You are on page 1of 8

European Journal of Internal Medicine 24 (2013) 721–728

Contents lists available at ScienceDirect

European Journal of Internal Medicine


journal homepage: www.elsevier.com/locate/ejim

Review article

Clinical indications for the albumin use: Still a controversial issue


Paolo Caraceni ⁎, Marco Domenicali, Alessandra Tovoli, Lucia Napoli, Carmen Serena Ricci,
Manuel Tufoni, Mauro Bernardi
U.O. Semeiotica Medica, Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy

a r t i c l e i n f o a b s t r a c t

Article history: Human serum albumin (HSA) is the most abundant circulating protein and accounts for about 70% of the
Received 13 May 2013 plasma colloid osmotic pressure. Beside the well known capacity to act as plasma-expander, HSA is provid-
Received in revised form 21 May 2013 ed of many other properties which are unrelated to the regulation of fluid compartmentalization, including
Accepted 24 May 2013
binding and transport of many endogenous and exogenous substances, antioxidant function, immuno-
Available online 20 June 2013
modulation, anti-inflammatory activity, and endothelial stabilization.
Keywords:
Treatment (hepatorenal syndrome) or prevention (renal failure after spontaneous bacterial peritonitis and post-
Albumin paracentesis circulatory dysfunction after large volume paracentesis) of severe clinical complications in patients
Liver cirrhosis with cirrhosis and fluid resuscitation in critically ill patients, when crystalloids and non-proteic colloids are not
Ascites effective or contra-indicated, represents the major evidence-based clinical indications for HSA administration.
Fluid resuscitation However, a large proportion of HSA prescription is inappropriate. Despite the existence of solid data against a
Sepsis real benefit, HSA is still given for nutritional interventions or for correcting hypoalbuminemia per se (without
hypovolemia). Other clinical uses for HSA administration not supported by definitive scientific evidence are
long-term treatment of ascites, nephrotic syndrome, pancreatitis, abdominal surgery, acute distress respira-
tory syndrome, cerebral ischemia, and enteric diseases.
HSA prescription should be not uncritically restricted. Enforcement of clinical practice recommendations has
been shown to allow a more liberal use for indications supported by strong scientific data and to avoid the futile
administration in settings where there is a lack of clinical evidence of efficacy. As a result, a more appropriate HSA
use can be achieved maintaining the health care expenditure under control.
© 2013 European Federation of Internal Medicine. Published by Elsevier B.V. All rights reserved.

1. Introduction 2. The molecule of albumin

Human serum albumin (HSA) is the most abundant circulating pro- 2.1. Structure
tein in the body provided of both oncotic and non-oncotic properties.
Administration of exogenous HSA dates back in the World War II when HSA is the main circulating protein in healthy individuals (3.5–5 g/dl),
it was used for fluid resuscitation. Since then, the use of HSA has been ex- representing about 50% of the total protein content in the plasma. HSA is a
tended to many other diseases since physicians commonly believe in its small protein with a molecular weight of 66.5 kDa, consisting of a single
efficacy. However, beside some clinical indications supported by solid chain of 585 amino acids organized in three repeated homologue
scientific evidence, the administration in many other settings is still domains (sites I, II, and III), each of which comprised of two separate
under debate or has been disproved by evidence-based medicine. As a sub-domains (A and B). In the human body, HSA assumes a globular
result, clinical practical recommendations have been proposed to ratio- heart-shaped conformation formed by α-helices for about 67%. Of the
nalize the prescription of HSA and avoid its futile use. 35 cysteine residues of the albumin molecule, 34 are involved in internal
disulfide bonds which stabilize the spatial conformation of the molecule,
while the cysteine at position 34 (Cys-34) remains free [1–3].

Abbreviations: HRS, hepatorenal syndrome; HSA, human serum albumin; ICU, intensive 2.2. Synthesis
care unit; NO, nitric oxide; PPCD, post-paracentesis circulatory dysfunction; SBP, spontane-
ous bacterial peritonitis; TNF-α, tumor necrosis factor-α. HSA is synthesized in the liver through the transcription of its gene
⁎ Corresponding author at: U.O. Semeiotica Medica, Department of Medical and Surgical
Sciences, Alma Mater Studiorum, University of Bologna, Via Albertoni15, 40138 Bologna,
on the long arm of chromosome 4. Under physiological conditions,
Italy. Tel.: +39 051 6362919; fax: +39 051 6362930. 20–30% of the hepatocytes are implicated in the synthesis and about
E-mail address: paolo.caraceni@unibo.it (P. Caraceni). 10–15 g are produced and released in the circulation every day, with

0953-6205/$ – see front matter © 2013 European Federation of Internal Medicine. Published by Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.ejim.2013.05.015
722 P. Caraceni et al. / European Journal of Internal Medicine 24 (2013) 721–728

