP4, Msx2, and Wnt pathways, induce protein energy wasting even before starting
which promote increased transcription of genes associated dialysis (figure 3D). Although decreased protein intake
with conversion of vascular smooth muscle cells to the could reduce generation of uraemic retention products,

4 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4


Review

ROS Uraemic retention Infectious agents Vasoactive agents Mitochondrial Dialysis-related Dyslipidaemia
products Infection, Angiotensin II, dysfunction factors OxLDL
AGEs, ADMA, IS, pCS, endotoxin (LPS), noradrenaline, Bioincompatibility,
TMAO, phosphate bacterial DNA, endothelin I, dialysis fluid impurities
peptidoglycan aldosterone

↑NF-κB

Cytokines and chemokines


Proinflammatory (eg, interleukin 6, MCP-1),
profibrotic (eg, TGF-β)

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.

A B C D

E F G

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).

muscle protein catabolism, indirectly caused by acidosis uraemic retention cause structural and functional
and inflammation, might enhance the accumulation of changes of the intestinal epithelial barrier, inducing
toxic metabolites. Thus, a diet containing the appropriate inflammation via different pathways.33 Although
nutrients in the appropriate quantity and balance is progression of kidney disease is clearly linked to changes
crucial for wellbeing and to reduce metabolic poisoning in intestinal microbiota, whether or not uraemic
in patients with chronic kidney disease. retention products play a part in these changes is not
The intestinal microbiota plays an important part in clear. Other conditions that are associated with kidney
regulating the host’s nutritional status, metabolism, and disease could also affect intestinal microbiota—eg,
different aspects of immunity. Dysbiosis has been linked ageing, diabetes, obesity, changes in nutrient intake and
to several chronic diseases. The few studies of dysbiosis diet, and antibiotic intake.
in patients with chronic kidney disease suggest changes
in the abundance and metabolic characteristics of Anaemia
intestinal microbiota as compared with healthy Kidney dysfunction is associated with decreased
individuals.32 These changes induce the preferential and concentrations of haemoglobin in blood, mainly due to
abundant generation of uraemic retention products with deficiency of and resistance to erythropoietin and absolute
toxic effects. Additionally, chronic kidney disease and and relative iron deficiency (figure 5). The kidneys

www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 5


Review

microbleeds; figure 3E–G) and cognitive disorders


↓ Nutritional status (linked to vascular abnormalities, neurodegenerative
abnormalities, or both; figure 6) have been reported in
Chronic kidney Uraemic ↑ Inflammation Protein energy wasting
chronic kidney disease.35 Cerebrovascular disorders
disease retention products could be linked to traditional and non-traditional
cardiovascular risk factors, such as oxidative stress,
chronic inflammation, endothelial dysfunction, vascular
Intestinal modifications
and translocation calcification, anaemia, dialysis-related haemodynamic
instability, and uraemic retention products.36 Uraemic
Figure 4: Role of malnutrition and dysbiosis and chronic kidney disease in defining the uraemic syndrome toxins could affect cerebrovascular diseases or cognition
either by direct functional effects or by modulation of
other factors (such as inflammation and oxidative
↓ Erythropoietin Anaemia Deficiency of folic acid stress). β2-microglobulin—a typical uraemic toxin—
generation and vitamin B12, was shown to impair cognitive function and act as a
erythrocyte fragility
systemic, pro-ageing factor.37 Other uraemic retention
Chronic kidney disease
products frequently associated with neurological
↑ Hepcidin Iron deficiency dysfunction are guanidines. Phenols and indoles have
production
also been linked to neurological disturbances. Further
contributing factors that are partly linked to retention
Inflammation, Deficient iron are inflammation, vascular disease, and hypertension.
infection intake, blood loss Factors that are more or less independent of solute
retention are depression, enhanced activity of drugs that
Figure 5: Role of renal anaemia and chronic kidney disease in defining the uraemic syndrome treat psychiatric disorders or have psychological effects
(eg, sleeping pills), and blood pressure changes related
or not to dialysis (figure 6).
Cognitive
dysfunction
Management
Oxidative stress, inflammation,
endothelial dysfunction, The studies, guidelines, position statements, and reviews
Chronic kidney Uraemic retention Neurological
disease products
vascular calcification,
damage referred to in this section are summarised in the
dialysis-related haemodynamic
instability
appendix. Treatment aimed at modifying risk factors will
be discussed in broad terms independently of underlying
Cerebrovascular intermediate mechanisms. Of note, positive interventions
disease
in the general population, could counterintuitively cause
or amplify complications in chronic kidney disease. This
Figure 6: Role of neurological dysfunction and chronic kidney disease in defining the uraemic syndrome
section on management includes 56 original studies
(appendix), of which only 35 (63%) are randomised
respond to low tissue oxygen by increasing erythropoietin controlled trials or meta-analyses. Only six (11%) of these
production, which induces erythropoiesis. Decline in randomised controlled trials or meta-analyses have an
kidney function impedes this mechanism. Iron deficiency unambiguously positive intervention in a study taking
is primarily attributable to ineffective intestinal iron into account more than 1000 patients (all but one of these
absorption, because inflammation associated with six are meta-analyses).
chronic kidney disease elevates hepcidin, an inhibitor of
iron export from enterocytes to plasma. Insufficient oral Lifestyle
iron intake and blood loss from occult gastrointestinal or Emphasis is often laid on pharmacological drug
urogenital bleeding, repetitive blood sampling, and interventions in cardiovascular and kidney protection in
dialysis further drive iron deficiency. Finally, vitamin B12 chronic kidney disease, of which the benefit is not
and folate deficiency, shortened erythrocyte lifespan, and necessarily unequivocal. Several general lifestyle
frequent infections are also contributing factors.34 measures could also be beneficial, and need negligible
Erythropoietin and iron deficiencies are the main societal investment, except for educational initiatives.
drivers of renal anaemia, and these effects are largely In a large meta-analysis, regular exercise training was
independent of uraemic retention; however, uraemic associated with improved outcomes in chronic kidney
toxins can affect erythrocyte production, solidity, and disease. However, risk of bias of the included studies was
survival (figure 5). high.38 In a single-centre, observational study, heavy
smoking was associated with increased risk of
Neurological dysfunction progression of chronic kidney disease, especially in
High incidence of cerebrovascular diseases (such as patients with hypertensive and diabetic nephropathy.39 In
stroke, white matter lesions, and intracerebral a large, cross-sectional follow-up study, no significant

