Вы находитесь на странице: 1из 13

Anaesthesia 2015, 70 (Suppl. 1), 2937 doi:10.1111/anae.

12891

Review Article
The pathophysiology and consequences of red blood cell storage
D. Orlov1 and K. Karkouti2

1 Clinical Fellow, 2 Associate Professor, Department of Anesthesia and Pain Management, Toronto General Hospital,
University Health Network, University of Toronto, Toronto, Ontario, Canada

Summary
Red cell transfusion therapy is a common treatment modality in contemporary medical practice. Although blood col-
lection and administration is safer and more efcient than ever before, red cells undergo multiple metabolic and
structural changes during storage that may compromise their functionality and viability following transfusion. The
clinical relevance of these changes is a hotly debated topic that continues to be a matter of intense investigation. In
the current review, we begin with an in-depth overview of the pathophysiological mechanisms underlying red cell
storage, with a focus on altered metabolism, oxidative stress and red cell membrane damage. We proceed to review
the current state of evidence on the clinical relevance and consequences of the red cell storage lesion, while discuss-
ing the strengths and limitations of clinical studies.
.................................................................................................................................................................
Correspondence to: K. Karkouti; Email: keyvan.karkouti@uhn.ca; Accepted: 8 August 2014

Introduction moglobin in the unit, and that at least 75% of the red
Modern blood banking represents a major achievement cells survive for more than 24 h post-transfusion [3,
in the eld of medicine. In less than 500 years, we 4]. These criteria, which have not been substantially
have progressed from vein-to-vein xeno-human trans- altered since the 1940s [5], are somewhat arbitrary,
fusions that were nearly uniformly fatal (to both par- and do not consider whether transfusing better func-
ties), to a highly procient system that supplies a wide tioning and more viable fresh red cells have any clini-
array of remarkably safe blood components for an cal advantages over transfusing older red cells.
expanding number of indications. Several pivotal Nevertheless, based on these criteria, the allowable
discoveries have contributed to this advance, one of storage duration has increased from 21 days in the
which was the development of storage mediums in the 1940s to 42 days or higher with the currently available
1940s that allowed red blood cells to be stored for pro- storage mediums [1]. Over the past decade, the detri-
longed periods while maintaining much (but not all) mental effects of storage on red cell functionality and
of their functionality and viability. viability have come under greater scrutiny [6], raising
Modern storage mediums for the most part consist questions about the clinical relevance of the storage
of various combinations of saline, adenine, glucose/ lesion and the appropriateness of currently approved
dextrose, phosphate and mannitol [1, 2]. The primary storage durations. In this narrative review, we will
regulatory criteria for setting the maximum amount of explore this issue by outlining the pathophysiological
time that red cells can be stored (refrigerated at changes that red cells undergo during storage, and the
4  2 C) using these mediums are that the amount mechanisms by which these changes may lead to
of haemolysis during storage be < 1% of the total hae- adverse clinical outcomes. We will also review the

2014 The Association of Anaesthetists of Great Britain and Ireland 29


Anaesthesia 2015, 70 (Suppl. 1), 2937 Orlov and Karkouti | Storage of red cells

current state of evidence on the clinical relevance of Altered RBC metabolism


the red cell storage lesion. Most cells meet their metabolic needs by oxidative
phosphorylation of glucose, but red cells lack mito-
Pathophysiological changes during red chondria and therefore have to rely on anaerobic gly-
cell storage colysis to generate metabolites, primarily adenosine
The function of red cell storage is to maintain the triphosphate (ATP), 2,3-diphosphoglycerate (2,3-DPG),
functionality and viability of red cells throughout the and reduced nicotinamide adenine dinucleotide
approved storage period [7]. The difculty that is (NADH) [8]. Adenosine triphosphate is the energy
shared by modern storage mediums, however, is that source for red cells numerous biochemical reactions,
red cell functionality and viability are progressively 2,3-DPG regulates the afnity of haemoglobin to oxy-
impaired during storage by three interrelated mecha- gen, and NADH is an important co-factor that reverses
nisms: altered metabolism; increased oxidative stress; the spontaneous oxidation of oxyhaemoglobin to meth-
and membrane damage (Fig. 1). aemoglobin within the red cell (see below) [8, 9].

Figure 1 Red cell changes associated with storage. Under normal conditions (left panel), the process of glycolysis
converts glucose to lactate, with adenosine triphosphate (ATP), 2,3-diphosphoglycerate (2,3-DPG), reduced nicotin-
amide adenine dinucleotide (NADH) and nicotinamide adenine dinucleotide phosphate (NADPH) formed as by-
products. Auto-oxidation of oxyhaemoglobin to methaemoglobin (dotted-arrow), with resultant production of hae-
min, iron and reactive oxygen species, occurs very slowly and is of limited signicance. The structure and orientation
of membrane protein anion exchanger-1 (AE1), ankyrin and spectrin, as well as phosphatidylserine (PS), maintain
the integrity of the red cell membrane. With prolonged storage (right panel), the accumulation of lactate inhibits
glycolysis. Reduced ATP further inhibits glycolysis, whereas depletion of 2,3-DPG attenuates the ability for oxygen
delivery. Reduced ATP and cold-storage impair the exchange of sodium (Na+) and potassium (K+) across the mem-
brane, resulting in increased intracellular Na+, which affects cell volume and shape. Reduced NADH content pro-
motes auto-oxidation and formation of methaemoglobin, whereas reduction of NADPH decreases production of
reduced glutathione (GSH), which is a potent cytosolic antioxidant. Clustering of AE1 compromises red cell mem-
brane integrity, whereas externalisation of PS acts as a senescence signal. Increased formation of microvesicles in the
supernatant decreases endothelial-derived nitric oxide (NO) gas, which is a potent vasodilator and antioxidant.
Finally, elevated levels of free haemoglobin (Hb) and iron in the supernatant released from haemolysed cells contrib-
ute to oxidative stress. Together, these changes reduce red cell viability (pre- and post-transfusion) and functionality
(post-transfusion).