none or a very low intracellular storage. However, under certain compartment. As a result of its osmotic effect, most of the clinical use
circumstances, the HSA production can increase even up to 3–4 folds, of HSA is based on the capacity to act as a plasma-expander [4].
thus exploiting a large functional reserve [3,4]. Its synthesis is stimulat-
ed by hormonal factors, such as insulin, cortisol and growth hormone, 3.2. Non-oncotic properties
while pro-inflammatory mediators, including cytokines, such as
Interleukin-6 and Tumor Necrosis Factor-α, exert an inhibitory effect Many non-oncotic properties, mostly related to the peculiar struc-
[4,5]. ture and conformation of the albumin molecule, have been identified
in the last two decades (Fig. 1).
2.3. Metabolism A large number of binding sites with different affinities for plasmatic
compounds have been identified: the major domains are capable of fold-
Once produced, HSA is quickly released in the blood through the ing into hydrophobic pockets, which can open and close, and accommo-
sinusoids. Approximately 30–40% is maintained in the blood stream, date large insoluble anions; furthermore, cationic groups located on the
while the remaining pool is distributed in the interstitium, where its surface of the molecule allow the formation of ionic bonds with many
concentration is low (1.4 g/dl), as well as in the muscles and the skin. different ligands. As a result, HSA binds and carries a great variety of hy-
The protein leaves the vascular compartment at a rate of 5% per hour drophobic molecules, such as endogenous (i.e., cholesterol, fatty acids, bil-
(transcapillary escape rate), returning to it via the lymphatic system irubin, thyroxine) or exogenous substances (i.e., drugs), transition metal
in an amount comparable to the output. The circulatory half-life of ions, and gas (nitric oxide [NO]), with consequent implications on their
HSA, which is approximately 16–18 h, is therefore much shorter solubilization, transport and metabolism [3,6]. Structural and conforma-
than its total half-life, which varies from about 12 to 19 days in a tional changes at the molecular level have been demonstrated to occur
healthy young adult. Although its catabolism is a widespread process during chronic diseases, producing an impairment of the binding, trans-
involving many tissues, HSA is mainly degraded by the muscles, liver port and detoxification capacity of HSA [7,8].
and kidneys, while a little amount is lost into the gastrointestinal
tract [3–6]. 3.2.1. Antioxidant activity
HSA is the major source of extracellular reduced sulfhydryl groups,
3. The functions of albumin which act as potent scavengers of reactive oxygen species (ROS), thus
constituting the main circulating antioxidant system in the body.
Beside the well known oncotic function, HSA is provided of many According to the redox state of the sulfhydryl group in the Cys-34 posi-
other properties which are unrelated to the regulation of fluid compart- tion, three albumin isoforms can be identified. Under physiological con-
mentalization, the so-called non-oncotic properties. ditions, about 70–80% of albumin contains the Cys-34 residue with a
free sulfhydryl group (known as human mercaptalbumin). In 25–30%
3.1. Oncotic properties of albumin, the Cys-34 site is reversibly bound with other small
sulfhydril compounds, such as homocysteine or glutathione, forming
HSA is the main modulator of fluid distribution in the various com- the non-mercaptalbumin 1 isoform, which is provided of a reduced
partments of the body as it accounts for about 70–80% of the plasma redox potential. Finally, less than 5% circulates with the residue
oncotic pressure. The oncotic property derives for 2/3 from the direct Cys-34 completely and irreversibly oxidized (non-mercaptalbumin 2),
osmotic effect related to its molecular mass and for 1/3 from the with a permanent loss of its antioxidant activity [8]. In acute and chronic
Gibbs–Donnan effect, due to the negative net charge of the molecule disease states characterized by a pro-oxidant and pro-inflammatory cir-
at physiological pH that allows the protein to attract positively charged culatory microenvironment (myocardial infarction [9], diabetes [10,11],
molecules (i.e. sodium and, therefore, water) into the intravascular renal failure [12,13], liver cirrhosis [14]), the amount of the totally

ROS Oncotic
ONOO- pressure
hydroperoxide

Endogenous Solubilization
III B IB Transport
molecules
Bilirubin Metabolism
Biliary salts IA
Fatty acids
Binding/Transport

Antioxidant
Functions

Nitric oxide SH

Immuno-
Exogenous modulation
III A
molecules
II A
Benzene
Aflatossin G
Endothelial
stabilization
Drugs
NSAIDs NH2
Warfarin Capillary
Antibiotics II B
permeability
Furosemide

Metals Hemostasis
Cu, Ni, Co, Fe

Fig. 1. Human serum albumin is provided of a series of oncotic and non-oncotic properties (right side). A major function is the binding and transport of many endogenous and
exogenous substances (left side), which are capable to interact with several specific sites of the albumin molecule.
P. Caraceni et al. / European Journal of Internal Medicine 24 (2013) 721–728 723

oxidized HSA is increased several folds and correlates with poor clinical Today, we know that the cardiovascular abnormalities occurring in
outcome and patient survival [15]. advanced cirrhosis are the essential background for the development
Finally, the antioxidant function of HSA resides also on the capability of ascites formation and other severe clinical complications of the dis-
to bind at the N-terminal portion of the molecule several metal ions, in- ease [26,27]. The schematic representation of the underlying patho-
cluding copper, cobalt, nickel, zinc and iron, which are therefore inhibited physiological events is described in Fig. 2. Thus, the preservation of
to catalyze many chemical reactions generating free radicals [3,16,17]. the effective blood volume has emerged as a primary goal in the man-
agement of patients with advanced cirrhosis [28]. Based on its capacity
3.2.2. Antithrombotic effects to act as a plasma-expander, the use of HSA is currently proposed by the
These proprieties are mostly due to the capacity to bind NO at the international guidelines to treat or prevent clinical complications of cir-
Cys-34 site, with subsequent formation of the complex albumin-NO, rhosis all characterized by extreme effective hypovolemia, such as in
also known as nitroso-albumin. This new compound, whose physiolog- preventing post-paracentesis circulatory dysfunction (PPCD) and the
ic and pathologic functions are still partially determined, seems to pre- renal failure induced by spontaneous bacterial peritonitis (SBP) and in
vent the rapid inactivation of NO thus prolonging its anti-aggregant treating hepatorenal syndrome (HRS), in association with vasoconstric-
effect on platelets [18,19]. tor drugs [29,30] (Fig. 3).
Beside these universally accepted indications, the use of HSA has been
proposed in patients with cirrhosis for the chronic treatment of ascites,
3.2.3. Anti-inflammatory effect
hypervolemic hyponatremia and hepatic encephalopathy, although all
HSA appears to inhibit the secretion of pro-inflammatory cytokines
these indications await to be supported by a clear scientific evidence
(TNF-α) and complement factors (C5a) through the modulation of
[6,28].
the signaling systems between inflammatory cells, such as neutrophils
A final assumption deriving from the pathophysiological scenario
and endothelial cells [20,21].
is that serum albumin concentration should not be a guide for its
use and the correction of hypoalbuminemia per se should not be a
3.2.4. Capillary permeability and endothelial stabilization
goal to pursue.
HSA contributes to the integrity of the vessels, by binding the in-
terstitial matrix and interacting with the sub-endothelial space, thus
4.1.1. Prevention of PPCD after large volume paracentesis
participating in the maintenance of the normal capillary permeabil-
Paracentesis is the current first-line treatment for patients with tense
ity [22]. HSA may impact positively on endothelial function also by
and refractory ascites [29,30]. The removal of large volumes of ascites
reducing oxidative damage and modulating inflammation [23].
CIRRHOSIS
4. Clinical applications of albumin
PORTAL
HSA is largely used in clinical practice, but its administration is often HYPERTENSION
inappropriate. Indeed, physicians commonly believe in its efficacy al-
though many indications are still under debate or have been disproved CIRRHOTIC
SPLANCHNIC
by evidence-based medicine. VASODILATION
CARDIOMIOPATHY
(end-stage disease)
Hepatology is a setting where HSA is recommended by international
guidelines, since randomized clinical trials and meta-analyses have dem-
EFFECTIVE HYPOVOLEMIA
onstrated its efficacy to prevent or treat some severe complications of
cirrhosis. In critically ill patients, HSA is also widely used for fluid resus-
citation, although non-protein colloids and crystalloids are considered
the first-line treatment. Furthermore, HSA is prescribed for some other ACTIVATION OF
specific conditions, such as nephrotic syndrome, pancreatitis, major sur- COMPENSATORY REDUCED
gery, kernicterus, plasmapheresis, and graft versus host disease, even NEURO-HORMONAL RENAL PERFUSION
SYSTEMS
though these indications are not supported by definite scientific evi-
dence. Finally, the most common inappropriate use occurs when HSA
is given to correct hypoalbuminemia per se (i.e. not associated with
hypovolemia) or for nutritional intervention.
RENAL RETENTION RENAL RETENTION RENAL PERFUSION
Na+ and H2O Na+ and H2O
4.1. HSA in liver cirrhosis