6 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4


Review

association was recorded between pre-dialysis obesity increased to more than 1·0 g/kg per day of protein in
and progression of chronic kidney disease.40 Another patients on haemodialysis and 1·2 g/kg per day in
cohort study,41 however, showed a clear association peritoneal dialysis (table 2).45
between BMI more than 30 kg/m² and progression of In stable patients, protein intake can be calculated as
kidney disease, especially from the age of 40 years on. protein catabolic rate from pre-dialysis and post-dialysis
The relative survival advantage in patients who are urea measurements. Fluid intake is an indicator of
overweight and on dialysis (obesity paradox) is nutritional status. In patients with anuria, interdialytic
remarkable, but can be attributed to poor outcomes of weight gain was positively associated with protein catabolic
their malnourished counterparts,42 partly related to the rate.46 Reduced weight gain, or a low serum urea or
negative effects of inflammation. Unfortunately, focused creatinine pre-dialysis are negative signs and should prompt
and conclusive studies on lifestyle in chronic kidney rapid dietary intervention.47 Nevertheless, fluid intake that is
disease are scarce. too high and interdialytic weight gain increase intradialytic
ultrafiltration rate, which should be avoided because of its
Diet association with mortality in patients on dialysis.48
A key factor of the dietary management of the uraemic Dietary phosphate intake should be controlled without
syndrome is to supply enough nutrition, but not too causing a reduction in protein intake, because a reduction
much, because the body needs fuel, but cannot handle in protein intake is associated with worse survival.49 Foods
well enough the end products of cellular metabolism. and beverages rich in phosphate, such as preserves,
This notion is particularly true of proteins because they processed meat, frozen foods, dairy products, and soft
cannot be stored to adjust for intake fluctuations. drinks should be discouraged. Potassium intake that is too
Accumulation of uraemic retention products and high should also be avoided (table 2). For more detailed
metabolic poisoning in chronic kidney disease can be dietary advice see appendix.
ameliorated by reduced protein intake, which, rather
than being left to spontaneous evolution, should be Pharmacological treatment
prescribed as a well balanced, carefully controlled, low- Chronic kidney disease is a challenging disease with
protein diet. Thus, patients with stages 3–5 chronic few treatment options to prevent progression. In
kidney disease who are not yet on dialysis should be clinical practice, measurements of albumin-to-
educated to pursue an equilibrium between sufficient creatinine ratio and eGFR allow risk classification for
energy intake (at least 30 kcal/kg per day) and reduced chronic kidney disease progression and cardiovascular
protein intake (0·6 to 0·8 g/kg per day, 50% of which mortality. Additional biomarkers might be helpful in
should be high value proteins of animal origin containing refining these strategies, but their use needs further
essential aminoacids (table 2).43 This approach reportedly validation.
reduces serum urea by 30%, and improves insulin
resistance, phosphate and parathyroid metabolism, Chronic Receiving Receiving Had Had
blood pressure, and anaemia.43 Reduction of phosphorus kidney haemodialysis peritoneal transplant transplant
intake allows better control of mineral bone disease and disease dialysis <3 months ≥3 months
stages 3–5, ago ago
reduces the need for oral phosphate binders. Sodium not on
intake should be targeted to a maximum of 6 g sodium dialysis
chloride per day and, if possible, lower (table 2), except in Diet Low protein Standard Standard High protein Low protein
the case of salt-losing nephropathy, in which intake can protein protein
be higher. Specialised dietitians should regularly be Target protein intake 0·6–0·8 1·0–1·2 1·2 1·4 0·6–0·8
involved in the implementation of advice to correctly (g/kg per day) (or less with
educate the patient. At least three encounters per year keto-
analogues)
have been suggested for the first year of care.44 This only
Target energy intake 30–35 30–35 30–35 30–35 30–35
seems practical from chronic kidney disease stage 4 (kcal/kg per day)
onwards. 24 h urine collections allow the monitoring of Salt intake (mg per <6000 <5000 <5000 <6000 <6000
daily protein intake (via urea measurements) and sodium day)*
intake and should be done twice yearly to control and Potassium intake 2500 2500 2500 Free Free†
implement the diet. If intentional weight loss is planned, (mg per day)
the benefit should be balanced against the risk of protein Phosphorus intake <800 <1000 <1000 Free <800
(mg per day)
wasting, particularly in the late stages of chronic kidney
disease. Wasting risk ++ +++ +++ +++ +

On dialysis, additional catabolism takes place in Overweight risk ++ + + ++ +++


response to chronic inflammation, surgical interventions, *Except in case of salt-losing nephropathy. †Except in case of hyperkalaemia (>5·5 mmol/L). Risk of being overweight
recurrent sepsis, and nutrient losses during the dialysis indicated as mild (+), moderate (++), and severe (+++).
procedure. Whereas caloric intake can be maintained
Table 2: Integrated optimum nutrition of patients with chronic kidney disease
above 30 kcal/kg per day, protein intake should be