30 2014 The Association of Anaesthetists of Great Britain and Ireland


Orlov and Karkouti | Storage of red cells Anaesthesia 2015, 70 (Suppl. 1), 2937

Cold-storage of red cells at 4  2 C helps main- whereas outside, they are approximately 5 and
tain red cell functionality and viability by reducing 140 mM, respectively [13]. By the sixth week of stor-
the red cell metabolic rate. For each one degree drop age, red cell potassium content is decreased by about
in storage temperature, there is approximately a 10% 40% and sodium content is increased by about 300%
decrease in red cell metabolic rate [10], and at 4 C, [16]. At the same time, the potassium content in the
the metabolic rate is ten times lower than at 25 C supernatant of the stored units is markedly increased,
[11]. As metabolic activity does not completely cease which may lead to hyperkalaemia after transfusion [13,
when red cells are stored, glucose or dextrose are 17, 21]. Increased intracellular sodium content affects
added to storage mediums to allow red cells to con- cell volume and shape, such that the mean corpuscular
tinue glycolysis and thus produce sufcient ATP, 2,3- volume of stored red cells is increased after 3 weeks of
DPG and NADH to maintain adequate functionality storage [21]. This effect is more pronounced in older
and viability during storage. Continued glycolysis, red cells, impairing their deformability and viability
however, results in the accumulation of its primary [16, 25].
by-product, lactic acid, in the supernatant [8]. The
resulting acidosis inhibits glycolysis via a negative Increased red cell oxidative stress
feedback loop [8, 12, 13], which results in a progres- For haemoglobin to be able to reversibly bind oxygen
sive reduction in ATP, 2,3-DPG and NADH levels (oxyhaemoglobin deoxyhaemoglobin) within the
[1317]. By the sixth week of storage, lactate levels in red cell, its component haeme-irons must be main-
the supernatant are increased by several fold, the pH tained in their reduced, or ferrous (Fe2+), form. Under
is below 6.5, the ATP and NADH contents are sub- normal circumstances, a small amount of oxyhaemo-
stantially reduced, and 2,3-DPG content is depleted globin undergoes spontaneous oxidation, generating
[1521]. Reduced ATP levels impair all red cell meta- methaemoglobin (which has oxidised or ferric (Fe3+)
bolic activities including glycolysis itself (as ATP is iron and cannot bind oxygen) and reactive oxygen
needed for the initial steps of glycolysis, creating a species [24, 2628]. Methaemoglobin is inherently
vicious cycle), formation of cytosolic antioxidants unstable and is denatured into globin and, more
(thus reducing antioxidant defences), and mainte- importantly, haemin (also known as ferric or oxidised
nance of membrane integrity (thereby reducing red haeme) [8], some of which may in turn be catabolised
cell deformability and promoting alterations in their by erythroid haeme-oxygenase-1 to release its iron
discoid shape) [8, 22]. Although there exists a direct [29]. Free haemin and iron, in conjunction with reac-
relationship between reduced ATP levels and red cell tive oxygen species, can generate highly hazardous
viability, the overall role of ATP depletion in the hydroxyl radicals that can cause oxidative injury to
storage lesion seems to be limited [22, 23]. Deple- membrane lipids and proteins (see below) [9, 27, 30].
tion of 2,3-DPG reduces oxygen delivery, but levels Under normal circumstances, red cells are protected
are rapidly replenished after transfusion [8]. The against this oxidative injury because the rate of sponta-
most important contribution to the storage lesion is neous oxidation of haemoglobin is slow [24, 31], the
likely to be the depletion of reduced NADH, which NADH-dependent cytochrome-b5 reductase (CYTb5)
impairs the conversion of methaemoglobin back to reduces methaemoglobin back into oxyhaemoglobin,
oxyhaemoglobin within the red cell, thereby aggravat- and cytosolic antioxidants (primarily reduced glutathi-
ing oxidative stress (see next section) [9, 24]. one or GSH) and membrane anti-oxidants (primarily
Another effect of cold-storage is that it impairs the ascorbic acid or vitamin C) neutralise the generated
exchange of sodium and potassium across the red cell reactive oxygen species [9, 24, 26, 27, 32].
membrane by the sodium/potassium adenosine tri- All of these protective mechanisms are impaired
phosphatase (Na+/K+ ATPase) pump [13, 16]. Under during storage. Spontaneous oxidation of haemoglobin
normal circumstances, the concentration of potassium to methaemoglobin increases under conditions of
and sodium inside the red cell are maintained at increased oxygen partial pressure and acidosis [31],
approximately 90 mM and 5 mM, respectively, both of which are present during storage [9]. As a

2014 The Association of Anaesthetists of Great Britain and Ireland 31


Anaesthesia 2015, 70 (Suppl. 1), 2937 Orlov and Karkouti | Storage of red cells

result, the rate of this reaction is substantially rapid clearance of the red cell from the circulation
increased during storage [24, 26, 27, 33]. Moreover, [38].
due to the metabolic changes outlined earlier, levels of Metabolic alterations due to cold-storage and
NADH, GSH and ascorbic acid are markedly reduced reduced glycolysis as well as increased oxidative stress
during this period [18, 34, 35]. Increased spontaneous have profoundly deleterious effects on the red cell
oxidation of haemoglobin in the setting of reduced membrane. Increased auto-oxidation of haemoglobin
antioxidant defences exposes red cells to increased oxi- within the red cell leads to precipitation of structurally
dative stress, which is the predominant cause of red distorted forms of methaemoglobin (known as haemi-
cell membrane damage (see below) [9, 18, 3638]. chromes) near the cell membrane, and causes disrup-
tion of AE1 and cytoskeletal membrane proteins [8, 9].
Red cell membrane damage Denaturation of methaemoglobin results in the forma-
Red cell functionality and viability is critically depen- tion of haemin and iron, which freely partition into
dent on the integrity of the red cell membrane, to the membrane lipid bilayer to cause peroxidation of
maintain normal cell shape, deformability and membrane lipids, proteins, and carbohydrates [9, 20].
mechanical stability [37]. Storage-related alterations in Membrane disruption and peroxidation causes mem-
metabolism and oxidative stress have considerable del- brane cation leak, increased cholesterol to plasma
eterious effects on red cell membrane integrity. ratios, phosphatidylserine externalisation, clustering of
The red cell membrane consists of a lipid bilayer AE1, and increases production of microvesicles. These
that is interspersed with proteins. The lipid bilayer changes reduce red cell functionality and viability by
includes phospholipids, cholesterol and fatty acids that causing membrane instability and loss, reduced de-
are asymmetrically distributed between the inner and formability, alterations in red cell discoid shape and
outer layers [8]. Phosphatidylserine is an important increased senescence signalling [9, 10, 1820, 36, 37,
component of the lipid layer that under normal cir- 4042]. Microvesicle formation and accumulation in
cumstances is present entirely on the inner layer, but the supernatant increases exponentially during storage
in senescent red cells, it is expressed on the outer layer [19, 21, 4346]. The accumulated microvesicles, which
of the membrane [8]. When phosphatidylserine is are haemoglobin-laden and externalise phosphatidyl-
expressed on the outer layer, it is highly thrombogenic serine, can also cause post-transfusion physiological
and also acts as an important senescence signal that perturbations by contributing to the oversaturation of
leads to the removal of red cells by reticuloendothelial the bodys clearance systems for haemolysed red cells
macrophages [38, 39]. Cholesterol is another important (discussed below), by increasing thrombogenicity due
component of the lipid layer as increased cholesterol to externalisation of phosphatidylserine, and by scav-
to phospholipid ratios impair red cell viscosity and de- enging of endothelial-derived nitric oxide (NO) gas
formability, and promote alterations in red cell shape which is a potent vasodilator, antioxidant and inhibitor
[8]. Important membrane proteins include the trans- of platelet aggregation by its component haemoglo-
membrane protein anion exchanger 1 (AE1, also bin [24, 41, 4648]. When haemoglobin comes in con-
known as band 3), and the primary structural cytoskel- tact with NO, it near instantaneously consumes the
etal proteins spectrin and ankyrin. The AE1 is a trans- NO to form methaemoglobin and nitrate [24, 48].
port protein that regulates the exchange of chloride Under normal circumstances, encapsulation of haemo-
and bicarbonate across the membrane and also links globin within the red cell prevents this reaction
the lipid bilayer to the cytoskeleton by binding to because NO does not diffuse well across the red cell
ankyrin, which in turn binds to spectrin [8]. Normal membrane [24, 48]. It does readily permeate across the
protein organisation is crucial for maintaining red cell microvesicular membrane, such that the rate of reac-
stability, deformability and shape [8]. AE1 is also tion between haemoglobin and NO is increased by
involved in senescence signalling, as its breakdown (or 1000-fold [49]. Thus, the accumulated microvesicles in
clustering) generates a neo-antigen that results in the the supernatant can reduce endothelial-derived NO