Hypoalbuminemia is a typical feature of cirrhosis and represents an


independent adverse prognostic factor. Beside the decreased hepato- ASCITES HYPERVOLEMIC HEPATORENAL
HYPONATREMIA SYNDROME
cyte synthesis, the low serum albumin concentration results from the
dilution of the extracellular fluid protein content, due to the plasma vol-
ume expansion consequent to renal sodium and water retention, and
DISEASE SEVERITY
from the increased trans-capillary escape rate towards the extravascu-
lar space, at least in the most advanced stage of cirrhosis [24].
The therapeutic use of HSA in cirrhosis started more than 50 years Fig. 2. Cardiovascular abnormalities in cirrhosis. An imbalance of the vasoactive systems
with a predominance of vasodilators (i.e. nitric oxide, carbon monoxide, endocannabinoids)
ago, when hypoalbuminemia was thought to be crucial for the develop-
reduces the tone of resistance vessels and hampers their response to vasoconstrictors, induc-
ment of ascites, due to the alteration of Starling force balance in the ing vasodilation, mainly located in the splanchnic area. This leads to a reduction in the central
intrahepatic microcirculation. However, the net fluid flow from the intra- effective volemia, which, in turn, evokes the compensatory activation of neuro-humoral
vascular compartment to the interstitium is regulated by the colloid systems, such as the renin–angiotensin–aldosterone axis (RAAS), the sympathetic nervous
osmotic gradient rather than by the absolute level of intrasinusoidal os- system (SNS) and arginine–vasopressin (ADH), ultimately promoting renal sodium and
water retention. In the advanced stages of cirrhosis, effective hypovolemia is further im-
motic pressure. Interestingly, it has been shown that the intravascular– paired by cardiac dysfunction, as witnessed by a relative reduction in cardiac output. These
interstitial colloid osmotic gradient did not significantly differ between events represent the pathophysiological background for the development of clinical compli-
healthy controls and patients with cirrhosis and ascites [25]. cations of cirrhosis, including ascites, hyponatremia, and renal failure.
724 P. Caraceni et al. / European Journal of Internal Medicine 24 (2013) 721–728

Evidence based clinical indications for albumin admistration in cirrhosis

Spontaneous Hepatorenal syndrome


Paracentesis bacterial
peritonitis Diagnosis Treatment

Cr > 1.5 mg/dL

Prevention Prevention of renal Exclusion of Type I HRS


of PPCD failure hypovolemia

8 g/liter ascites Day 1: 1.5 g/kg 1 g/kg per day Day 1: 1g/kg (max 100 g),
removed Day 3: 1 g/kg (at least two days) then 20-40 g/day
+ antibiotics + diuretics + vasoconstrictors in HRS 1
withdrawal

Ascites
Bacterial
Hepatic encephalopathy
infections (other than SBP)
Hypervolemic hyponatremia

grey zone
Lack of evidences to make recomendations

Fig. 3. Evidence-based and controversial clinical indications (gray zone) for the use of human serum albumin in patients with complicated liver cirrhosis. HRS: hepatorenal syndrome;
PPCD: post-paracentesis circulatory dysfunction; SBP: spontaneous bacterial peritonitis.

(>5 l) has been associated with an increased risk to develop PPCD, which [42,43]. Similarly, the dose of HSA proposed is arbitrary and chosen
is diagnosed by a significant increase (>50%) in plasma renin activity based on pathophysiological considerations and not according compara-
6 days after paracentesis. PPCD is a circulatory dysfunction characterized tive clinical studies. At this regard, a reduced-dose regimen of albumin
by an exacerbation of arteriolar vasodilatation, reduction of effective (1.0 g/kg on day 1 and 0.5 g/kg on day 3) seemed to be as effective as
blood volume, rapid re-accumulation of ascites, increased risk of HRS, the standard regimen in preventing renal failure and HRS [44].
dilutional hyponatremia, and increased mortality [31,32]. In several ran- Until more information are available, the Clinical Practice Guidelines
domized trials, administration of HSA at the time of paracentesis was of the European Association for the Study of the Liver (EASL) recom-
able to reduce significantly the incidence of PPCD, showing also its supe- mend that all patients who develop SBP should be treated with broad
riority when compared with other plasma expanders or vasoconstrictors spectrum antibiotics and HSA (1.5 g/kg on day 1 and 1 g/kg on day 3)
[33–35]. Importantly, a recent meta-analysis has shown that HSA not only [29]. Supporting this recommendation, a very recent meta-analysis
reduces the occurrence of PPCD more efficiently than any other plasma from Salerno at al. has confirmed the capacity of HSA to improve out-
expander, but is also superior to either artificial plasma expanders or va- comes in patients with SBP [45].
soconstrictors in lowering the incidence of hyponatremia and mortality
[36]. On the basis of such evidences, international guidelines recommend
the administration of 8 g of HSA per liter of ascites removed, with a great- 4.1.3. Diagnosis and treatment of HRS
er strength of recommendation for paracentesis of at least 4–5 l [29,30]. HRS is a potentially reversible renal failure developing in patients
with advanced cirrhosis and ascites and it is characterized by marked
4.1.2. Prevention of renal failure after SBP renal hypoperfusion related to the extreme effective hypovolemia, due
SBP is a frequent and life-threatening infection of ascitic fluid that oc- to splanchnic vasodilatation and cardiac dysfunction, and the conse-
curs in patients with advanced cirrhosis and requires prompt recognition quent overactivation of compensatory mechanisms that induce severe
and treatment. Diagnosis is based on the presence of >250 polymorpho- intrarenal vasoconstriction [26,46].
nuclear cells/mmc of ascites, in the absence of an intra-abdominal source The diagnostic criteria for HRS have been published in 2007 by the In-
of infection or malignancy [37]. Even in absence of septic shock, SBP may ternational Club of Ascites (Table 1). HRS is diagnosed in patients with cir-
trigger hemodynamic decompensation precipitating renal failure that rhosis and ascites and the exclusion of organic and other functional forms
can become progressive as type 1 HRS. Thus, despite infection resolution, of renal failure. At this regard, diuretic withdrawal and HSA administra-
SBP still carries approximately a 10–30% hospital mortality [38–40]. tion (1 g/kg of body weight per day up to a maximum of 100 g/day) for
A prospective randomized comparative study reported that adminis- at least 2 days are recommended to exclude hypovolemic renal failure
tration of high-dose of HSA (1.5 g/kg at diagnosis of SBP and 1 g/kg on [46]. The expert panel of the International Club of Ascites agreed that vol-
day 3), together with antibiotic treatment, decreased the incidence of ume expansion should be performed with HSA rather than saline, be-
type 1 HRS (from 30% to 10%) and improved in-hospital and 3-month cause of its greater and more sustained expansion [46].
survival [39]. Whether all patients with SBP should be treated with anti- Once diagnosis in established, the current therapeutic approach in-
biotics and HSA is still matter of debate. Indeed, available data suggest cludes the administration of both vasoconstrictors and HSA with the
that the most striking effects are obtained in patients with liver failure aim of improving effective volemia. Several vasoconstrictors have
(serum bilirubin > 4 mg/dl) and renal dysfunction (serum creatinine been used in the management of type 1 HRS, but terlipressin, which
greater than 1 mg/dl) at the time of diagnosis, suggesting that HSA ad- mainly acts on the splanchnic vascular bed where vasodilation is maxi-
ministration could be restricted to these high-risk patients [39,41]. How- mal, is the most studied and widely used [47–51]. As recently shown by
ever, two large subsequent retrospective studies showed a not negligible a systematic review of randomized studies, terlipressin is able to resolve
incidence of poor outcome in low-risk patients not treated with albumin HRS in about 40% of the cases and to improve patient survival [52]. In
P. Caraceni et al. / European Journal of Internal Medicine 24 (2013) 721–728 725