www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 7


Review

The mainstays of non-specific prevention of chronic pressure burden in patients on haemodialysis.60 Out-of-
kidney disease progression, irrespective of cause, include dialysis systolic blood pressure, in fact, predicts a linear
blood pressure control and proteinuria-directed strategies increase in the risk of death from 110 mm Hg or higher,61
to preserve residual kidney function, with special emphasis as in the general population.
on angiotensin-converting enzyme (ACE) inhibitors or Antihypertensive treatment reduces the incidence of
angiotensin-receptor blockers.50 In diabetic nephropathy, death and cardiovascular events at all risk levels and in
strategies that focus on strict glycaemic control slow down absolute terms most of all in patients with a higher
progression.51 Although hyperlipidaemia could play a role baseline risk.62 In the SPRINT trial,63 patients with
in progression, there is no consensus regarding statin hypertension randomised to target systolic blood
treatment for nephroprotection.52,53 pressures of less than 120 mm Hg had fewer
In a randomised controlled trial54 of bicarbonate cardiovascular events than in those targeted to less than
supplementation with a target serum bicarbonate 140 mm Hg, in general and in the subgroup with chronic
concentration greater than 23 mmol/L, progression of kidney disease. In patients with end-stage kidney disease,
chronic kidney disease was slowed and nutritional status including patients with heart failure, the use of
improved. In another randomised controlled trial,55 antihypertensive drugs reduces mortality,64 but treatment
reduction of uric acid by allopurinol treatment was should be titrated to tolerable levels—ie, to minimise the
nephroprotective. In a trial56 in patients with diabetic risk of hypotension by autonomic dysfunction or arterial
nephropathy, pentoxifylline reduced the rate of decline in stiffness,65 which enhance the risk for ischaemic events.
eGFR and proteinuria. The findings of these three Optimisation of volume control at constant ultrafiltration
studies should, however, be confirmed in larger rate (eg, by more frequent or extended dialysis), and
controlled trials. judicious use of antihypertensive drugs accounting for
Uraemia is a strong, independent risk factor for comorbidities and pharmacokinetic profile66 could reduce
cardiovascular disease, therefore, patients with chronic cardiovascular risk in patients on dialysis. In a single
kidney disease need close cardiovascular follow-up as centre-based observational study,67 extended haemo-
part of their management strategy to prevent chronic dialysis (24 h per week) in an older population resulted in
kidney disease progression. good blood pressure control without antihypertensive
Approaches targeting inflammation (eg, anti- drugs.
inflammatory drugs or statins) did not provide health Controlling hyperglycaemia in patients with renal
benefits for patients with chronic kidney disease. In the failure and diabetes is difficult because of the higher risk
AURORA trial,57 rosuvastatin lowered serum C-reactive of hypoglycaemia as compared with the diabetic
protein concentrations by 27%, but failed to reduce population without chronic kidney disease.68 Target
mortality or the risk of cardiovascular events. Resistance HbA1c levels should account for additional risk factors,
to interventions targeting inflammation could depend with the aim to achieve strict control in those with low
on the severity of inflammation in end-stage kidney risk, but more leniency in those with comorbidities and
disease (in the AURORA trial,57 median C-reactive high risk for hypoglycaemia.69 Patients with diabetes,
protein in the treatment group remained four times chronic kidney disease, and heart failure, ischaemic
above upper normal concentrations), and on its heart disease, or hypertension, can be treated with renin–
multifactorial origin. Interference of proinflammatory angiotensin system blockers at a maximally tolerated
and oxidative mechanisms—by the reduction of NF-κB dose. However, combinations of ACE inhibitors and
activation and activation of nuclear factor-erythroid-2- angiotensin II receptor blockers should be used carefully
related factor 2—seemed an attractive option to reduce in patients with chronic kidney disease not yet on
the enhanced risk of all-cause and cardiovascular death dialysis69 to avoid hyperkalaemia, kidney impairment,
in advanced chronic kidney disease. However, a trial58 and hypotensive symptoms,50 although in a meta-
testing a NF-κB blocker (bardoxolone) in patients with analysis,70 a positive effect on kidney function
chronic kidney disease and type 2 diabetes was preservation was suggested with this combination in
prematurely terminated because of excessive risk of people with diabetes.
cardiovascular events in the bardoxolone group. Empagliflozin, a sodium-glucose co-transporter
In pre-dialysis chronic kidney disease, the well inhibitor, significantly reduced a composite of cardio-
established cardiovascular benefit of antihypertensive vascular events and death in patients with diabetes type 2
treatment has led to specific guidelines recommending and eGFR more than 30 mL/min per 1·73 m² at high
target blood pressures of 140/90 mm Hg or less in cardiovascular risk (26% with chronic kidney disease
chronic kidney disease (130/80 mm Hg or less in the stage 3).71
presence of proteinuria).59 The relation between pre- Because of malnutrition and inflammation, hyper-
dialysis or post-dialysis blood pressure and mortality in cholesterolaemia often regresses as chronic kidney
patients on dialysis is inverse or U-shaped, which is a disease advances. Identification of dyslipidaemia (high
classical example of reverse causality. However, dialysis- total or LDL cholesterol, low HDL cholesterol, high
related blood pressure values hardly reflect true blood triglycerides) in patients with end-stage kidney disease is

8 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4


Review

deemed useful by existing Kidney Disease Improving laboratory, and body composition measurements.
Global Outcomes (KDIGO) guidelines to assess overall Wasting scores can predict survival and should be used
cardiovascular risk,72 but follow-up of lipid concentrations more frequently to help identify patients at risk of death
is not recommended because of the absence of evidence from wasting. An increase in nutrient intake is the key
that this approach improves clinical outcomes. intervention and has been shown to be effective even in
Findings from the SHARP trial73 and subsequent the context of chronic inflammation. One or two units of
meta-analysis53 showed the usefulness of an absolute oral supplements taken separately from regular meals to
reduction of LDL cholesterol in patients with chronic avoid further reductions in spontaneous intake should be
kidney disease to decrease the associated cardiovascular used as a primary measure. The FINE study,83 which
risk. Cholesterol-lowering therapy is especially compared oral nutritional supplements with or without
recommended by KDIGO in all patients with chronic parenteral nutrition in dialysis patients with protein
kidney disease at stage 3–5, who are more than 50 years energy wasting, was negative for its primary endpoints
of age and not on dialysis.72 However, the benefit of (eg, mortality). Oral supplements were well tolerated and
statins was less prominent in end-stage kidney increase total nutrient intake, serum albumin, and
disease,57,74 which discouraged their use in end-stage serum prealbumin.83 Results of a multivariate analysis
kidney disease, irrespective of inflammation or showed that mortality was reduced if serum prealbumin
malnutrition. In patients already treated, however, this increased above 30 mg/L in the first 3 months of
therapy can be continued.69 supplementation.83 Therefore, in ambulatory patients, if
Results from a large end-stage kidney disease cohort in nutritional status does not improve within 4 weeks,
the USA showed no survival benefit of lowering enteral feeding should be considered. For hospital
phosphate concentrations with calcium carbonate or inpatients, intensive nutrition through a nasogastric tube
acetate.75 Findings from another observational study76 or percutaneous gastrostomy can be considered if oral
showed a reduction in death risk in patients who were energy intake is less than 20 kcal/kg per day for more
given any type of phosphate binder. Results from a meta- than 10 days.
analysis77 of seven trials in haemodialysis patients and Few randomised controlled trials have been done that
one in moderate chronic kidney disease showed lower investigate the effect of the restoration of intestinal
mortality in patients given the non-calcium-based symbiosis in chronic kidney disease by giving patients
phosphate binder sevelamer. However, this apparent prebiotics (selectively fermentable ingredients),
benefit (heterogeneity I²=89%) is potentially skewed by a probiotics (live biotherapeutics), or synbiotics (com-
study in patients on haemodialysis in which sevelamer bination of prebiotics and probiotics) to reduce
reduced the death risk by 91%, hence, the issue remains circulating concentrations of uraemic retention products,
unresolved. Serum phosphate concentrations between inflammation, oxidative stress, and progression of
0·68 and 1·93 mmol/L do not associate with an excessive chronic kidney disease. Specific prebiotics decrease
death risk in dialysis patients.78 serum indoxyl sulfate and urea nitrogen.84,85 Results from
The decision to treat hyperphosphataemia should take a study86 in patients receiving peritoneal dialysis showed
into account serum calcium, PTH, and serum vitamin D a significant reduction in concentrations of pro-
concentrations from the early stages of chronic kidney inflammatory cytokines, an increase in concentrations of
disease-associated mineral bone disorder.78 Secondary anti-inflammatory cytokines, and better preservation of
hyperparathyroidism has been implicated in the residual kidney function in the group receiving
cardiovascular risk of chronic kidney disease. However, probiotics. Synbiotic therapy reduces the concentration
improvement of mortality and cardiovascular outcome of p-cresol.87 In several of the above trials, however, the
was inconclusive in a large trial79 of calcium receptor intervention was coupled to a low-protein diet, which
agonist cinacalcet in secondary hyperparathyroidism, and could itself decrease uremic toxin concentration and
this finding was confirmed by meta-analysis.80 progression of kidney failure. The actual effect of
Many patients with chronic kidney disease have low restoring intestinal symbiosis on the intestinal
serum concentrations of 25-hydroxyvitamin D or microbiota profile or on hard outcomes was not assessed
calcitriol, but a randomised, placebo-controlled trial in any of the above trials.
testing their supplementation on hard endpoints is not Reduction of indoxyl sulfate by intestinal sorbents, such
yet available. However, these supplements might help as AST-120, stopped progression of chronic kidney
control hyperparathyroidism. The effect of modulation of disease in small Japanese randomised trials.88,89 This
uraemic retention product concentrations on metabolic benefit was not confirmed in a large European–American
bone disease and cardiovascular outcomes needs to be randomised controlled trial.90 Interventions improving
investigated because several of these retention products symbiosis need further study before their efficacy can be
are associated with both complications.81 accepted.
Malnutrition should be diagnosed using the The conventional approaches for iron repletion in
International Society for Renal Nutrition and Metabolism chronic kidney disease are oral ferrous salts or intravenous
protein energy wasting criteria,82 which include clinical, colloidal compounds. High concentrations of hepcidin,