32 2014 The Association of Anaesthetists of Great Britain and Ireland


Orlov and Karkouti | Storage of red cells Anaesthesia 2015, 70 (Suppl. 1), 2937

bioavailability in the recipient, which can lead to tissue Under normal circumstances, approximately 1% of
ischaemia and end-organ injury [24, 4852]. the bodys 2.5 9 1013 circulating red cells
(5 9 106.ll 1 of blood) become senescent and are
Reduced red cell viability cleared from the circulation each day (which works
The nal common pathway of injury due to the stor- out to approximately 1 x 1010 cells per hour), with the
age-related alterations in red cell metabolism, increased majority (8090%) undergoing extravascular haemoly-
oxidative stress and membrane damage may be reduced sis in the macrophages of the reticuloendothelial
red cell viability resulting in pre- and post-transfusion system and the remaining 1020% undergoing intra-
haemolysis.[53] Pre-transfusion haemolysis leads to the vascular haemolysis [8] (Fig. 2). When red cells are
release of red cell contents in the supernatant, which destroyed in the intravascular space, the released
leads to an approximately eightfold increase in the free haemoglobin is rapidly bound to haptoglobin and
haemoglobin levels in the supernatant after 6 weeks of removed from the circulation by macrophages via
storage [20, 32, 49, 51, 54, 55]. As a result of similar receptor-mediated endocystosis [8, 5962]. The scav-
processes that occur within the red cell (outlined enged haemoglobin is then safely degraded within the
above), some of the free haemoglobin in the superna- macrophages as described below. Once the haptoglobin
tant undergoes spontaneous oxidation, resulting in the scavenging capacity has been exceeded, the remaining
accumulation of methaemoglobin, haemin, iron and free haemoglobin scavenges endothelial-derived NO to
reactive oxygen species in the supernatant, thus aggra- form methaemoglobin [24, 48, 52]. As noted above,
vating the oxidative stress that red cells are exposed to reduced NO bioavailability can cause endothelial dys-
during storage [49]. Methaemoglobin levels, which nor- function, platelet aggregation and oxidative injury,
mally constitute < 1% of total haemoglobin in the which can lead to tissue ischaemia and end-organ
supernatant [8], are increased by twofold after 3 weeks injury [48, 49, 52]. The formed methaemoglobin is in
of storage [56]. Free iron, which is essentially undetect- turn converted to free haemin, which is then rapidly
able during the rst few days of storage, is ubiquitous cleared from the circulation and transferred to the
in the supernatant after 3 weeks of storage [35, 57]. liver by haemopexin and albumin, thereby protecting
Similar to the post-transfusion effects of microvesicles against its potent pro-oxidant and pro-inammatory
(outlined above), the contents of the supernatant may effects [8, 6164]. Thus, intravascular clearance of
also reduce endothelial-derived NO bioavailability and haemolysed red cells protects against the toxic effects
contribute to the oversaturation of the clearance of free haemoglobin and haemins at the cost of
systems for haemolysed red cells in the recipient. How- reduced NO bioavailability.
ever, given that the amount of free haemoglobin in the Extravascular haemolysis involves the reticuloendo-
supernatant constitutes < 1% of the total haemoglobin thelial macrophages that recognise and phagocytose not
in a unit of blood [1921], this contribution is likely to only senescent red cells, but also the haptoglobin-hae-
be minor. moglobin and haemopexin-haemin complexes that are
A much more onerous burden on the recipient is formed during intravascular haemolysis as well as any
likely to be the progressively increasing number of red haemoglobin-laden microvesicles [8, 41]. Once inside
cells that become senescent but do not undergo haem- the macrophages, the scavenged haemoglobin and hae-
olysis during storage. These comprise nearly 25% of mins are catalysed by haeme-oxygenase-1 to carbon
the total cells in a unit of blood by the fourth week of monoxide, biliverdin (a precursor of bilirubin), and
storage [5, 17, 58], and are haemolysed and removed most importantly, iron [8, 65]. The majority of the
from the recipients circulation within 1 h of transfu- recovered iron is buffered and stored within the macro-
sion [17]. Their rapid removal may overwhelm the phage in ferritin, and the remainder is released into the
normal systems of haemolysis that include several circulation where it is bound by the iron-carrier protein
highly efcient but saturable pathways for clearing the transferrin [8, 66]. Excessive extravascular haemolysis
contents of red cells from the circulation, thereby cur- can lead to the presence of highly toxic free iron inside
tailing their toxic effects. the macrophages as well as in the circulation.

2014 The Association of Anaesthetists of Great Britain and Ireland 33


Anaesthesia 2015, 70 (Suppl. 1), 2937 Orlov and Karkouti | Storage of red cells

Senescent RBCs
Normally 80-90% Normally 10-20%

Extravascular Haemolysis Intravascular Haemolysis

Free Hb
Free Hb

Haeme NO

2NO3-
Iron
+ Met-Hb (Fe3+)
CO
+
Biliverdin Haemin

Macrophage
Microvesicles

Figure 2 Haemolytic pathways of senescent red cells following transfusion. Following red cell transfusion, intravas-
cular haemolysis results in the formation of free haemoglobin (Hb) which binds to haptoglobin and is delivered to
macrophages for safe degradation. If this system is saturated, free Hb may form methaemoglobin (Met-Hb) and hae-
min at the expense of endothelial nitric oxide (NO) consumption. Haemin a known pro-oxidant and pro-inam-
matory mediator is further scavenged by haemopexin and albumin and delivered to macrophages for processing.
Extravascular haemolysis occurs within reticuloendothelial macrophages and results in the production of iron, carbon
monoxide and biliverdin from the breakdown of free Hb. Outside of the macrophage, free iron binds to transferrin
before transport. Macrophages are further responsible for phagocytosis and degradation of haemoglobin-laden micro-
vesicles. The combination of macrophage over-activation and overwhelmed safety mechanisms (pink shading) may
contribute to end-organ dysfunction secondary to oxidative injury (from accumulation of free iron, free Hb, and
haemin) and tissue ischaemia (from vasoconstriction due to NO-consumption).

As noted above, under normal circumstances, decreased endothelial-derived NO bioavailability and


the systems of intravascular and extravascular haem- the presence of free haemoglobin, haemin and iron
olysis clear approximately 1 9 1010 red cells per in the recipients circulation as well as within the
hour. A single unit of blood that has been stored reticuloendothelial macrophages [48, 49, 55, 6772].
for more than 4 weeks, however, contains approxi- The potential clinical consequences of these effects
mately 1.5 9 1012 red cells, of which about 25%, or are many, and include: increased risk of infection
approximately 4 9 1011, will have become senescent due to macrophage activation or dysfunction and
during storage and are cleared from the circulation cytokine release; tissue ischaemia due to endothelial
within 1 h of transfusion [17]. Moreover, each unit dysfunction; and end-organ dysfunction due to oxi-
also contains a large number of haemoglobin-laden dative injury [28, 41, 49, 54, 55, 61, 67, 69, 71, 73
microvesicles, free haemoglobin, haemin and iron in 79]. Whether or not these effects are clinically rele-
the supernatant that also need to be rapidly cleared vant remains unproven and a matter of intense
from the circulation. Since the number of senescent investigation.
red cells alone are one order of magnitude greater
than the number that are cleared per hour under Current state of clinical evidence on
normal circumstances, it is quite possible that trans- the clinical relevance of the red cell
fusing a single unit of old blood may oversaturate storage lesion
the protective systems of extravascular and intravas- There has been a plethora of studies investigating the
cular haemolysis in some patients, leading to effects of prolonged blood storage on clinical out-