Table 1 albumin, bacterial infections, refractory ascites and renal failure [56].
Diagnostic criteria and classification of hepatorenal syndrome (HRS) in cirrhosis according Hypervolemic hyponatremia in cirrhosis is the result of effective
the International Club of Ascites [46].
hypovolemia that leads to greater free water reabsorption mediated
Diagnostic criteria by the non-osmotic secretion of vasopressin [56]. Although some physi-
• Cirrhosis with ascites cians prescribe HSA to correct moderate (serum sodium b 130 mmol/l)
• Serum creatinine > 1.5 mg/dl (133 mmol/l)
or severe (serum sodium b 125 mmol/l) hyponatremia with the aim to
• Absence of shock
• Absence of hypovolemia as defined by no sustained improvement of renal function expand the central volume and suppress the ADH release, there is no ac-
(creatinine decreasing to b1.5 mg/dl–133 mmol/l) following at least 2 days tual recommendation about its use because of the lack of clinical studies
of diuretic withdrawal (if on diuretics), and volume expansion with albumin specifically dedicated to this topic.
at 1 g/kg/day up to a maximum of 100 g/day
• No current or recent treatment with nephrotoxic drugs
• Absence of parenchymal renal disease as defined by proteinuria b0.5 g/day,
4.2. Albumin in critically ill patients
no microhematuria (b50 red cells/high powered field), and normal renal
ultrasonography Fluid resuscitation is widely practiced in intensive care units (ICU)
Classification for the treatment of critically ill patients. Although HSA has been
• Type 1 HRS: a rapid progressive renal failure as defined by doubling of the initial
given for many decades in a large number of diseases, including
serum creatinine concentration to a level greater than 2.5 mg/dl in less than
2 weeks. It often appears after a precipitating event, most frequently spontaneous shock, sepsis, trauma, acute respiratory distress syndrome, burns or
bacterial peritonitis. acute clinical situations associated with hypoproteinemia, a long de-
• Type 2 HRS: a moderate renal failure (serum creatinine from 1.5 to 2.5 mg/dl), with bate is still ongoing since the choice of fluid is controversial with crys-
a steady or slowly progressive course. It often appears spontaneously and is
talloids and non-protein colloids being also proposed for treatment.
typically associated with refractory ascites.
In 1998, a Cochrane meta-analysis of 30 randomized clinical trials
strongly suggested that the infusion of HSA solutions in the management
contrast, the use of vasoconstrictors and albumin in type 2 HRS is still of patients with hypovolemia from injury or surgery, burns and
controversial [29,30]. hypoproteinemia increased the risk of death by 6% (pooled relative
risk: 1.68) [57]. However, few years later, a meta-analysis including 55
4.1.4. Other potential indications for the use of HSA in cirrhosis trials on the comparison of HSA therapy with crystalloid therapy, no
HSA or lower HSA doses in patients with trauma or surgery, burns,
4.1.4.1. Long-term treatment of ascites. The chronic and prolonged use hypoalbuminemia, ascites or high-risk neonates found no significant ef-
of HSA to treat ascites is still debated, due to the lack of a definitive fect of HSA on mortality [58]. The latter finding was then confirmed by
evidence showing a clinical benefit and an adequate assessment of the third meta-analysis published on this topic by the Cochrane group
the cost/effectiveness ratio. in 2004 [59]. At the same time, the Saline versus Albumin Fluid Evalua-
So far, the effect of the combination of HSA and diuretics has only been tion (SAFE study) evaluating 6997 patients admitted to the ICU and ran-
assessed in two controlled clinical trials conducted in Italy and involving a domized to receive saline or 4% HSA solution for fluid resuscitation
relatively small number of patients. In the first study, two groups of pa- demonstrated that the 28-day mortality was similar in the two groups,
tients with cirrhosis and ascites were randomized to receive diuretics supporting the safety use of HSA in critically ill patients [60]. More re-
with or without daily infusion of 12.5 g of albumin during hospitalization cently, in 2011, another Cochrane meta-analysis on 38 trials, comparing
followed by 25 g/week after discharge, with a median follow-up of HSA or plasma protein fraction with no HSA or plasma protein fraction or
20 months [53]. The treatment with diuretics plus HSA was overall with a crystalloid solution in critically ill patients with hypovolemia,
more effective than diuretics alone in resolving ascites and reducing the burns or hypoalbuminemia, showed no evidence of survival benefit of
length of the hospital stay. However, these results were only achieved HSA compared to the other cheaper alternatives [61].
in the subset of patients receiving a low dose of diuretics, while the ad- However, all these studies included a very heterogeneous population
vantage was lost when higher doses were needed [53]. In a subsequent of critically ill patients, while the effect of HSA may be quite different
study, a cohort of patients was randomized to receive chronic treatment depending on the specific clinical conditions. Indeed, while consistent
with either diuretic alone or diuretic plus HSA (25 g/week for the first evidence have indicated that HSA administration for resuscitation of
year, then 25 g every 2 weeks) (median follow up 84 months) [54]. patients with traumatic brain injury is associated to an increased risk of
The recurrence rate of moderate to severe ascites and mortality were sig- mortality, accumulating data support its use in patients with sepsis
nificantly reduced in patients supplemented with HSA; however, the rel- [62,63]. Indeed, the SAFE group in a pre-defined subgroup analysis of
atively low sample size prevented to reach firm conclusions [54]. 1218 patients with severe sepsis randomized to receive HSA or saline
In order to fill this gap of knowledge, an open-label, multicenter, showed that, at multivariate analysis, those treated with HSA administra-
randomized clinical trial (NCT01288794, www.clinicaltrials.gov, the tion may have a decreased risk of death with an adjusted odd ratio of 0.71
“ANSWER Study”) that will enroll more than 400 patients is currently [63]. Similarly, a meta-analysis by Delaney et al. comparing crystalloid
ongoing in Italy. This study has the potential to assess the effective- and albumin in septic patients showed a survival benefit with HSA [64].
ness of long-term administration of HSA in improving ascites treat- Furthermore, besides acting as plasma-expander, HSA might exert an ad-
ment and patient mortality. ditional beneficial effect in these patients by increasing the plasma
anti-oxidant and anti-inflammatory capacity and by reducing the capil-
4.1.4.2. Bacterial infections other than SBP. Even less defined is the indi- lary permeability and thus the fluid loss towards the interstitium [6]. As
cation on the prophylactic use of HSA in patients with bacterial infec- a result, the most recent guidelines of the surviving sepsis campaign sup-
tions other than SBP. In a recent randomized controlled trial, HSA port, even if the level of recommendation is still low, the use of HSA for
administration together with antibiotics, as compared with standard fluid resuscitation in patients with severe sepsis and septic shock when
antibiotic therapy alone, had some beneficial effects on the renal and substantial amount of crystalloids are required, suggesting also to avoid
circulatory functions, showing also a potential survival benefit at least the use of hydroxyethyl starch [65]. A definitive indication on the use of
in high-risk patients. However, due to the relatively low sample size, a HSA in this specific clinical setting will be hopefully provided by a ran-
clear recommendation cannot be given as yet [55]. domized prospective multicenter clinical trial, actually ongoing in Italy,
comparing HSA with crystalloids among patients with severe sepsis
4.1.4.3. Hypervolemic hyponatremia. Hyponatremia (serum sodium b (NCT00707122, www.clinicaltrials.gov, “the ALBIOS study”).
135 mmol/l) frequently complicates advanced cirrhosis and its de- Thus, if HSA administration for resuscitation is now considered safe
velopment is associated to the use of diuretics, paracentesis without in critically patients except for those with traumatic brain injury, its
726 P. Caraceni et al. / European Journal of Internal Medicine 24 (2013) 721–728