www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 9


Review

however, make intestinal iron uptake ineffective. interventional trials have targeted these abnormalities.
Additionally, oral iron intake further contributes to the However, improvement of intra-dialytic haemodynamic
pill burden of patients with chronic kidney disease and stability by cooling dialysate has been shown to protect
causes severe gastrointestinal side-effects. Therefore, against brain injury.65
intravenous iron has gained popularity in the past 5 years.
However, experimental and observational data suggest Renal replacement therapy
unwanted immunological, cardiovascular, and renal Replacement of kidney function requires trans-
effects of such treatment,91,92 which might differ between plantation or dialysis, and haemodialysis and peritoneal
classic preparations and new, more stable formulations.91 dialysis are the main dialysis modalities (for more on
Long-term prospective studies are, however, unavailable. different dialysis strategies see appendix). Haemodialysis
The introduction of recombinant human erythropoietin is usually done three times weekly, whereas peritoneal
was one of the major steps forward in the treatment of dialysis clears retention products with lower efficiency
renal anaemia. Both short-acting and long-acting but does so continuously. The high efficiency solute
erythropoiesis stimulating agents are in use at present. removal provided by a single haemodialysis session over
According to the existing guidelines, the decision of 4 h should be extrapolated to the full interdialytic interval
whether and when to start treatment with an of 48–72 h, by comparison with kidney removal capacity.
erythropoiesis stimulating agent should be individualised Thus, renal replacement therapy through dialysis allows
and take into account related risks and anaemic the anuric patient to have a kidney-equivalent filtration
symptoms.34 The target values for erythropoiesis rate of only 5–10 mL/min for small molecules that are
stimulating agent therapy are 10·0–12·0 g/dL.34 These the size of urea or creatinine, which results in higher
drugs should be used with care in patients at high risk of uraemic solute concentrations than is normal. In
stroke or with active or past malignancy.34 general, dialysis removes retention products of higher
Blood transfusions might be considered, especially in molecular weight or those bound to protein less
patients with haemoglobin concentration of less than efficiently than the normal kidney. Oral phosphate
7·0 g/dL, who are resistant to erythropoiesis-stimulating binders are frequently needed to maintain serum
agents, or at potential risk of complications with phosphate concentrations that are close to normal. The
erythropoiesis stimulating agent therapy.93 However, the standard dialysis clearance of small proteins such as
risk of developing panel reactive antibodies should be cytokines, FGF-23, and β2-microglobulin is negligible,
taken into account in transplant candidates. as compared with normal kidneys. Clearance of
Few data are available on the effect of treatment for β2-microglobulin (11·8 kDa), deemed representative of
chronic kidney disease on risk of stroke and other middle-sized molecules, was estimated at 3–19 mL/min
neurological disorders in patients with this disease, for a conventional haemodialysis session (ie,
particularly in those with end-stage kidney disease. 0·17–1·0 mL/min when averaged over 72 h), as compared
Patients with chronic kidney disease have been under- with 65 mL/min in a normally functioning kidney.96
represented in the cardiovascular trials that prove net Conventional haemodialysis relies on diffusion for
benefit of commonly used preventive treatments (eg, solute removal, whereas haemodiafiltration also
antihypertensive drugs, low-dose aspirin, carotid revascu- depends on convective transport. This additional
larisation, and thromboprophylaxis for atrial fibrillation), mechanism enhances the removal of middle-sized
and safety and efficacy of many of these treatments in molecules, such as β2-microglobulin and FGF-23, but
chronic kidney disease remains uncertain. Conflicting has little effect on many protein-bound retention
results have been reported on the effect of statins in products.97,98 However, β2-microglobulin clearance per
patients with chronic kidney disease. In a Cochrane single haemodiafiltration session is still far from
review,53 the authors concluded that statins did not normal kidney removal rates when averaged over
necessarily have effects on stroke in patients with chronic 48–72 h (3·6 mL/min higher than conventional
kidney disease who did not need dialysis, by contrast haemodialysis).99 Despite this poor efficiency, findings
with another meta-analysis94 in which statins were from a randomised controlled trial100 published in 2013,
associated with a decrease in cardiovascular disease in suggested that haemodiafiltration could improve hard
chronic kidney disease, including stroke. Correcting outcomes versus standard dialysis, but a meta-analysis
anaemia might not prevent stroke and could even of all studies showed improvement only for cardio-
increase its risk in chronic kidney disease.95 Moreover, vascular mortality, but not overall mortality.
the effect of anaemia treatment on cognitive disorders Additionally, the reliability of potential benefits found
remains a matter of debate. in this meta-analysis was deemed debatable because
Oxidative stress, inflammation, or uraemic retention the studies included in the analysis had several
products could contribute to neurological disorders in methodological limitations.101
patients with chronic kidney disease. Thus, treatments Dialyser design changes might further improve removal
modulating these factors might improve neurological of middle molecules.102 However, for each additional
outcomes. To the best of our knowledge, no direct 20 mg of β2-microglobulin removed in a single session

10 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4


Review

cannot always be extrapolated from the general


Search strategy and selection criteria population, because beneficial interventions in the
We searched PubMed, Embase, Web of Science, and the general population often have a different effect in
Cochrane Library for articles published in English up to Dec 1, populations with chronic kidney disease. Treatment of
2015. Search terms related to the main topics of this Review one aspect of the uraemic syndrome might exacerbate
were used—eg, “metabolic bone disease” or “inflammation” other deleterious elements. In future studies, a more
and “chronic kidney disease”, “chronic renal failure” or “chronic holistic therapeutic approach to cope with the high
kidney disease” and “physiopathology”, “treatment”, “therapy”, mortality of this disease could be more useful rather
“outcomes”, “morbidity”, or “mortality”. Randomised than the pursuit of one single factor as is usual practice
controlled trials or large observational studies on hard in randomised controlled trials. Therapeutic approaches
outcomes were retained by preference. Reference lists of the should focus on additional outcomes beyond mortality,
resulting articles were searched for other relevant papers. especially standardised quality of life. Consultation with
Review articles are cited to provide readers with more details patients to understand what is important to them might
and more references than space permits in this narrative be useful for the definition of patient-related outcomes.
review, and were selected for their comprehensiveness and Contributors
compactness. 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.

