Sei sulla pagina 1di 8

PREOPERATIVE ASSESSMENT

Combining fluid intake from food (1200 ml) and drink (1000 ml)
Fluid and electrolyte and adding water generated metabolically (300 ml) produces an
‘average’ intake of 2.5 l. This is balanced by insensible losses
balance, anaemia and blood from the skin and lungs (900 ml), faecal water content (100 ml)
and urine (1500 ml). This fluid ‘turnover’ represents a modest
transfusion fraction of the total fluid content of the body, which remains
relatively constant, and which is divided between the intracellular,
Iain Mackenzie interstitial, and circulating fluid volumes (Figure 1). The fluid in
each compartment has a specific composition (Figure 2). Losses
of water and electrolytes can be met by about 35 ml/kg/day of
water, together with 1–2 ml/kg day of sodium and 0.5–1 mmol/
kg/day of potassium.
This issue focuses on preoperative assessment, but this contribu-
tion addresses the management of fluid and electrolyte balance, Fluid movement
anaemia and blood transfusion in the perioperative period. Two f o rc e s d r i ve t h e m ove m e n t o f wa t e r b e t we e n
The UK National Confidential Enquiry into Patient Outcome compartments:
and Death (NCEPOD) report, ‘An acute problem?’, published in • osmotic gradients (created by differences in the concentration
2005, identified poor management of fluid as contributing to pre- of solutes either side of a membrane that is impermeable to the
ventable morbidity and mortality, and reflected the conclusions solutes, but permeable with respect to the solvent (water))
of earlier studies. Optimal management of fluid has the capacity • hydrostatic pressure gradients, determined within each com-
to significantly improve outcomes, although whether this is by partment by the relationship between compartment volume
augmenting or limiting fluid compared to ‘usual’ care depends (e.g. circulating volume) and the tension of the walls of the
on the patient populations studied. compartments (e.g. venous tone).
There is little or no hydrostatic pressure gradient between the intra-
cellular fluid (ICF) and extracellular fluid (ECF) compartments,
Volume regulation and electrolytes
so fluid movement between them is regulated by the osmotic
In health, euvolaemia is maintained by the regulation of: gradient. This is predominantly determined by the concentration
• fluid intake (thirst) in response to plasma osmolality via osmo- of electrolytes because of their overwhelmingly greater concentra-
receptors in the anterior hypothalamus which relay signals to tion compared to other osmotically active molecules (e.g. glucose,
the cerebral cortex proteins). Plasma osmolality can be measured in the laboratory
• excretion of urinary water in response to osmolality and volume or plasma osmolarity (see Denman, CROSS REFERENCE) can be
sensors in the major vessels of the thorax and the cardiac estimated:
chambers
• reabsorption of urinary sodium in response to the volume Estimated plasma osmolarity =
sensors. 2 × [Na+] plasma +2×[K+]plasma + [urea]plasma +[glucose]plasma

Iain Mackenzie is a Consultant in Intensive Care and Anaesthesia at Because of the difference between osmolality and osmolarity, the
Addenbrooke’s Hospital, Cambridge, UK. estimated plasma osmolarity would be expected to be less than

Distribution of fluid among body compartments

volume
ular
e asc
m trav volume Osmotic Notes
In
ma
lu

as RBC movement • The proportion of body weight accounted for by body water is
vo

Pl
inversely proportional to body fat, and ranges between 50% and 70%.
ui d

Hydrostatic In general, the proportion is greater in men (60%) than women (50%),
r fl

Colloids
movement
llula

and falls with age (increasing obesity and decreasing lean muscle mass).
me

This is not true for young children and neonates, where 70–80% of body
Extrace

titial fluid volu

Ions weight is body water. In general:


Total body water (TBW) in litres = 0.6 × body weight in kg (men)
Interstitial
0.5 × body weight in kg (women)
Intracellul

fluid Water
• Intracellular fluid (ICF) volume = 2/3 × TBW
• Extracellular fluid (ECF) volume = 1/3 × TBW
Inters

• Plasma volume = 1/4 × ECF


(Falls with age and physical inactivity from 1/4 to 1/5, except in trained
ar fl

Ions athletes.)
ui

• The circulating blood volume (Vb) is the sum of the red cell volume
d volu

and the plasma volume, and is estimated at 75 ml/kg for adults or


m

calculated from the plasma volume (Vplasma) and haematocrit (hct) as


e

follows:
1
Vb = × Vplasma
1–hct

SURGERY 23:12 453 © 2005 The Medicine Publishing Company Ltd


PREOPERATIVE ASSESSMENT

Electrolyte composition, pH and osmolality of body compartments, secretions and commonly available fluids for
intravenous infusion
Plasma volume Clinical
Na+ K+ Ca2+ Mg2+ Cl– HCO3– pH Osmolality T½ (minutes) use
Body compartment
Plasma 142 4 2.5 1 100 25 7.4 280 – –
Interstitial fluid 145 4 2.4 0.9 118 27 7.4 280 – –
Intracellular fluid 12 155 ~0 15 8 10 7.2 280 – –