benefit above cheaper alternatives (crystalloids and non-proteic col- We recently reported the impact of internal practice guidelines for the
loids) has not been unequivocally demonstrated. However, recent appropriate use of albumin in the S. Orsola-Malpighi Academic Hospital
evidence indicate that HSA could be instead more effective at least in in Bologna, Italy, a third-level referral center for many diseases, including
some specific clinical conditions, such as sepsis, severe sepsis and septic liver cirrhosis and transplantation, with more than 1.700 beds and 70.000
shock [63,64]. admissions per year [69]. Due to the fact that HSA consumption and costs
had been relentlessly increasing in the period comprised between 1997
4.3. Other clinical indications and inappropriate use of albumin and 2003, practical recommendations were elaborated using a systematic
literature-based consensus method by a multi-disciplinary team, which
Beside liver cirrhosis and critical care illnesses, some niche indica- identified conditions where HSA had to be considered the first- or
tions for the appropriate use of HSA can reflect the peculiar settings of the second-line treatment and others where HSA was formally not
highly specialized centers. An example is the extracorporeal liver sup- recommended (Table 2) [69].
port device Molecular Adsorbant Recirculating System (MARS), which As a result of their implementation in 2003, the HSA consumption
is based on the binding and detoxification capacity of albumin, for the and the related costs dropped suddenly about 20% to remain steady
treatment of advanced liver failure, hepatic encephalopathy and intrac- over the following year. Even more interestingly, the decrease observed
table cholestatic pruritus [66–68]. in the “non-hepatological” units after guideline implementation was
However, there is no doubt that, in many other clinical conditions, the mirrored by a parallel increase in the “hepatological” units of our hospi-
lack of definitive scientific evidence have produced some confusion and a tal. Furthermore, no significant changes occurred in the prescription by
great variability regarding which indication is perceived to be appropri- physicians working in ICUs, probably because they were already accus-
ate. As a result, a consistent part of the HSA prescription, ranging from tomed to established international guidelines, despite the dispute on
40% up to 90%, is not supported by clinical evidence, guidelines or prac- the HSA administration in critically ill patients lasts almost two decades
tical recommendations, as it has been consistently revealed by several [69].
studies and surveys in different countries [69–73]. In few words, the enforcement of in-hospital guidelines allowed a
Most of the inappropriate HSA prescription derives from the use for more liberal use of HSA for indications supported by strong scientific
nutritional interventions or for correcting hypoalbuminemia per se data and avoided its futile administration in settings where there is a
(without hypovolemia) that still occurs in many clinical areas (i.e. sur- lack of clinical evidence of efficacy. As a result, a more appropriate
gery, internal medicine, geriatrics, oncology), despite the existence of HSA prescription was achieved maintaining the health care expendi-
solid data against a real benefit. Other clinical uses for HSA administra- ture under control.
tion not supported by solid scientific evidence are nephrotic syndrome,
pancreatitis, abdominal surgery, acute distress respiratory syndrome, Learning points
cerebral ischemia, and enteric diseases [69–73].
Finally, inappropriate prescription can also occur even in presence of • Clinical indications for HSA administration have emerged from
clinical guidelines and recommendations, i.e. when HSA is prescribed as evidence-based medicine.
first-line treatment for fluid resuscitation even if other cheaper plasma- • HSA is the first choice to expand effective volemia in patients with
expanders are not contraindicated or for chronic treatment of cirrhosis. advanced cirrhosis and should be used to prevent renal failure
after spontaneous bacterial peritonitis and the post-paracentesis
5. Impact of clinical recommendations for albumin prescription circulatory dysfunction after large volume paracentesis or to diag-
nose and treat hepatorenal syndrome.
Beside the high proportion of inappropriate use, the elevated cost, • HSA is also the second-line treatment for fluid resuscitation in critical-
the theoretical risk of disease transmission and the existence of more ly ill patients when crystalloids and non-proteic colloids are not effec-
economical alternatives of comparable efficacy have prompted several tive or contra-indicated. Among the heterogeneous population of ICU
clinical and economical evaluations aiming to rationalize and render patients, accumulating data indicate that those with sepsis, severe
more appropriate the use of HSA [69–73]. sepsis, and septic shock benefit more from HSA administration.