(less than 10% increase), 1·24 g of albumin were lost (40% Declaration of interests
RV has received travel grants and honoraria from Nipro, Bayer, and
increase).102 Researchers who aim to increase protein- Fresenius Medical Care, and has acted as a consultant for Fresenius
bound solute removal are exploring how to increase the Medical Care and Debiotech. DF has acted as a consultant for Fresenius
release of uraemic retention products from their protein- Kabi. GHH has received study grants from Pharmacosmos and
bound states,103 for example by including adsorbents, but Fresenius Medical Care. ZAM has received speaker’s honoraria and
research grants from Amgen, Genzyme, Fresenius Medical Care, and
length of dialysis using these technologies is a limiting Shire. AO has received speaker’s honoraria and research grants from
factor.97 Extension of haemodialysis time, in daily or Amgen, Genzyme, Fresenius Medical Care, Servier, and Shire. PR has
extended regimens, either at home or in centre, can received honoraria from Baxter-Gambro, Fresenius, and Relypsa. AW has
improve weekly solute clearance.97,98 received speaker’s and consulting honoraria from Amgen, Fresenius
Medical Care, Astellas, Roche, GlaxoSmithKline, Pharmacosmos, and
Transplanted kidneys develop hypertrophy and mean Teva. GG, MK, FM, CZ, and GML declare no competing interests.
eGFR is 60–70 mL/min per 1·73 m² 1 year after
Acknowledgments
transplantation.104 However, in a cross-sectional study of The European Renal and Cardiovascular Medicine (EURECA-m)
graft recipients 5 years after their transplantation, more working group is one of the working groups of the European Renal
than 70% of patients had measured GFR of less than Association—European Dialysis and Transplant Association
(ERA-EDTA) and is composed of members whose careers have been
60 mL/min per 1·73 m², and thus, had limited removal
devoted to unravelling the pathophysiology of cardiovascular disease in
capacity.105 Serum β2-microglobulin concentrations of chronic kidney disease and to defining accurate therapeutic approaches
3·0 mg/L or more were observed in 58% of kidney to address this problem.
transplant recipients at discharge.106 Kidney graft References
handling of certain uraemic retention products might be 1 Almeras C, Argilés A. The general picture of uremia. Semin Dial
abnormal. For example, phosphate leakage is frequent 2009; 22: 329–33.
2 Meyer TW, Hostetter TH. Uremia. N Engl J Med 2007; 357: 1316–25.
early after transplantation, especially in patients with 3 Roberts MA, Polkinghorne KR, McDonald SP, Ierino FL.
severe hyperparathyroidism.105 Over time, kidney grafts Secular trends in cardiovascular mortality rates of patients receiving
lose function—the half-life of a cadaveric kidney graft is dialysis compared with the general population. Am J Kidney Dis
2011; 58: 64–72.
9 years and 12 years for a living-donor graft.
4 Chapman JR. What are the key challenges we face in kidney
transplantation today? Transplant Res 2013; 2 (suppl 1): S1.
Conclusion 5 Matsushita K, van der Velde M, Astor BC, et al, for the Chronic
Patients with chronic kidney disease have complex Kidney Disease Prognosis Consortium. Association of estimated
glomerular filtration rate and albuminuria with all-cause and
pathophysiology for which the underlying mechanisms cardiovascular mortality in general population cohorts:
intertwine (appendix). Inflammation and disturbed a collaborative meta-analysis. Lancet 2010; 375: 2073–81.
bone homoeostasis in particular lead to complications 6 Vanholder R, Massy Z, Argiles A, et al. Chronic kidney disease as
cause of cardiovascular morbidity and mortality.
and high and accelerated mortality. Management Nephrol Dial Transplant 2005; 20: 1048–56.
(appendix) cannot always be based on high-level 7 Jun M, Lv J, Perkovic V, Jardine MJ. Managing cardiovascular risk in
evidence, because of difficulties in the recruitment of people with chronic kidney disease: a review of the evidence from
randomized controlled trials. Ther Adv Chronic Dis 2011; 2: 265–78.
patients with sufficiently homogeneous background of 8 Ortiz A, Covic A, Fliser D, et al, for the Board of the EURECA-m
primary disease, metabolic features, and response to the Working Group of ERA-EDTA. Epidemiology, contributors to, and
uraemic syndrome. Specific therapeutic recom- clinical trials of mortality risk in chronic kidney failure. Lancet 2014;
383: 1831–43.
mendations are, therefore, based on an amalgam of
9 de Jager DJ, Grootendorst DC, Jager KJ, et al. Cardiovascular and
high-level and lower-level evidence and uraemia-related noncardiovascular mortality among patients starting dialysis. JAMA
pathophysiological reasoning. Therapeutic approaches 2009; 302: 1782–89.