Secretions
Saliva 50–70 15–20 – – 10–15 30–50 6–7 – –
Gastric juice 150 5–10 – – 100–160 10–20 1–3.5 – –
Bile 180–220 6–8 – – 60–70 60–70 7–8 – –
Pancreatic juice 160 4 – – 30–60 80–120 8–8.3 – –
Small bowel 140 4 – – 100 25 7.8–8 – –

Crystalloids
Normal saline (0.9% NaCl) 154 – – – 154 – 5 300 80 R
Hartmann’s solution 131 5 2 – 111 a 6.5 275 68 R
Dextrose 4%, saline 0.19% 31 – – – 31 – 4.5 286 M
Dextrose 5% – – – – – – 4.2 278 19 M
Bicarbonate 8.4% 1000 – – – – 1000 8 2000 – A
PlasmaLyte 148™ 140 5 – – 98b – 5.5 294 R

Colloids
Haemaccel™ 145 5.1 6.25 – 145 – 7.25 293 240 R
Gelofusine™ 154 0.4 0.4 0.4 120–125 – 7.4 274 180 R
Hydroxyethyl starch (450/0.7) 154 – – – 154 – 5.5 300–310 75 R
Hydroxyethyl starch (130/0.4) 154 – – – 154 – 5.0 308 700 R
Dextran 40 154 – – – 154 – 3.5–7.0 280–324 180 T
Dextran 70 154 – – – 154 – 5.0 280–324 1500 R
HAS 4.5–5% 100–160 <2.25 – – 100–160 – 5.5 200–310 1000 R
HAS 20–25% 50–120 <10 – – 50–120 – 7.4 100–240 1000 R

a and A: Alkanization; M: Maintenance; R: Replacement; T: Thromboprophylaxis; b: Contains lactate as buffer.

the measured plasma osmolality by a quantity called the ‘osmolal and oncotic pressures according to the Starling equation:
gap’, which is usually 9 ± 6 mosm/kg. An osmolal gap greater than
this suggests the presence of osmotically active (but unmeasured) Jv ∝ [(Pc – Pi) − σ(πc – πi)]
substances in plasma (e.g. ethanol, methanol, ethylene glycol,
acetone, mannitol, glycerol). At equilibrium, the osmolality of the where Jv is the fluid flux, P is the hydrostatic pressure in the circula-
ECF and ICF are equal (Figure 2). tion (Pc) and interstitium (Pi) respectively, π is the oncotic pressure
Fluid movement between the intravascular volume (IVV) and in the circulation (πc) and interstitium (πi) respectively, and σ is
interstitial fluid (ISF) compartments (where the semipermeable the basement membrane reflectance coefficient (an index of how
membrane is formed by the vascular endothelium and its basement ‘porous’ the barrier is, which varies by tissue type).
membrane) is governed by hydrostatic and osmotic pressures. The The oncotic pressure gradient is largely a function of the plasma
osmotic gradient is called the ‘colloid oncotic pressure’ because protein (colloid) concentration because the interstitial fluid nor-
this barrier is impermeable only to large molecules (colloids). mally has little protein.
Albumin represents only 60% of plasma protein, but accounts
for 80% of the colloid oncotic pressure because its high negative Sodium
charge holds small cations in the plasma (Gibbs–Donnan effect). Sodium is the principal extracellular cation and, with its associated
Water flux between the IVV and ISF is determined by hydrostatic anions, accounts for most of the osmolality of the ECF. It is the

SURGERY 23:12 454 © 2005 The Medicine Publishing Company Ltd


PREOPERATIVE ASSESSMENT

Disorders of sodium and water

Hypovolaemia Euvolaemia Oedema


Hyponatraemia
Sodium Sodium = Sodium
Water Water Water
Loss of fluid replaced by low-sodium or Stress (postoperative) Renal failure
sodium-free solutions Dilutional hyponatraemia Cardiac failure
Hypothyroidism Cirrhosis
Glucocorticoid deficiency Nephrotic syndrome