Table 2
Practical recommendations for the prescription of human serum albumin at the S. Orsola-Malpighi University Hospital, Bologna, Italy [69].

Acute diseases First-line treatment Second-line treatment

Hypovolemic shock Colloid/crystalloid solutions Human albumin if:


• Sodium intake restriction
• Hypersensitivity to colloid or crystalloids
• Lack of response to combined use of colloids and crystalloid
Major surgery: Colloid/crystalloid solutions Human albumin if:
• Cardiovascular • Lack of response to combined use of colloid/crystalloid
• Other surgery
Burns Colloid/crystalloid solutions Human albumin plus crystalloid solutions if:
• Lack of response to colloid or crystalloid solutions alone
• Severe burns (>50% body surface)
Paracentesis Human albumin 8 g/l of removed ascites if paracentesis >4 l
Spontaneous bacterial peritonitis Human albumin 1.5 g/kg at diagnosis and 1 g/kg on third
day + antibiotic therapy
Hepatorenal syndrome Human albumin 1 g/kg at diagnosis followed by
20–40 g/die + vasoconstrictors
Ascites Diuretic treatment Human albumin if:
• Ascites resistant to diuretics
Plasmapheresis Human albumin if plasma changes >20 ml/kg per week
Protein wasting enteropathy/malnutrition Enteral or parenteral nutrition Human albumin if:
• Severe diarrhea (>2 l/die)
• Albuminemia b 2 g/dl
• Clinical hypovolemia
P. Caraceni et al. / European Journal of Internal Medicine 24 (2013) 721–728 727