www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 11


Review

10 Morton RL, Schlackow I, Mihaylova B, Staplin ND, Gray A, Cass A. 34 Locatelli F, Barany P, Covic A, et al. Kidney Disease: Improving
The impact of social disadvantage in moderate-to-severe chronic Global Outcomes guidelines on anaemia management in chronic
kidney disease: an equity-focused systematic review. kidney disease: a European Renal Best Practice position statement.
Nephrol Dial Transplant 2016; 31: 46–56. Nephrol Dial Transplant 2013; 28: 1346–59.
11 Weiner DE, Tighiouart H, Elsayed EF, et al. The Framingham 35 Bugnicourt JM, Godefroy O, Chillon JM, Choukroun G, Massy ZA.
predictive instrument in chronic kidney disease. J Am Coll Cardiol Cognitive disorders and dementia in CKD: the neglected
2007; 50: 217–24. kidney-brain axis. J Am Soc Nephrol 2013; 24: 353–63.
12 Matsushita K, Coresh J, Sang Y, et al. Estimated glomerular filtration 36 Chen YC, Su YC, Lee CC, Huang YS, Hwang SJ. Chronic kidney
rate and albuminuria for prediction of cardiovascular outcomes: a disease itself is a causal risk factor for stroke beyond traditional
collaborative meta-analysis of individual participant data. cardiovascular risk factors: a nationwide cohort study in Taiwan.
Lancet Diabetes Endocrinol 2015; 3: 514–25. PLoS One 2012; 7: e36332.
13 Vanholder R, De Smet R, Glorieux G, et al, for the European 37 Smith LK, He Y, Park JS, et al. β2-microglobulin is a systemic
Uremic Toxin Work Group (EUTox). Review on uremic toxins: pro-aging factor that impairs cognitive function and neurogenesis.
classification, concentration, and interindividual variability. Nat Med 2015; 21: 932–37.
Kidney Int 2003; 63: 1934–43. 38 Heiwe S, Jacobson SH. Exercise training in adults with CKD:
14 Duranton F, Cohen G, De Smet R, et al, for the European Uremic a systematic review and meta-analysis. Am J Kidney Dis 2014;
Toxin Work Group. Normal and pathologic concentrations of 64: 383–93.
uremic toxins. J Am Soc Nephrol 2012; 23: 1258–70. 39 Yacoub R, Habib H, Lahdo A, et al. Association between smoking
15 Vanholder R, Baurmeister U, Brunet P, Cohen G, Glorieux G, and chronic kidney disease: a case control study.
Jankowski J, for the European Uremic Toxin Work Group. A bench BMC Public Health 2010; 10: 731.
to bedside view of uremic toxins. J Am Soc Nephrol 2008; 19: 863–70. 40 Babayev R, Whaley-Connell A, Kshirsagar A, et al. Association of
16 Tripepi G, Kollerits B, Leonardis D, et al. Competitive interaction race and body mass index with ESRD and mortality in CKD stages
between fibroblast growth factor 23 and asymmetric dimethylarginine 3-4: results from the Kidney Early Evaluation Program (KEEP).
in patients with CKD. J Am Soc Nephrol 2015; 26: 935–44. Am J Kidney Dis 2013; 61: 404–12.
17 Inker LA, Tonelli M, Hemmelgarn BR, Levitan EB, Muntner P. 41 Lu JL, Molnar MZ, Naseer A, Mikkelsen MK, Kalantar-Zadeh K,
Comparison of concurrent complications of CKD by 2 risk Kovesdy CP. Association of age and BMI with kidney function and
categorization systems. Am J Kidney Dis 2012; 59: 372–81. mortality: a cohort study. Lancet Diabetes Endocrinol 2015; 3: 704–14.
18 Wühl E, Schaefer F. Managing kidney disease with blood-pressure 42 Park J, Ahmadi SF, Streja E, et al. Obesity paradox in end-stage
control. Nat Rev Nephrol 2011; 7: 434–44. kidney disease patients. Prog Cardiovasc Dis 2014; 56: 415–25.
19 Gordon JW, Shaw JA, Kirshenbaum LA. Multiple facets of NF-κB in 43 Fouque D, Aparicio M. Eleven reasons to control the protein intake
the heart: to be or not to NF-κB. Circ Res 2011; 108: 1122–32. of patients with chronic kidney disease. Nat Clin Pract Nephrol
20 Ruiz S, Pergola PE, Zager RA, Vaziri ND. Targeting the transcription 2007; 3: 383–92.
factor Nrf2 to ameliorate oxidative stress and inflammation in 44 Laville M, Fouque D. Nutritional aspects in hemodialysis.
chronic kidney disease. Kidney Int 2013; 83: 1029–41. Kidney Int Suppl 2000; 58 (suppl 76): S133–39.
21 Kooman JP, Kotanko P, Schols AM, Shiels PG, Stenvinkel P. 45 Fouque D, Vennegoor M, ter Wee P, et al. EBPG guideline on
Chronic kidney disease and premature ageing. Nat Rev Nephrol nutrition. Nephrol Dial Transplant 2007; 22 (suppl 2): ii45–87.
2014; 10: 732–42. 46 Testa A, Plou A. Clinical determinants of interdialytic weight gain.
22 Briet M, Boutouyrie P, Laurent S, London GM. Arterial stiffness J Ren Nutr 2001; 11: 155–60.
and pulse pressure in CKD and ESRD. Kidney Int 2012; 82: 388–400. 47 Moreau-Gaudry X, Jean G, Genet L, et al. A simple protein-energy
23 Minamino T, Komuro I. Vascular cell senescence: contribution to wasting score predicts survival in maintenance hemodialysis
atherosclerosis. Circ Res 2007; 100: 15–26. patients. J Ren Nutr 2014; 24: 395–400.
24 Boström KI, Rajamannan NM, Towler DA. The regulation of 48 Saran R, Bragg-Gresham JL, Levin NW, et al. Longer treatment
valvular and vascular sclerosis by osteogenic morphogens. Circ Res time and slower ultrafiltration in hemodialysis: associations with
2011; 109: 564–77. reduced mortality in the DOPPS. Kidney Int 2006; 69: 1222–28.
25 Lim K, Lu TS, Molostvov G, et al. Vascular Klotho deficiency 49 Shinaberger CS, Greenland S, Kopple JD, et al. Is controlling
potentiates the development of human artery calcification and phosphorus by decreasing dietary protein intake beneficial or
mediates resistance to fibroblast growth factor 23. Circulation 2012; harmful in persons with chronic kidney disease? Am J Clin Nutr
125: 2243–55. 2008; 88: 1511–18.
26 London GM, Marchais SJ, Guérin AP, de Vernejoul MC. 50 Yusuf S, Diener HC, Sacco RL, et al, for the PRoFESS Study Group.
Ankle-brachial index and bone turnover in patients on dialysis. Telmisartan to prevent recurrent stroke and cardiovascular events.
J Am Soc Nephrol 2015; 26: 476–83. N Engl J Med 2008; 359: 1225–37.
27 Nitta K, Okada K, Yanai M, Takahashi S. Aging and chronic kidney 51 Writing Team for the Diabetes Control and Complications
disease. Kidney Blood Press Res 2013; 38: 109–20. Trial/Epidemiology of Diabetes Interventions and Complications
28 Raschenberger J, Kollerits B, Titze S, et al. Association of relative Research Group. Sustained effect of intensive treatment of type 1
telomere length with cardiovascular disease in a large chronic kidney diabetes mellitus on development and progression of diabetic
disease cohort: the GCKD study. Atherosclerosis 2015; 242: 529–34. nephropathy: the Epidemiology of Diabetes Interventions and
29 Zawada AM, Rogacev KS, Heine GH. Clinical relevance of Complications (EDIC) study. JAMA 2003; 290: 2159–67.
epigenetic dysregulation in chronic kidney disease-associated 52 Haynes R, Lewis D, Emberson J, et al, and the SHARP
cardiovascular disease. Nephrol Dial Transplant 2013; 28: 1663–71. Collaborative Group, and the SHARP Collaborative Group.
30 Zawada AM, Rogacev KS, Hummel B, et al. SuperTAG Effects of lowering LDL cholesterol on progression of kidney
methylation-specific digital karyotyping reveals uremia-induced disease. J Am Soc Nephrol 2014; 25: 1825–33.
epigenetic dysregulation of atherosclerosis-related genes. 53 Palmer SC, Navaneethan SD, Craig JC, et al. HMG CoA reductase
Circ Cardiovasc Genet 2012; 5: 611–20. inhibitors (statins) for people with chronic kidney disease not
31 Kalantar-Zadeh K, Block G, McAllister CJ, Humphreys MH, requiring dialysis. Cochrane Database Syst Rev 2014; 5: CD007784.
Kopple JD. Appetite and inflammation, nutrition, anemia, and 54 de Brito-Ashurst I, Varagunam M, Raftery MJ, Yaqoob MM.
clinical outcome in hemodialysis patients. Am J Clin Nutr 2004; Bicarbonate supplementation slows progression of CKD and
80: 299–307. improves nutritional status. J Am Soc Nephrol 2009; 20: 2075–84.
32 Wong J, Piceno YM, Desantis TZ, Pahl M, Andersen GL, Vaziri ND. 55 Goicoechea M, de Vinuesa SG, Verdalles U, et al. Effect of
Expansion of urease- and uricase-containing, indole- and allopurinol in chronic kidney disease progression and
p-cresol-forming and contraction of short-chain fatty acid-producing cardiovascular risk. Clin J Am Soc Nephrol 2010; 5: 1388–93.
intestinal microbiota in ESRD. Am J Nephrol 2014; 39: 230–37. 56 Navarro-González JF, Mora-Fernández C, Muros de Fuentes M,
33 Anders HJ, Andersen K, Stecher B. The intestinal microbiota, et al. Effect of pentoxifylline on renal function and urinary albumin
a leaky gut, and abnormal immunity in kidney disease. Kidney Int excretion in patients with diabetic kidney disease: the PREDIAN
2013; 83: 1010–16. trial. J Am Soc Nephrol 2015; 26: 220–29.