34 2014 The Association of Anaesthetists of Great Britain and Ireland


Orlov and Karkouti | Storage of red cells Anaesthesia 2015, 70 (Suppl. 1), 2937

comes. These have included healthy volunteers [67, Human observational studies
8082], numerous retrospective and prospective No study subtype is more ubiquitous in the realm of
observational studies [77, 83114], and a handful of red cell storage research than human observational
older [115123], more recent [124], and ongoing studies. Indeed, we have come a long way from the
randomised controlled trials. Overall, study results seminal investigation by Purdy et al. [99, 125], with
have been conicting and affected by multiple meth- upwards of thirty observational studies published in
odological issues including confounding, systematic the literature [77, 83114], and multiple recent system-
bias, limited external validity and multiple statistical atic reviews [1, 126131] attempting to consolidate this
shortcomings. data. All but one of these reviews [130] have not sup-
ported the hypothesis that transfusion of older red
Volunteer studies cells is worse than that of fresh red cells. The review
Human volunteer studies have examined the effect of by Wang et al. [130] was the only one that found a
blood storage on post-transfusion physiological vari- signicant association between transfusion of older red
ables, including anaemia-induced cognitive dysfunc- cells and mortality, but this review included several
tion [81], extravascular haemolysis [67], serum iron primary studies that are evidently confounded [129].
elevation [67], propensity for bacteraemia (evaluated In all, no rm conclusions can be drawn from existing
in vitro) [67], attenuation of NO-mediated hypera- studies owing to important limitations in their design
emia [80] and gas exchange [82]. All of these studies or analyses.
involved cross-over designs where autologous blood Observational studies are fraught with numerous
was used for transfusion following both a short (any- potential sources of bias that may hinder their ability
where from 34 h to 37 days) and prolonged (any- to answer the research question. For instance, in the
where from 23 to 42 days) storage duration. In all absence of randomisation, sampling bias might occur,
cases, each subject participated in both study arms, such that older red cells are predominantly transfused
thereby serving as their own control. Of all these to sicker patients or those with a more precarious
parameters, only transient increases in extravascular peri-operative course, based on a combination of
haemolysis and serum iron elevations were observed increased demand and institutional pressures on blood
in relation to storage latency [67], supporting the piv- banks for efciency and subsequent utilisation of the
otal role of reduced red cell viability in the storage rst in-rst out principle for cost-containment. If a
lesion. causal association between storage latency and adverse
Volunteer studies have the advantage of investigat- outcomes does indeed exist, the presence of this bias
ing outcomes in humans while achieving great tempo- would overestimate this effect. Similarly, although a
ral separation between the study arms to allow for multicentre, international, observational study would
maximal investigation of any treatment effect. Never- improve the power of an analysis, its results should
theless, they suffer from a myriad of drawbacks which not be analysed in aggregate, independent of the coun-
warrant careful interpretation. Specically, volunteers try of origin. Given that the maximum duration of red
are frequently devoid of comorbidities, receive slow, cell storage is a function of the preservative solution
low-volume infusions of autologous blood (thereby which varies by country, unadjusted pooling of
minimising the potential adverse effects of volume national outcomes would render the results both non-
overload and immunomodulation, respectively), and generalisable and meaningless.
are unsuitable for studying certain outcomes such as Multiple confounders should also be considered
mortality. Moreover, inherent in a crossover study is when evaluating observational data. For example, in a
the potential for order and carry-over effects if tech- study of prolonged duration, the year of transfusion
niques such as counterbalancing and an adequate should be accounted for, as it may affect both storage
wash-out period are overlooked. Most importantly, latency (through advances in preservative solution) and
volunteer studies have limited external validity for outcomes (secondary to rapid advances in technology
many of the study populations of interest.

2014 The Association of Anaesthetists of Great Britain and Ireland 35


Anaesthesia 2015, 70 (Suppl. 1), 2937 Orlov and Karkouti | Storage of red cells

and standards of care). The nature of ABO-matching (8.3) days in the trial) red cell transfusions [124].
between donor and recipient should also be carefully Although robust in its design, this trial is limited in its
noted. Specically, Group O red cells are considered the external generalisability for two reasons. First, the pop-
universal donor, thereby decreasing their turnover time ulation was limited to premature infants a very
and inating the number of Group O transfusions in unique subgroup of patients, mainly cared for in spec-
the fresh blood subgroup of patients. It follows that the ialised institutions. Second, the standard-of-care arm
old transfusion subgroup usually contains a greater pro- still received relatively fresh blood only 12 infants
portion of non-O blood recipients, as demonstrated by received red cells that were exclusively more than
the largest and perhaps most inuential observational 14 days old. Given that the average blood storage in
study on the topic [90]. Due to the known association the US is 18 days [1], and certain centres routinely
of non-O blood subtypes with an increased rate of store red cells for longer than 21 days [117], these
venous thrombo-embolic events and possible increased results may not be applicable to the broader popula-
mortality [132134], the nature of ABO-matching may tion.
overestimate the causal relationships of interest in the Five other large RCTs have either been completed,
absence of appropriate statistical adjustment. Finally, or are ongoing. The age of blood evaluation study
the number of red cell transfusions is a parameter (ABLE; ISRCTN44878718) has completed recruiting
requiring special attention. For one, each additional and randomising 2510 ICU patients across Canada to
unit of transfused red cells simultaneously brings the receive either fresh ( 7 days old) or older (standard
average age of transfused red cells closer to the blood blood bank procedure of using the oldest blood units
bank mean, while selecting for a more critical patient rst, irrespective of age) red cells with the primary
population with increased likelihood of experiencing outcome being all-cause 90-day mortality. These
adverse outcomes. Moreover, this confounded direc- results are yet to be published. The red cell storage
tionality between storage latency and adverse outcomes duration and outcomes in cardiac surgery study
can be further inuenced by the precise denition of (NCT00458783) has been enrolling patients for 7 years
what constitutes fresh and old blood [89, 95, 114]. with a goal to recruit and randomise a total of 2800
primary or re-operative adult cardiac surgical patients
Randomised controlled trials (RCT) to receive either fresh (< 14 days old) or older
Although close to a dozen RCTs have been published (> 20 days old) red cells at a single centre, with the
on the relationship between storage duration and outcome being all-cause postoperative 30-day mortal-
adverse outcomes [115124], the majority have been ity. The red cell storage duration study (RECESS;
either small or not adequately powered to evaluate NCT00991341) has completed randomising 1696
mortality. Moreover, none of them have found any sig- patients undergoing complex cardiac surgery across 33
nicant differences in outcomes between fresh and old centres in the USA to receive either fresh ( 10 days
subgroups of transfusion recipients across a variety of old) or older ( 21 days old) red cells. The primary
clinical settings. A handful of large, ongoing RCTs, outcome is a change from baseline in the multi-organ
including one that has been completed and published dysfunction score at 7 days postoperatively. The
[124], aim to contribute valuable information to this informing fresh vs old red cell management study
area of interest. (INFORM; ISRCTN08118744) is continuing to rando-
One recently published double-blinded RCT that mise a projected 24 400 acute care inpatients across
included 377 premature infants found no difference in Canada, Australia and the US to receive either fresh
the rates of morbidity (necrotising enterocolitis, reti- (youngest available) or old (oldest product compatible
nopathy, bronchopulmonary dysplasia or intraventric- in stock) red cells, with the primary outcome being in-
ular haemorrhage), or mortality among those who hospital mortality. Finally, the standard issue transfu-
received fresh ( 7 days old as stated by protocol; sion vs fresher red blood cell use in intensive care
mean (SD) 5.1 (2.0) days in the trial) vs older (stan- study (TRANSFUSE; NCT01638416) is an interna-
dard practice as stated by protocol; mean (SD) 14.6 tional investigation set to randomise 5000 ICU patients