• A large proportion of the HSA use is not supported by solid scientif- [22] Qiao R, Siflinger-Birnboim A, Lum H, Tiruppathi C, Malik AB. Albumin and Ricinus
communis agglutinin decrease endothelial permeability via interactions with matrix.
ic evidence. Most of this inappropriate prescription derives from Am J Physiol 1993;265:C439–46.
the administration for nutritional interventions or for correcting [23] Lang JD, Figueroa M, Chumley P, Aslan M, Hurt J, Tarpey MM, et al. Albumin and
hypoalbuminemia per se (without hypovolemia), despite the exis- hydroxyethyl starch modulate oxidative inflammatory injury to vascular endo-
thelium. Anesthesiology 2004;100:51–8.
tence of solid data against a real benefit. [24] Henriksen JH, Siemssen O, Krintel JJ, Malchow-Møller A, Bendtsen F, Ring-Larsen
• Clinical practical recommendations should be implemented to rational- H. Dynamics of albumin in plasma and ascitic fluid in patients with cirrhosis.
ize the prescription of HSA and avoid its futile use. J Hepatol 2001;34:53–60.
[25] Rector WG, Ibarra F, Openshaw K, Hoefs JC. Ascites kinetics in cirrhosis: relation-
ship to plasma-ascites hydrostatic–oncotic balance and intensity of renal sodium
retention. J Lab Clin Med 1986;107:412–9.
Conflict of interests [26] Ginès P, Schrier RW. Renal failure in cirrhosis. N Engl J Med 2009;361:1279–90.
[27] Møller S, Henriksen JH. Cardiovascular complications of cirrhosis. Postgrad Med J
2009;85:44–54.
Paolo Caraceni has been a speaker for Grifols S.p.A. [28] Bernardi M, Maggioli C, Zaccherini G. Human albumin in the management of com-
Mauro Bernardi has been a speaker and consultant for Grifols Italia plications of liver cirrhosis. Crit Care 2012;16:211.
S.p.A, CLS Behring GNBH, PPTA Europe. [29] EASL clinical practice guidelines on the management of ascites, spontaneous bacterial
peritonitis, and hepatorenal syndrome in cirrhosis. J Hepatol 2010;53:397–417.
Marco Domenicali, Alessandra Tovoli, Lucia Napoli, Carmen Serena [30] Runyon BA. Management of adult patients with ascites due to cirrhosis: update
Ricci and Manuel Tufoni have no conflict of interest. 2012. Available from: http://www.aasld.org . [Accessed 2013, May 9].
[31] Ruiz-del-Arbol L, Monescillo A, Jimenéz W, Garcia-Plaza A, Arroyo V, Rodés J.
Paracentesis-induced circulatory dysfunction: mechanism and effect on hepatic
References hemodynamics in cirrhosis. Gastroenterology 1997;113:579–86.
[32] Ginès P, Titó L, Arroyo V, Planas R, Panés J, Viver J, et al. Randomized comparative
[1] Sugio S, Kashima A, Mochizuki S, Noda M, Kobayashi K. Crystal structure of human study of therapeutic paracentesis with and without intravenous albumin in cir-
serum albumin at 2.5 A resolution. Protein Eng 1999;12:439–46. rhosis. Gastroenterology 1988;94:1493–502.
[2] Fasano M, Curry S, Terreno E, Galliano M, Fanali G, Narciso P, et al. The extraordinary [33] Salerno F, Badalamenti S, Lorenzano E, Moser P, Incerti P. Randomized comparative
ligand binding properties of human serum albumin. IUBMB Life 2005;57:787–96. study of hemaccel vs. albumin infusion after total paracentesis in cirrhotic patients
[3] Fanali G, di Masi A, Trezza V, Marino M, Fasano M, Ascenzi P. Human serum albumin: with refractory ascites. Hepatology 1991;13:707–13.
from bench to bedside. Mol Aspects Med 2012;33:209–90. [34] Ginès A, Fernández-Esparrach G, Monescillo A, Vila C, Domènech E, Abecasis R, et al.
[4] Evans TW. Review article: albumin as a drug–biological effects of albumin Randomized trial comparing albumin, dextran 70, and polygeline in cirrhotic patients
unrelated to oncotic pressure. Aliment Pharmacol Ther 2002;16:6–11. with ascites treated by paracentesis. Gastroenterology 1996;111:1002–10.
[5] Nicholson JP, Wolmarans MR, Park GR. The role of albumin in critical illness. Br J [35] Sola-Vera J, Miñana J, Ricart E, Planella M, González B, Torras X, et al. Random-
Anaesth 2000;85:599–610. ized trial comparing albumin and saline in the prevention of paracentesis-
[6] Garcia-Martinez R, Caraceni P, Bernardi M, Gines P, Arroyo V, Jalan R. Albumin: induced circulatory dysfunction in cirrhotic patients with ascites. Hepatology
pathophysiologic basis of its role in the treatment of cirrhosis and its compli- 2003;37:1147–53.
cations. Hepatology 2013. http://dx.doi.org/10.1002/hep.26338 [[Epub ahead [36] Bernardi M, Caraceni P, Navickis RJ, Wilkes MM. Albumin infusion in patients under-
of print]. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23423799. going large-volume paracentesis: a meta-analysis of randomized trials. Hepatology
Accessed 2013, Mar 2]. 2012;55:1172–81.
[7] Jalan R, Schnurr K, Mookerjee RP, Sen S, Cheshire L, Hodges S, et al. Alterations in the [37] Rimola A, García-Tsao G, Navasa M, Piddock LJ, Planas R, Bernard B, et al. Diagno-
functional capacity of albumin in patients with decompensated cirrhosis is associated sis, treatment and prophylaxis of spontaneous bacterial peritonitis: a consensus
with increased mortality. Hepatology 2009;50:555–64. document. International Ascites Club. J Hepatol 2000;32:142–53.
[8] Oettl K, Stauber RE. Physiological and pathological changes in the redox state of [38] Follo A, Llovet JM, Navasa M, Planas R, Forns X, Francitorra A, et al. Renal impair-
human serum albumin critically influence its binding properties. Br J Pharmacol ment after spontaneous bacterial peritonitis in cirrhosis: incidence, clinical
2007;151:580–90. course, predictive factors and prognosis. Hepatology 1994;20:1495–501.
[9] Van Belle E, Dallongeville J, Vicaut E, Degrandsart A, Baulac C, Montalescot G. [39] Sort P, Navasa M, Arroyo V, Aldeguer X, Planas R, Ruiz-del-Arbol L, et al. Effect of
Ischemia-modified albumin levels predict long-term outcome in patients with intravenous albumin on renal impairment and mortality in patients with cirrhosis
acute myocardial infarction. The French Nationwide OPERA study. Am Heart J and spontaneous bacterial peritonitis. N Engl J Med 1999;341:403–9.
2010;159:570–6. [40] Tandon P, Garcia-Tsao G. Bacterial infections, sepsis, and multiorgan failure in cirrho-
[10] Bourdon E, Loreau N, Blache D. Glucose and free radicals impair the antioxidant sis. Semin Liver Dis 2008;28:26–42.
properties of serum albumin. FASEB J 1999;13:233–44. [41] Sigal SH, Stanca CM, Fernandez J, Arroyo V, Navasa M. Restricted use of albumin
[11] Faure P, Wiernsperger N, Polge C, Favier A, Halimi S. Impairment of the antioxidant for spontaneous bacterial peritonitis. Gut 2007;56:597–9.
properties of serum albumin in patients with diabetes: protective effects of metformin. [42] Terg R, Gadano A, Cartier M, Casciato P, Lucero R, Muñoz A, et al. Serum creatinine and
Clin Sci (Lond) 2008;114:251–6. bilirubin predict renal failure and mortality in patients with spontaneous bacterial peri-
[12] Wratten ML, Sereni L, Tetta C. Oxidation of albumin is enhanced in the presence of tonitis: a retrospective study. Liver Int 2009;29:415–9.
uremic toxins. Ren Fail 2001;23:563–71. [43] Poca M, Concepción M, Casas M, Alvarez-Urturi C, Gordillo J, Hernández-Gea V,
[13] Himmelfarb J, McMonagle E. Albumin is the major plasma protein target of oxidant et al. Role of albumin treatment in patients with spontaneous bacterial peritonitis.
stress in uremia. Kidney Int 2001;60:358–63. Clin Gastroenterol Hepatol 2012;10:309–15.
[14] Oettl K, Stadlbauer V, Petter F, Greilberger J, Putz-Bankuti C, Hallström S, et al. Oxida- [44] De Araujo A, De Barros Lopes A, Rossi G, Da Silva GV, Ananias P, Ness S, et al.
tive damage of albumin in advanced liver disease. Biochim Biophys Acta 2008;1782: Low-dose albumin in the treatment of spontaneous bacterial peritonitis: should
469–73. we change the standard treatment? Gut 2012;61:1371–2.
[15] Stauber R, Spindelboeck W, Haas J, Putz-Bankuti C, Stadlbauer V, Lackner C, et al. [45] Salerno F, Navickis RJ, Wilkes MM. Albumin infusion improves outcomes of patients
Human nonmercaptalbumin-2: a novel prognostic marker in chronic liver failure. with spontaneous bacterial peritonitis: a meta-analysis of randomized trials.Clin
Ther Apher Dial 2013. http://dx.doi.org/10.1111/1744-9987.12024 [[Epub ahead Gastroenterol Hepatol 2013;11:123–30 [e1].
of print]. Available from: http://http://onlinelibrary.wiley.com. Accessed 2013, [46] Salerno F, Gerbes A, Ginès P, Wong F, Arroyo V. Diagnosis, prevention and treatment
May 7]. of hepatorenal syndrome in cirrhosis. Gut 2007;56:1310–8.
[16] Sadler PJ, Tucker A, Viles JH. Involvement of a lysine residue in the N-terminal Ni2+ and [47] Sanyal AJ, Boyer T, Garcia-Tsao G, Regenstein F, Rossaro L, Appenrodt B, et al. A
Cu2+ binding site of serum albumins. Comparison with Co2+, Cd2+ and Al3+. Eur J randomized, prospective, double-blind, placebo-controlled trial of terlipressin
Biochem 1994;220:193–200. for type 1 hepatorenal syndrome. Gastroenterology 2008;134:1360–8.
[17] Sokołowska M, Wszelaka-Rylik M, Poznański J, Bal W. Spectroscopic and thermody- [48] Martín-Llahí M, Pépin M-N, Guevara M, Díaz F, Torre A, Monescillo A, et al.
namic determination of three distinct binding sites for Co(II) ions in human serum Terlipressin and albumin vs albumin in patients with cirrhosis and hepatorenal
albumin. J Inorg Biochem 2009;103:1005–13. syndrome: a randomized study. Gastroenterology 2008;134:1352–9.
[18] Keaney JF, Simon DI, Stamler JS, Jaraki O, Scharfstein J, Vita JA, et al. NO forms an [49] Ortega R, Ginès P, Uriz J, Cárdenas A, Calahorra B, De Las Heras D, et al. Terlipressin
adduct with serum albumin that has endothelium-derived relaxing factor-like therapy with and without albumin for patients with hepatorenal syndrome: results
properties. J Clin Invest 1993;91:1582–9. of a prospective, nonrandomized study. Hepatology 2002;36:941–8.
[19] Kim SB, Chi HS, Park JS, Hong CD, Yang WS. Effect of increasing serum albumin on [50] Angeli P, Volpin R, Gerunda G, Craighero R, Roner P, Merenda R, et al. Reversal of type
plasma D-dimer, von Willebrand factor, and platelet aggregation in CAPD patients. 1 hepatorenal syndrome with the administration of midodrine and octreotide.
Am J Kidney Dis 1999;33:312–7. Hepatology 1999;29:1690–7.
[20] Kitano H, Fukui H, Okamoto Y, Kikuchi E, Matsumoto M, Kikukawa M, et al. Role of [51] Alessandria C, Ottobrelli A, Debernardi-Venon W, Todros L, Cerenzia MT, Martini S,
albumin and high-density lipoprotein as endotoxin-binding proteins in rats with et al. Noradrenalin vs terlipressin in patients with hepatorenal syndrome: a prospec-
acute and chronic alcohol loading. Alcohol Clin Exp Res 1996;20:73A–6A. tive, randomized, unblinded, pilot study. J Hepatol 2007;47:499–505.
[21] Chen T-A, Tsao Y-C, Chen A, Lo G-H, Lin C-K, Yu H-C, et al. Effect of intravenous [52] Gluud LL, Christensen K, Christensen E, Krag A. Terlipressin for hepatorenal syn-
albumin on endotoxin removal, cytokines, and nitric oxide production in patients drome. Cochrane Database Syst Rev 2012;9:CD005162. http://dx.doi.org/10.1002/
with cirrhosis and spontaneous bacterial peritonitis. Scand J Gastroenterol 14651858.CD005162.pub3 [Available from: http://www.ncbi.nlm.nih.gov/pubmed/
2009;44:619–25. 22972083. Accessed 2013, May 2].
728 P. Caraceni et al. / European Journal of Internal Medicine 24 (2013) 721–728