12 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4


Review

57 Fellström BC, Jardine AG, Schmieder RE, et al, for the AURORA 78 Tentori F, Blayney MJ, Albert JM, et al. Mortality risk for dialysis
Study Group. Rosuvastatin and cardiovascular events in patients patients with different levels of serum calcium, phosphorus, and
undergoing hemodialysis. N Engl J Med 2009; 360: 1395–407. PTH: the Dialysis Outcomes and Practice Patterns Study (DOPPS).
58 de Zeeuw D, Akizawa T, Audhya P, et al, and the BEACON Trial Am J Kidney Dis 2008; 52: 519–30.
Investigators. Bardoxolone methyl in type 2 diabetes and stage 4 79 Chertow GM, Block GA, Correa-Rotter R, et al, and the EVOLVE
chronic kidney disease. N Engl J Med 2013; 369: 2492–503. Trial Investigators. Effect of cinacalcet on cardiovascular disease in
59 Verbeke F, Lindley E, Van Bortel L, et al. A European Renal Best patients undergoing dialysis. N Engl J Med 2012; 367: 2482–94.
Practice (ERBP) position statement on the Kidney Disease: 80 Palmer SC, Nistor I, Craig JC, et al. Cinacalcet in patients with
Improving Global Outcomes (KDIGO) clinical practice guideline chronic kidney disease: a cumulative meta-analysis of randomized
for the management of blood pressure in non-dialysis-dependent controlled trials. PLoS Med 2013; 10: e1001436.
chronic kidney disease: an endorsement with some caveats for 81 Liabeuf S, Neirynck N, Drüeke TB, Vanholder R, Massy ZA.
real-life application. Nephrol Dial Transplant 2014; 29: 490–96. Clinical studies and chronic kidney disease: what did we learn
60 Agarwal R, Peixoto AJ, Santos SF, Zoccali C. Pre- and postdialysis recently? Semin Nephrol 2014; 34: 164–79.
blood pressures are imprecise estimates of interdialytic ambulatory 82 Fouque D, Kalantar-Zadeh K, Kopple J, et al. A proposed
blood pressure. Clin J Am Soc Nephrol 2006; 1: 389–98. nomenclature and diagnostic criteria for protein-energy wasting in
61 Bansal N, McCulloch CE, Rahman M, et al, and the CRIC Study acute and chronic kidney disease. Kidney Int 2008; 73: 391–98.
Investigators. Blood pressure and risk of all-cause mortality in 83 Cano NJ, Fouque D, Roth H, et al, for the French Study Group for
advanced chronic kidney disease and hemodialysis: the chronic Nutrition in Dialysis. Intradialytic parenteral nutrition does not
renal insufficiency cohort study. Hypertension 2015; 65: 93–100. improve survival in malnourished hemodialysis patients: a 2-year
62 The Blood Pressure Lowering Treatment Trialists’ Collaboration. multicenter, prospective, randomized study. J Am Soc Nephrol 2007;
Blood pressure-lowering treatment based on cardiovascular risk: 18: 2583–91.
a meta-analysis of individual patient data. Lancet 2014; 84 Sirich TL, Plummer NS, Gardner CD, Hostetter TH, Meyer TW.
384: 591–98. Effect of increasing dietary fiber on plasma levels of colon-derived
63 The SPRINT Research Group. A randomized trial of intensive solutes in hemodialysis patients. Clin J Am Soc Nephrol 2014;
versus standard blood-pressure control. N Engl J Med 2015; 9: 1603–10.
373: 2103–16. 85 Bliss DZ, Stein TP, Schleifer CR, Settle RG. Supplementation
64 Heerspink HJ, Ninomiya T, Zoungas S, et al. Effect of lowering with gum arabic fiber increases fecal nitrogen excretion and
blood pressure on cardiovascular events and mortality in patients lowers serum urea nitrogen concentration in chronic renal failure
on dialysis: a systematic review and meta-analysis of randomised patients consuming a low-protein diet. Am J Clin Nutr 1996;
controlled trials. Lancet 2009; 373: 1009–15. 63: 392–98.
65 Eldehni MT, Odudu A, McIntyre CW. Randomized clinical trial of 86 Wang IK, Wu YY, Yang YF, et al. The effect of probiotics on serum
dialysate cooling and effects on brain white matter. levels of cytokine and endotoxin in peritoneal dialysis patients:
J Am Soc Nephrol 2015; 26: 957–65. a randomised, double-blind, placebo-controlled trial. Benef Microbes
66 Levin NW, Kotanko P, Eckardt KU, et al. Blood pressure in chronic 2015; 6: 423–30.
kidney disease stage 5D-report from a Kidney Disease: Improving 87 Guida B, Germanò R, Trio R, et al. Effect of short-term synbiotic
Global Outcomes controversies conference. Kidney Int 2010; treatment on plasma p-cresol levels in patients with chronic renal
77: 273–84. failure: a randomized clinical trial. Nutr Metab Cardiovasc Dis 2014;
67 Charra B, Calemard E, Ruffet M, et al. Survival as an index of 24: 1043–49.
adequacy of dialysis. Kidney Int 1992; 41: 1286–91. 88 Akizawa T, Asano Y, Morita S, et al. Effect of a carbonaceous oral
68 Moen MF, Zhan M, Hsu VD, et al. Frequency of hypoglycemia and adsorbent on the progression of CKD: a multicenter, randomized,
its significance in chronic kidney disease. Clin J Am Soc Nephrol controlled trial. Am J Kidney Dis 2009; 54: 459–67.
2009; 4: 1121–27. 89 Ueda H, Shibahara N, Takagi S, Inoue T, Katsuoka Y. AST-120, an
69 Guideline development Group. Clinical practice guideline on oral adsorbent, delays the initiation of dialysis in patients with
management of patients with diabetes and chronic kidney disease chronic kidney diseases. Ther Apher Dial 2007; 11: 189–95.
stage 3b or higher (eGFR <45 mL/min). Nephrol Dial Transplant 90 Schulman G, Berl T, Beck GJ, et al. Randomized placebo-controlled
2015; 30 (suppl 2): ii1–142. EPPIC Trials of AST-120 in CKD. J Am Soc Nephrol 2015; 26: 1732–46.
70 Palmer SC, Mavridis D, Navarese E, et al. Comparative efficacy and 91 Fell LH, Zawada AM, Rogacev KS, Seiler S, Fliser D, Heine GH.
safety of blood pressure-lowering agents in adults with diabetes and Distinct immunologic effects of different intravenous iron
kidney disease: a network meta-analysis. Lancet 2015; 385: 2047–56. preparations on monocytes. Nephrol Dial Transplant 2014; 29: 809–22.
71 Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, 92 Bailie GR, Larkina M, Goodkin DA, et al. Data from the Dialysis
cardiovascular outcomes, and mortality in type 2 diabetes. Outcomes and Practice Patterns Study validate an association
N Engl J Med 2015; 373: 2117–28. between high intravenous iron doses and mortality. Kidney Int 2015;
72 Tonelli M, Wanner C, for the Kidney Disease: Improving Global 87: 162–68.
Outcomes Lipid Guideline Development Work Group Members. 93 Macdougall IC, Obrador GT. How important is transfusion
Lipid management in chronic kidney disease: synopsis of the avoidance in 2013? Nephrol Dial Transplant 2013; 28: 1092–99.
Kidney Disease: Improving Global Outcomes 2013 clinical practice 94 Zhang X, Xiang C, Zhou YH, Jiang A, Qin YY, He J. Effect of statins
guideline. Ann Intern Med 2014; 160: 182–89. on cardiovascular events in patients with mild to moderate chronic
73 Baigent C, Landray MJ, Reith C, et al. The effects of lowering LDL kidney disease: a systematic review and meta-analysis of randomized
cholesterol with simvastatin plus ezetimibe in patients with chronic clinical trials. BMC Cardiovasc Disord 2014; 14: 19.
kidney disease (Study of Heart and Renal Protection): a randomised 95 Seliger SL, Zhang AD, Weir MR, et al. Erythropoiesis-stimulating
placebo-controlled trial. Lancet 2011; 377: 2181–92. agents increase the risk of acute stroke in patients with chronic
74 Massy ZA, de Zeeuw D. LDL cholesterol in CKD—to treat or not to kidney disease. Kidney Int 2011; 80: 288–94.
treat? Kidney Int 2013; 84: 451–56. 96 Floege J, Bartsch A, Schulze M, Shaldon S, Koch KM, Smeby LC.
75 Winkelmayer WC, Liu J, Kestenbaum B. Comparative effectiveness Clearance and synthesis rates of beta 2-microglobulin in patients
of calcium-containing phosphate binders in incident U.S. dialysis undergoing hemodialysis and in normal subjects. J Lab Clin Med
patients. Clin J Am Soc Nephrol 2011; 6: 175–83. 1991; 118: 153–65.
76 Isakova T, Gutiérrez OM, Chang Y, et al. Phosphorus binders and 97 Cornelis T, van der Sande FM, Eloot S, et al. Acute hemodynamic
survival on hemodialysis. J Am Soc Nephrol 2009; 20: 388–96. response and uremic toxin removal in conventional and extended
77 Zoccali C, Bolignano D, D’Arrigo G, et al, for the European Renal hemodialysis and hemodiafiltration: a randomized crossover study.
and Cardiovascular Medicine (EURECA-m) Working Group of the Am J Kidney Dis 2014; 64: 247–56.
European Renal Association–European Dialysis Transplantation 98 Cornelis T, Eloot S, Vanholder R, et al. Protein-bound uraemic
Association (ERA-EDTA). Validity of vascular calcification as a toxins, dicarbonyl stress and advanced glycation end products in
screening tool and as a surrogate end point in clinical research. conventional and extended haemodialysis and haemodiafiltration.
Hypertension 2015; 66: 3–9. Nephrol Dial Transplant 2015; 30: 1395–402.