36 2014 The Association of Anaesthetists of Great Britain and Ireland


Orlov and Karkouti | Storage of red cells Anaesthesia 2015, 70 (Suppl. 1), 2937

to oldest vs freshest available red cells, with 90-day mechanisms underlying red cell storage, the answers to
mortality being the primary outcome. many fundamental questions continue to evade us.
These large-scale RCTs are likely to enhance the Although we have succeeded in prolonging the maxi-
state of knowledge and contribute to standard-of-care mum storage shelf-life of blood with advances in pre-
transfusion practice, whereas they are still subject to servative solution, the criteria on which this is based
numerous limitations. First, given the ethical con- are almost completely arbitrary and have remained
straints, it is unlikely that any randomised trial will stagnant since the 1940s. We are also no closer to
allow for a substantial inter-group contrast that reects delineating the temporal pattern of changes with red
the spectrum of the red cell storage shelf-life. This cell storage, nor, more importantly, the point at which
drawback substantially limits the power of an RCT these changes begin to manifest in clinically-signicant
due to both a small intervention effect, and also the sequelae following transfusion. We have likely only hit
unknown temporal trajectory of red cell degradation. the tip of the iceberg in beginning to identify predis-
A recent simulation study examined the power of mul- posing comorbidities and contexts that may worsen
tiple pre-existing RCT designs for ve hypothetical red prognosis following red cell transfusions of prolonged
cell degradation patterns [135]. Only a single pattern storage duration. All of these questions warrant system-
of red cell degradation occurring between 17 and atic investigation with a simultaneous understanding of
25 days allowed for adequate power of the RCTs in research design and its many limitations.
question. Second, it is becoming increasingly clear that The multiple ongoing, large-scale RCTs are a tes-
certain patient subpopulations might be more suscepti- tament to the importance of the storage lesion and its
ble to the effects of the red cell storage lesions com- potential effects on patient outcomes. Over the next
pared to others. These subpopulations including decade, ndings from these RCTs are likely to have
patients with an infection, renal failure, or those signicant implications for international transfusion
requiring a massive transfusion need to be dened a practice, policy makers, and regulatory bodies. Never-
priori, but could only be identied using alternative theless, it must be appreciated that all of the aforemen-
study designs. Finally, as with any longitudinal out- tioned study designs are complementary and
come measure (e.g. 90-day mortality), a lengthy follow invaluable for the holistic exploration of the broader
up may be resource-intensive, costly and may result in research question. Only through a large, concerted
multiple patient dropouts. In this case, adequate trial effort, will we be closer to understanding the patho-
funding, the indirect use of large administrative data- physiology and consequences of the red cell storage
bases or statistical imputation may be warranted to lesion in clinical practice.
maintain data integrity.
Acknowledgements
Conclusion KK is supported in part by a merit award from the
Over the past half-millennium, we have witnessed Department of Anesthesia, University of Toronto.
incredible progress in the efciency and safety of blood
transfusion therapy. Nevertheless, although we continue Competing interests
to acquire insight into the intricacies of metabolic No competing interests declared.

2014 The Association of Anaesthetists of Great Britain and Ireland 37


Anaesthesia 2015, 70 (Suppl. 1), e9e12 Orlov and Karkouti | Storage of red cells

References blood bank storage of packed red blood cells. Blood Trans-
1. Flegel WA, Natanson C, Klein HG. Does prolonged storage of fusion 2012; 10: 45361.
red blood cells cause harm? British Journal of Haematology 21. Salzer U, Zhu R, Luten M, et al. Vesicles generated during
2014; 165: 316. storage of red cells are rich in the lipid raft marker stoma-
2. Hardwick J. Blood storage and transportation. ISBT Science tin. Transfusion 2008; 48: 45162.
Series 2014; 3: 17796. 22. Almac E, Ince C. The impact of storage on red cell function
3. Hess JR. Scientific problems in the regulation of red blood in blood transfusion. Best Practice & Research Clinical Ana-
cell products. Transfusion 2012; 52: 182735. esthesiology 2007; 21: 195208.
4. Hess JR, Sparrow RL, van der Meer PF, Acker JP, Cardigan 23. Karger R, Lukow C, Kretschmer V. Deformability of red blood
RA, Devine DV. Red blood cell hemolysis during blood cells and correlation with ATP content during storage as
bank storage: using national quality management data to leukocyte-depleted whole blood. Transfusion Medicine and
answer basic scientific questions. Transfusion 2009; 49: Hemotherapy 2012; 39: 27782.
2599603. 24. Umbreit J. Methemoglobin its not just blue: a concise
5. Ross JF, Finch CA, Peacock WC, Sammons ME. The in vitro review. American Journal of Hematology 2007; 82: 13444.
preservation and post-transfusion survival of stored blood. 25. Hentschel WM, Wu LL, Tobin GO, et al. Erythrocyte cation
Journal of Clinical Investigation 1947; 26: 687703. transport activities as a function of cell age. Clinica Chimica
6. Ness PM. Does transfusion of stored red blood cells cause Acta 1986; 157: 3343.
clinically important adverse effects? A critical question in 26. Lutz HU, Bogdanova A. Mechanisms tagging senescent red
search of an answer and a plan. Transfusion 2011; 51: 666 blood cells for clearance in healthy humans. Fronteries in
7. Physiology 2013; 4: 387.
7. Hogman CF, Meryman HT. Storage parameters affecting red 27. Kanias T, Acker JP. Biopreservation of red blood cells the
blood cell survival and function after transfusion. Transfu- struggle with hemoglobin oxidation. FEBS Journal 2010;
sion Medicine Reviews 1999; 13: 27596. 277: 34356.
8. Greer P. Wintrobes Clinical Hematology, 13th edn. Philadel- 28. Buehler PW, Karnaukhova E, Gelderman MP, Alayash AI.
phia, PA: Lippincott Williams & Wilkins Publishers, 2014. Blood aging, safety, and transfusion: capturing the radical
9. Yoshida T, Shevkoplyas SS. Anaerobic storage of red blood menace. Antioxidants & Redox Signaling 2011; 14: 171328.
cells. Blood Transfusion 2010; 8: 22036. 29. Garcia-Santos D, Schranzhofer M, Horvathova M, et al.
10. Hess JR. Measures of stored red blood cell quality. Vox San- Heme oxygenase 1 is expressed in murine erythroid cells
guinis 2014; 107: 19. where it controls the level of regulatory heme. Blood 2014;
11. Ruddell JP, Lippert LE, Babcock JG, Hess JR. Effect of 24-hour 123: 226977.
storage at 25 degrees C on the in vitro storage characteris- 30. Hess JR. Red cell storage. Journal of Proteomics 2010; 73:
tics of CPDA-1 packed red cells. Transfusion 1998; 38: 424 36873.
8. 31. Reeder BJ. The redox activity of hemoglobins: from physio-
12. Graubarth H, Mackler B, Guest GM. Effects of acidosis on uti- logic functions to pathologic mechanisms. Antioxidants &
lization of glucose in erythrocytes and leucocytes. American Redox Signaling 2010; 13: 1087123.
Journal of Physiology 1953; 172: 3018. 32. Raval JS, Fontes J, Banerjee U, Yazer MH, Mank E, Palmer
13. Flatt JF, Bawazir WM, Bruce LJ. The involvement of cation AF. Ascorbic acid improves membrane fragility and
leaks in the storage lesion of red blood cells. Frontiers in decreases haemolysis during red blood cell storage. Transfu-
Physiology 2014; 5: 214. sion Medicine 2013; 23: 8793.
14. Tinmouth A, Fergusson D, Yee IC, Hebert PC. Clinical conse- 33. Balagopalakrishna C, Manoharan PT, Abugo OO, Rifkind
quences of red cell storage in the critically ill. Transfusion JM. Production of superoxide from hemoglobin-bound oxy-
2006; 46: 201427. gen under hypoxic conditions. Biochemistry 1996; 35:
15. Bennett-Guerrero E, Veldman TH, Doctor A, et al. Evolution 63938.
of adverse changes in stored RBCs. Proceedings of the 34. Jozwik M, Jozwik M, Jozwik M, Szczypka M, Gajewska J,
National Academy of Sciences USA 2007; 104: 170638. Laskowska-Klita T. Antioxidant defence of red blood cells
16. Minetti G, Ciana A, Profumo A, et al. Cell age-related mono- and plasma in stored human blood. Clinica Chimica Acta
valent cations content and density changes in stored 1997; 267: 12942.
human erythrocytes. Biochimica et Biophysica Acta 2001; 35. Collard K, White D, Copplestone A. The influence of storage
1527: 14955. age on iron status, oxidative stress and antioxidant protec-
17. Luten M, Roerdinkholder-Stoelwinder B, Schaap NP, de Grip tion in paediatric packed cell units. Blood Transfusion 2014;
WJ, Bos HJ, Bosman GJ. Survival of red blood cells after 12: 2109.
transfusion: a comparison between red cells concentrates 36. Mohanty JG, Nagababu E, Rifkind JM. Red blood cell oxida-
of different storage periods. Transfusion 2008; 48: 1478 tive stress impairs oxygen delivery and induces red blood
85. cell aging. Frontiers in Physiology 2014; 5: 84.
18. DAlessandro A, DAmici GM, Vaglio S, Zolla L. Time-course 37. Kriebardis AG, Antonelou MH, Stamoulis KE, Economou-Pet-
investigation of SAGM-stored leukocyte-filtered red bood ersen E, Margaritis LH, Papassideri IS. Progressive oxidation
cell concentrates: from metabolism to proteomics. Haema- of cytoskeletal proteins and accumulation of denatured
tologica 2012; 97: 10715. hemoglobin in stored red cells. Journal of Cellular and
19. Almizraq R, Tchir JD, Holovati JL, Acker JP. Storage of red Molecular Medicine 2007; 11: 14855.
blood cells affects membrane composition, microvesicula- 38. Antonelou MH, Kriebardis AG, Papassideri IS. Aging and
tion, and in vitro quality. Transfusion 2013; 53: 225867. death signalling in mature red cells: from basic science to
20. Karon BS, van Buskirk CM, Jaben EA, Hoyer JD, Thomas DD. transfusion practice. Blood Transfusion 2010; 8(Suppl 3):
Temporal sequence of major biochemical events during s3947.