[53] Gentilini P, Casini-Raggi V, Di Fiore G, Romanelli RG, Buzzelli G, Pinzani M, et al. [64] Delaney A, Dan A, McCaffrey J, Finfer S. The role of albumin as a resuscitation fluid
Albumin improves the response to diuretics in patients with cirrhosis and ascites: for patients with sepsis: a systematic review and meta-analysis. Crit Care Med
results of a randomized, controlled trial. J Hepatol 1999;30:639–45. 2011;39:386–91.
[54] Romanelli R-G, La Villa G, Barletta G, Vizzutti F, Lanini F, Arena U, et al. Long-term al- [65] Dellinger RP, Levy MM, Rhodes A, Annane D, Gerlach H, Opal SM, et al. Surviving
bumin infusion improves survival in patients with cirrhosis and ascites: an unblind- sepsis campaign: international guidelines for management of severe sepsis and
ed randomized trial. World J Gastroenterol 2006;12:1403–7. septic shock, 2012. Intensive Care Med 2013;39:165–228.
[55] Guevara M, Terra C, Nazar A, Solà E. Albumin for bacterial infections other than spon- [66] Bañares R, Nevens F, Larsen FS, Jalan R, Albillos A, Dollinger M, et al. Extracorporeal al-
taneous bacterial peritonitis in cirrhosis. A randomized, controlled study. J Hepatol bumin dialysis with the molecular adsorbent recirculating system in acute-on-chronic
2012;57:759–65. liver failure: the RELIEF trial. Hepatology 2012;57:1153–62.
[56] Ginès P, Guevara M. Hyponatremia in cirrhosis: pathogenesis, clinical significance, [67] Sen S, Williams R, Jalan R. Emerging indications for albumin dialysis. Am J
and management. Hepatology 2008;48:1002–10. Gastroenterol 2005;100:468–75.
[57] Human albumin administration in critically ill patients: systematic review of [68] Schaefer B, Schmitt CP. The role of molecular adsorbent recirculating system dialysis
randomised controlled trials. Cochrane Injuries Group Albumin Reviewers. BMJ for extracorporeal liver support in children.Pediatr Nephrol 2012 [[in press; Epub
1998;317:235–40. ahead of print]. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23179193.
[58] Wilkes MM, Navickis RJ. Patient survival after human albumin administration. A Accessed 2013, May 6].
meta-analysis of randomized, controlled trials. Ann Intern Med 2001;135:149–64. [69] Mirici-Cappa F, Caraceni P, Domenicali M, Gelonesi E, Benazzi B, Zaccherini G, et al.
[59] Alderson P, Bunn F, Lefebvre C, Li WPA, Li L, Roberts I, et al. Human albumin solution How albumin administration for cirrhosis impacts on hospital albumin consumption
for resuscitation and volume expansion in critically ill patients.Cochrane Database and expenditure. World J Gastroenterol 2011;17:3479–86.
Syst Rev 2004:CD001208 [Available from: http://www.ncbi.nlm.nih.gov/pubmed/ [70] Tanzi M, Gardner M, Megellas M, Lucio S, Restino M. Evaluation of the appropriate
15495011. Accessed 2013, May 2]. use of albumin in adult and pediatric patients. Am J Health Syst Pharm 2003;60:
[60] Finfer S, Bellomo R, Boyce N, French J, Myburgh J, Norton R. A comparison of albumin 1330–5.
and saline for fluid resuscitation in the intensive care unit. N Engl J Med 2004;350: [71] Tarín Remohí MJ, Sánchez Arcos A, Santos Ramos B, Bautista Paloma J, Guerrero
2247–56. Aznar MD. Costs related to inappropriate use of albumin in Spain. Ann Pharmacother
[61] Human albumin solution for resuscitation and volume expansion in critically ill 2000;34:1198–205.
patients.Cochrane Database Syst Rev 2011:CD001208. http://dx.doi.org/10.1002/ [72] Debrix I, Combeau D, Stephan F, Benomar A, Becker A. Clinical practice guidelines
14651858.CD001208.pub3 [Available from: http://www.ncbi.nlm.nih.gov/pubmed/ for the use of albumin: results of a drug use evaluation in a Paris hospital. Tenon
21975732. Accessed 2013, May 3]. Hospital Paris. Pharm World Sci 1999;21:11–6.
[62] Myburgh J, Cooper DJ, Finfer S, Bellomo R, Norton R, Bishop N, et al. Saline or albumin [73] Vargas E, De Miguel V, Portolés A, Avendaño C, Ambit MI, Torralba A, et al. Use of
for fluid resuscitation in patients with traumatic brain injury. N Engl J Med 2007;357: albumin in two Spanish university hospitals. Eur J Clin Pharmacol 1997;52:
874–84. 465–70.
[63] Finfer S, McEvoy S, Bellomo R, McArthur C, Myburgh J, Norton R. Impact of albumin
compared to saline on organ function and mortality of patients with severe sepsis.
Intensive Care Med 2011;37:86–96.

You might also like