www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4 13


Review

99 Susantitaphong P, Siribamrungwong M, Jaber BL. 103 Böhringer F, Jankowski V, Gajjala PR, Zidek W, Jankowski J.
Convective therapies versus low-flux hemodialysis for chronic Release of uremic retention solutes from protein binding by
kidney failure: a meta-analysis of randomized controlled trials. hypertonic predilution hemodiafiltration. ASAIO J 2015; 61: 55–60.
Nephrol Dial Transplant 2013; 28: 2859–74. 104 Budde K, Becker T, Arns W, et al. Everolimus-based,
100 Maduell F, Moreso F, Pons M, et al, for the ESHOL Study Group. calcineurin-inhibitor-free regimen in recipients of de-novo kidney
High-efficiency postdilution online hemodiafiltration reduces transplants: an open-label, randomised, controlled trial. Lancet 2011;
all-cause mortality in hemodialysis patients. J Am Soc Nephrol 2013; 377: 837–47.
24: 487–97. 105 Perrin P, Caillard S, Javier RM, et al. Persistent hyperparathyroidism
101 Nistor I, Palmer SC, Craig JC, et al. Convective versus diffusive dialysis is a major risk factor for fractures in the five years after kidney
therapies for chronic kidney failure: an updated systematic review of transplantation. Am J Transplant 2013; 13: 2653–63.
randomized controlled trials. Am J Kidney Dis 2014; 63: 954–67. 106 Astor BC, Muth B, Kaufman DB, Pirsch JD, Michael Hofmann R,
102 Maduell F, Arias-Guillen M, Fontseré N, et al. Elimination of large Djamali A. Serum β2-microglobulin at discharge predicts mortality
uremic toxins by a dialyzer specifically designed for high-volume and graft loss following kidney transplantation. Kidney Int 2013;
convective therapies. Blood Purif 2014; 37: 125–30. 84: 810–17.

14 www.thelancet.com/diabetes-endocrinology Published online March 2, 2016 http://dx.doi.org/10.1016/S2213-8587(16)00033-4

Vous aimerez peut-être aussi