e9 2014 The Association of Anaesthetists of Great Britain and Ireland


Orlov and Karkouti | Storage of red cells Anaesthesia 2015, 70 (Suppl. 1), e9e12

39. Cardo LJ, Hmel P, Wilder D. Stored packed red blood cells 57. Marwah SS, Blann A, Harrison P, et al. Increased non-trans-
contain a procoagulant phospholipid reducible by leukode- ferrin bound iron in plasma-depleted SAG-M red blood cell
pletion filters and washing. Transfusion and Apheresis Sci- units. Vox Sanguinis 2002; 82: 1226.
ence 2008; 38: 1417. 58. Ebaugh G Jr, Emerson CP, Ross JF. The use of radioactive
40. Berezina TL, Zaets SB, Morgan C, et al. Influence of storage chromium 51 as an erythrocyte tagging agent for the deter-
on red blood cell rheological properties. Journal of Surgical mination or red cell survival in vivo. Journal of Clinical
Research 2002; 102: 612. Investigation 1953; 32: 126076.
41. Bosman GJ, Werre JM, Willekens FL, Novotny VM. Erythro- 59. Buehler PW, Abraham B, Vallelian F, et al. Haptoglobin pre-
cyte ageing in vivo and in vitro: structural aspects and serves the CD163 hemoglobin scavenger pathway by
implications for transfusion. Transfusion Medicine 2008; 18: shielding hemoglobin from peroxidative modification. Blood
33547. 2009; 113: 257886.
42. Frank SM, Abazyan B, Ono M, et al. Decreased erythrocyte 60. Thomsen JH, Etzerodt A, Svendsen P, Moestrup SK. The hap-
deformability after transfusion and the effects of erythro- toglobin-CD163-heme oxygenase-1 pathway for hemoglobin
cyte storage duration. Anesthesia and Analgesia 2013; 116: scavenging. Oxidative Medicine and Cellular Longevity
97581. 2013; 2013: 523652.
43. Greenwalt TJ, McGuinness CG, Dumaswala UJ. Studies in red 61. Rother RP, Bell L, Hillmen P, Gladwin MT. The clinical
blood cell preservation: 4. Plasma vesicle hemoglobin sequelae of intravascular hemolysis and extracellular
exceeds free hemoglobin. Vox Sanguinis 1991; 61: 147. plasma hemoglobin: a novel mechanism of human dis-
44. Kriebardis AG, Antonelou MH, Stamoulis KE, Economou- ease. Journal of the American Medical Association 2005;
Petersen E, Margaritis LH, Papassideri IS. RBC-derived vesi- 293: 165362.
cles during storage: ultrastructure, protein composition, oxi- 62. Schaer DJ, Buehler PW, Alayash AI, Belcher JD, Vercellotti
dation, and signaling components. Transfusion 2008; 48: GM. Hemolysis and free hemoglobin revisited: exploring
194353. hemoglobin and hemin scavengers as a novel class of ther-
45. Rubin O, Crettaz D, Canellini G, Tissot JD, Lion N. Microparti- apeutic proteins. Blood 2013; 121: 127684.
cles in stored red blood cells: an approach using flow cytom- 63. Wagener FA, Eggert A, Boerman OC, et al. Heme is a potent
etry and proteomic tools. Vox Sanguinis 2008; 95: 28897. inducer of inflammation in mice and is counteracted by
46. Gao Y, Lv L, Liu S, Ma G, Su Y. Elevated levels of thrombin- heme oxygenase. Blood 2001; 98: 180211.
generating microparticles in stored red blood cells. Vox San- 64. Graca-Souza AV, Arruda MA, de Freitas MS, Barja-Fidalgo C,
guinis 2013; 105: 117. Oliveira PL. Neutrophil activation by heme: implications for
47. Rubin O, Delobel J, Prudent M, et al. Red blood cell-derived inflammatory processes. Blood 2002; 99: 41605.
microparticles isolated from blood units initiate and propa- 65. Sassa S. Why heme needs to be degraded to iron, biliverdin
gate thrombin generation. Transfusion 2013; 53: 174454. IXalpha, and carbon monoxide? Antioxidants & Redox Sig-
48. Gladwin MT, Kim-Shapiro DB. Storage lesion in banked naling 2004; 6: 81924.
blood due to hemolysis-dependent disruption of nitric oxide 66. Nemeth E, Ganz T. Regulation of iron metabolism by hepci-
homeostasis. Current Opinion in Hematology 2009; 16: din. Annual Review of Nutrition 2006; 26: 32342.
51523. 67. Hod EA, Brittenham GM, Billote GB, et al. Transfusion of
49. Donadee C, Raat NJ, Kanias T, et al. Nitric oxide scavenging human volunteers with older, stored red blood cells pro-
by red blood cell microparticles and cell-free hemoglobin as duces extravascular hemolysis and circulating non-transfer-
a mechanism for the red cell storage lesion. Circulation rin-bound iron. Blood 2011; 118: 667582.
2011; 124: 46576. 68. Stapley R, Owusu BY, Brandon A, et al. Erythrocyte storage
50. Liu C, Liu X, Janes J, et al. Mechanism of faster NO scaveng- increases rates of NO and nitrite scavenging: implications
ing by older stored red blood cells. Redox Biology 2014; 2: for transfusion-related toxicity. The Biochemical Journal
2119. 2012; 446: 499508.
51. Liu C, Zhao W, Christ GJ, Gladwin MT, Kim-Shapiro DB. Nitric 69. Baek JH, DAgnillo F, Vallelian F, et al. Hemoglobin-driven
oxide scavenging by red cell microparticles. Free Radical pathophysiology is an in vivo consequence of the red blood
Biology and Medicine 2013; 65: 116473. cell storage lesion that can be attenuated in guinea pigs by
52. Minneci PC, Deans KJ, Zhi H, et al. Hemolysis-associated haptoglobin therapy. Journal of Clinical Investigation 2012;
endothelial dysfunction mediated by accelerated NO inacti- 122: 144458.
vation by decompartmentalized oxyhemoglobin. Journal of 70. Wojczyk BS, Kim N, Bandyopadhyay S, et al. Macrophages
Clinical Investigation 2005; 115: 340917. clear refrigerator storage-damaged red blood cells and sub-
53. Bosman GJ. Survival of red blood cells after transfusion: pro- sequently secrete cytokines in vivo, but not in vitro, in a
cesses and consequences. Frontiers in Physiology 2013; 4: murine model. Transfusion 2014 Jul 20. doi 10.1111/trf.
376. 12755.
54. Ozment CP, Mamo LB, Campbell ML, Lokhnygina Y, Ghio AJ, 71. Ozment CP, Turi JL. Iron overload following red blood cell
Turi JL. Transfusion-related biologic effects and free hemo- transfusion and its impact on disease severity. Biochimica
globin, heme, and iron. Transfusion 2013; 53: 73240. et Biophysica Acta 2009; 1790: 694701.
55. Vermeulen Windsant IC, de Wit NC, Sertorio JT, et al. Blood 72. Hod EA, Zhang N, Sokol SA, et al. Transfusion of red blood
transfusions increase circulating plasma free hemoglobin cells after prolonged storage produces harmful effects that
levels and plasma nitric oxide consumption: a prospective are mediated by iron and inflammation. Blood 2010; 115:
observational pilot study. Critical Care 2012; 16: R95. 428492.
56. Uchida I, Tashiro C, Koo YH, Mashimo T, Yoshiya I. Carbo- 73. Roback JD. Vascular effects of the red blood cell storage
xyhemoglobin and methemoglobin levels in banked blood. lesion. Hematology American Society of Hematology Educa-
Journal of Clinical Anesthesia 1990; 2: 8690. tion Program 2011; 2011: 4759.

2014 The Association of Anaesthetists of Great Britain and Ireland e10


Anaesthesia 2015, 70 (Suppl. 1), e9e12 Orlov and Karkouti | Storage of red cells

74. Karkouti K. Transfusion and risk of acute kidney injury in 92. Leal-Noval SR, Munoz-Gomez M, Arellano-Orden V, et al.
cardiac surgery. British Journal of Anaesthesia 2012; 109 Impact of age of transfused blood on cerebral oxygenation
(Suppl 1): i2938. in male patients with severe traumatic brain injury. Critical
75. Karkouti K, Wijeysundera DN, Yau TM, et al. Advance tar- Care Medicine 2008; 36: 12906.
geted transfusion in anemic cardiac surgical patients for 93. McKenny M, Ryan T, Tate H, Graham B, Young VK, Dowd N.
kidney protection: an unblinded randomized pilot clinical Age of transfused blood is not associated with increased
trial. Anesthesiology 2012; 116: 61321. postoperative adverse outcome after cardiac surgery. British
76. Baron DM, Yu B, Lei C, et al. Pulmonary hypertension in Journal of Anaesthesia 2011; 106: 6439.
lambs transfused with stored blood is prevented by breath- 94. Murrell Z, Haukoos JS, Putnam B, Klein SR. The effect of
ing nitric oxide. Anesthesiology 2012; 116: 63747. older blood on mortality, need for ICU care, and the length
77. Brown CH, Grega M, Selnes OA, et al. Length of red cell unit of ICU stay after major trauma. The American Surgeon
storage and risk for delirium after cardiac surgery. Anesthe- 2005; 71: 7815.
sia and Analgesia 2014; 119: 24250. 95. Mynster T, Nielsen HJ. Storage time of transfused blood and
78. Yazdanbakhsh K, Bao W, Zhong H. Immunoregulatory disease recurrence after colorectal cancer surgery. Diseases
effects of stored red blood cells. Hematology American of the Colon and Rectum 2001; 44: 95564.
Society of Hematology Education Program 2011; 2011: 96. Offner PJ, Moore EE, Biffl WL, Johnson JL, Silliman CC.
4669. Increased rate of infection associated with transfusion of
79. Hod EA, Spitalnik SL. Stored red blood cell transfusions: iron, old blood after severe injury. Archives of Surgery 2002;
inflammation, immunity, and infection. Transfusion Clinique 137: 7116.
et Biologique 2012; 19: 849. 97. Pettila V, Westbrook AJ, Nichol AD, et al. Age of red blood
80. Berra L, Coppadoro A, Yu B, et al. Transfusion of stored cells and mortality in the critically ill. Critical Care 2011; 15:
autologous blood does not alter reactive hyperemia index R116.
in healthy volunteers. Anesthesiology 2012; 117: 5663. 98. Phelan HA, Eastman AL, Aldy K, et al. Prestorage leukoreduc-
81. Weiskopf RB, Feiner J, Hopf H, et al. Fresh blood and aged tion abrogates the detrimental effect of aging on packed red
stored blood are equally efficacious in immediately cells transfused after trauma: a prospective cohort study.
reversing anemia-induced brain oxygenation deficits in American Journal of Surgery 2012; 203: 198204.
humans. Anesthesiology 2006; 104: 91120. 99. Purdy FR, Tweeddale MG, Merrick PM. Association of mor-
82. Weiskopf RB, Feiner J, Toy P, et al. Fresh and stored red tality with age of blood transfused in septic ICU patients.
blood cell transfusion equivalently induce subclinical pulmo- Canadian Journal of Anaesthesia 1997; 44: 125661.
nary gas exchange deficit in normal humans. Anesthesia 100. Robinson SD, Janssen C, Fretz EB, et al. Red blood cell stor-
and Analgesia 2012; 114: 5119. age duration and mortality in patients undergoing percuta-
83. Edgren G, Kamper-Jorgensen M, Eloranta S, et al. Duration neous coronary intervention. American Heart Journal 2010;
of red blood cell storage and survival of transfused patients. 159: 87681.
Transfusion 2010; 50: 118595. 101. Saager L, Turan A, Dalton JE, Figueroa PI, Sessler DI, Kurz A.
84. Eikelboom JW, Cook RJ, Liu Y, Heddle NM. Duration of red Erythrocyte storage duration is not associated with
cell storage before transfusion and in-hospital mortality. increased mortality in noncardiac surgical patients: a retro-
American Heart Journal 2010; 159: 73743. spective analysis of 6,994 patients. Anesthesiology 2013;
85. Gajic O, Rana R, Mendez JL, et al. Acute lung injury after 118: 518.
blood transfusion in mechanically ventilated patients. Trans- 102. Sanders J, Patel S, Cooper J, et al. Red blood cell storage is
fusion 2004; 44: 146874. associated with length of stay and renal complications after
86. Gajic O, Rana R, Winters JL, et al. Transfusion-related acute cardiac surgery. Transfusion 2011; 51: 228694.
lung injury in the critically ill: prospective nested case-con- 103. Spinella PC, Carroll CL, Staff I, et al. Duration of red blood
trol study. American Journal of Respiratory and Critical Care cell storage is associated with increased incidence of deep
Medicine 2007; 176: 88691. vein thrombosis and in hospital mortality in patients with
87. Gauvin F, Spinella PC, Lacroix J, et al. Association between traumatic injuries. Critical Care 2009; 13: R151.
length of storage of transfused red blood cells and multiple 104. Taylor RW, OBrien J, Trottier SJ, et al. Red blood cell trans-
organ dysfunction syndrome in pediatric intensive care fusions and nosocomial infections in critically ill patients.
patients. Transfusion 2010; 50: 190213. Critical Care Medicine 2006; 34: 23028.
88. Juffermans NP, Vlaar AP, Prins DJ, Goslings JC, Binnekade JM. 105. Vamvakas EC, Carven JH. Transfusion and postoperative
The age of red blood cells is associated with bacterial infec- pneumonia in coronary artery bypass graft surgery: effect
tions in critically ill trauma patients. Blood Transfusion of the length of storage of transfused red cells. Transfusion
2012; 10: 2905. 1999; 39: 70110.
89. Keller ME, Jean R, LaMorte WW, Millham F, Hirsch E. Effects 106. Vamvakas EC, Carven JH. Length of storage of transfused
of age of transfused blood on length of stay in trauma red cells and postoperative morbidity in patients undergo-
patients: a preliminary report. The Journal of Trauma 2002; ing coronary artery bypass graft surgery. Transfusion 2000;
53: 10235. 40: 1019.
90. Koch CG, Li L, Sessler DI, et al. Duration of red-cell storage 107. van de Watering L, Lorinser J, Versteegh M, Westendord R,
and complications after cardiac surgery. New England Jour- Brand A. Effects of storage time of red blood cell transfu-
nal of Medicine 2008; 358: 122939. sions on the prognosis of coronary artery bypass graft
91. Leal-Noval SR, Jara-Lopez I, Garcia-Garmendia JL, et al. Influ- patients. Transfusion 2006; 46: 17128.
ence of erythrocyte concentrate storage time on postsurgi- 108. van Straten AH, Soliman Hamad MA, Martens EJ, et al.
cal morbidity in cardiac surgery patients. Anesthesiology Effect of storage time of transfused plasma on early and
2003; 98: 81522. late mortality after coronary artery bypass grafting. Journal

e11 2014 The Association of Anaesthetists of Great Britain and Ireland


Orlov and Karkouti | Storage of red cells Anaesthesia 2015, 70 (Suppl. 1), e9e12

of Thoracic and Cardiovascular Surgery 2011; 141: 238 coronary bypass surgery: a prospective randomized study.
43. British Journal of Haematology 1989; 72: 814.
109. Vandromme MJ, McGwin G Jr, Marques MB, Kerby JD, Rue 123. Yuruk K, Milstein DM, Bezemer R, Bartels SA, Biemond BJ,
LW III, Weinberg JA. Transfusion and pneumonia in the Ince C. Transfusion of banked red blood cells and the effects
trauma intensive care unit: an examination of the temporal on hemorrheology and microvascular hemodynamics in ane-
relationship. The Journal of Trauma 2009; 67: 97101. mic hematology outpatients. Transfusion 2013; 53: 134652.
110. Weinberg JA, McGwin G Jr, Griffin RL, et al. Age of trans- 124. Fergusson DA, Hebert P, Hogan DL, et al. Effect of fresh red
fused blood: an independent predictor of mortality despite blood cell transfusions on clinical outcomes in premature,
universal leukoreduction. The Journal of Trauma 2008; 65: very low-birth-weight infants: the ARIPI randomized trial.
27982. Journal of The American Medical Association 2012; 308:
111. Weinberg JA, McGwin G Jr, Marques MB, et al. Transfusions 144351.
in the less severely injured: does age of transfused 125. Karkouti K. From the Journal archives: the red blood cell
blood affect outcomes? The Journal of Trauma 2008; 65: storage lesion: past, present, and future. Canadian Journal
7948. of Anesthesia 2014; 61: 5836.
112. Weinberg JA, McGwin G Jr, Vandromme MJ, et al. Duration 126. Zimrin AB, Hess JR. Current issues relating to the transfusion
of red cell storage influences mortality after trauma. The of stored red blood cells. Vox Sanguinis 2009; 96: 93103.
Journal of Trauma 2010; 69: 142731. 127. Lelubre C, Piagnerelli M, Vincent JL. Association between
113. Yap CH, Lau L, Krishnaswamy M, Gaskell M, Yii M. Age of duration of storage of transfused red blood cells and mor-
transfused red cells and early outcomes after cardiac sur- bidity and mortality in adult patients: myth or reality?
gery. Annals of Thoracic Surgery 2008; 86: 5549. Transfusion 2009; 49: 138494.
114. Zallen G, Offner PJ, Moore EE, et al. Age of transfused blood 128. Triulzi DJ, Yazer MH. Clinical studies of the effect of blood
is an independent risk factor for postinjury multiple organ storage on patient outcomes. Transfusion and Apheresis
failure. American Journal of Surgery 1999; 178: 5702. Science 2010; 43: 95106.
115. Aubron C, Syres G, Nichol A, et al. A pilot feasibility trial of 129. Vamvakas EC. Meta-analysis of clinical studies of the pur-
allocation of freshest available red blood cells versus stan- ported deleterious effects of old (versus fresh) red
dard care in critically ill patients. Transfusion 2012; 52: blood cells: are we at equipoise? Transfusion 2010; 50:
1196202. 60010.
116. Fernandes CJ Jr, Akamine N, De Marco FV, De Souza JA, 130. Wang D, Sun J, Solomon SB, Klein HG, Natanson C. Transfu-
Lagudis S, Knobel E. Red blood cell transfusion does not sion of older stored blood and risk of death: a meta-analy-
increase oxygen consumption in critically ill septic patients. sis. Transfusion 2012; 52: 118495.
Critical Care 2001; 5: 3627. 131. Zubair AC. Clinical impact of blood storage lesions. Ameri-
117. Fernandes da Cunha DH, Nunes Dos Santos AM, Kopelman BI, can Journal of Hematology 2010; 85: 11722.
et al. Transfusions of CPDA-1 red blood cells stored for up to 132. ODonnell J, Boulton FE, Manning RA, Laffan MA. Amount of
28 days decrease donor exposures in very low-birth-weight H antigen expressed on circulating von Willebrand factor is
premature infants. Transfusion Medicine 2005; 15: 46773. modified by ABO blood group genotype and is a major
118. Hebert PC, Chin-Yee I, Fergusson D, et al. A pilot trial evalu- determinant of plasma von Willebrand factor antigen levels.
ating the clinical effects of prolonged storage of red cells. Arteriosclerosis, Thrombosis, and Vascular Biology 2002;
Anestheisa and Analgesia 2005; 100: 14338. 22: 33541.
119. Heddle NM, Cook RJ, Arnold DM, et al. The effect of blood 133. Wiggins KL, Smith NL, Glazer NL, et al. ABO genotype and
storage duration on in-hospital mortality: a randomized con- risk of thrombotic events and hemorrhagic stroke. Journal
trolled pilot feasibility trial. Transfusion 2012; 52: 120312. of Thrombosis and Haemostasis 2009; 7: 2639.
120. Kor DJ, Kashyap R, Weiskopf RB, et al. Fresh red blood cell 134. Carpeggiani C, Coceani M, Landi P, Michelassi C, Labbate A.
transfusion and short-term pulmonary, immunologic, and ABO blood group alleles: a risk factor for coronary artery
coagulation status: a randomized clinical trial. American disease. An angiographic study. Atherosclerosis 2010; 211:
Journal of Respiratory and Critical Care Medicine 2012; 4616.
185: 84250. 135. Pereira A. Will clinical studies elucidate the connection
121. Walsh TS, McArdle F, McLellan SA, et al. Does the storage between the length of storage of transfused red blood cells
time of transfused red blood cells influence regional or glo- and clinical outcomes? An analysis based on the simulation
bal indexes of tissue oxygenation in anemic critically ill of randomized controlled trials. Transfusion 2013; 53: 34
patients? Critical Care Medicine 2004; 32: 36471. 40.
122. Wasser MN, Houbiers JG, DAmaro J, et al. The effect of
fresh versus stored blood on post-operative bleeding after

2014 The Association of Anaesthetists of Great Britain and Ireland e12

Вам также может понравиться