Hypernatraemia
Sodium Sodium = Sodium
Water Water Water
Chronic or subacute loss of fluid (diuretics, Mild dehydration Primary hyperaldosteronism
burns, diarrhoea, fistula), post-obstruction, Sodium administration
polyuric renal failure, diabetes insipidus

key regulator of water flux. Unlike other electrolytes, disorders of Potassium


sodium homeostasis must be assessed in the context of hydration The ECF contains only about 1% of total potassium in the body
(Figure 3). In surgical patients, the commonest cause of hyponat- because potassium is predominantly an intracellular cation. Potas-
raemia is due to the replacement of fluid losses with low-sodium or sium homeostasis is achieved by matching urinary losses to intake
sodium-free solutions, particularly postoperatively, when increased by secretion of potassium in the distal convoluted tubule. The ratio
secretion of antidiuretic hormone is part of the stress response. of intracellular to extracellular potassium is maintained by the
Acute hyponatraemia (developing in <48 hours) causes cerebral action of Na+/K+-ATPase in the cell membrane, and determines the
oedema, with symptoms progressing from nausea, vomiting and resting membrane potential of excitable cells (see ‘Action potential
headache to somnolence, confusion, and finally seizures, brain- and nervous conduction’, page 425). Alterations in the potassium
stem herniation and death. In these patients, prompt correction to concentration in the ECF may therefore reflect disturbance of total
increase the concentration of sodium in plasma by 2 mmol/l/hour potassium content in the body (intake, loss) or changes in the ratio
should continue until symptoms resolve. This may be achieved of extracellular to intracellular potassium (Figure 5).
using 3% sodium chloride infused at 1–2 ml/kg/hour together Hyperkalaemia sufficient to cause ECG changes (prolonged PR,
with a loop diuretic. In contrast, symptomatic hyponatraemia widened QRS, peaked T waves) requires immediate treatment.
(confusion, somnolence, ataxia) that develops insidiously must be Calcium gluconate (10 ml of 10% i.v.) stabilizes the myocardium,
treated with gradual correction because the brain compensates for reducing the immediate risk of a dysrhythmia. It must be accom-
plasma hypo-osmolality by reducing intracellular osmolality. Under panied by treatment to reduce potassium concentrations in ECF,
these circumstances, correction of >15 mmol/l/24 hours may such as dextrose and insulin (50 ml of 50% dextrose containing
precipitate central pontine myelinolysis. Chronic asymptomatic 15 IU soluble insulin infused over 30 minutes), nebulized salb-
hyponatraemia can be treated by fluid restriction or by identifying utamol (5 mg) or, if central venous access is available, sodium
and treating the underlying cause (e.g. dilutional hyponatraemia, bicarbonate (1–2 ml/kg infused over 30 minutes). These treatments
endocrine disorders, drugs). reduce potassium in plasma by shifting potassium from the ECF
Hypernatraemia is much less common because it is associated to the ICF and do not, therefore, reduce total potassium in the
with increased plasma osmolality, and usually accompanied by body. Reduction of total potassium in the body requires enhanced
thirst. Patients without access to water, unable to respond to thirst renal elimination (diuretics) or renal replacement therapy if renal
(e.g. unconscious, cognitively impaired, psychiatric conditions, function is impaired.
bed-bound, very young) or with a depressed thirst response are Hypokalaemia can precipitate dysrhythmias, particularly in
at risk. Symptoms of hypernatraemia include a depressed level patients on digoxin, and also requires prompt treatment when
of consciousness, agitation, muscle twitching, hyperreflexia and, symptomatic. Intravenous correction of severe hypokalaemia
particularly in children, seizures. The mortality in adults with a requires cardiac monitoring, close observation and central venous
sodium concentration in plasma of 160 mmol/l is about 75%, access if solutions of >40 mmol/l potassium are used. Accidental
which reflects the severity of the underlying conditions with over-rapid administration of solutions containing high concentra-
which hypernatraemia is usually associated. In those with a deficit tions of potassium are likely to be fatal, and a pump (e.g. syringe
in circulating volume (Figures 4a and b), treatment consists of driver, volumetric or peristaltic pump) to control the infusion rate
returning circulating volume to normal with isotonic fluids, and is essential.
hypotonic fluids to restore total body water.

SURGERY 23:12 455 © 2005 The Medicine Publishing Company Ltd


PREOPERATIVE ASSESSMENT

Fluid loss

a RBC b RBC 'Third-space' fluid losses.


Plasma Interstitial fluid losses. For simplicity, This term is applied if the
transcellular fluids (lymph, gastro- Plasma plasma volume is lost by
2° intestinal secretions, cerebrospinal sequestration to the
fluid, ocular fluid and urine) can be interstitium or body cavities
1° considered as a type of interstitial 1°
that usually contain little or
Interstitial fluid
fluid, although they are separated Interstitial fluid
no fluid, and therefore where
from the interstitial fluid by an the fluid loss is covert.
epithelial layer which secretes This usually occurs when
them and regulates their electrolyte 2°
Na+ the mechanisms that
2° composition. In this example, the 3° normally reabsorb fluid from
Water
losses are overt because the fluid these spaces fail or are
is lost from the body in the form of overwhelmed. Examples
urine, vomit, diarrhoea or insensible include tissue oedema,
losses (sweat, water vapour) or is ascites, pleural effusions, and
evacuated by drains (nasogastric the gastrointestinal tract.
c losses). d Blood loss

RBC RBC
Plasma Plasma


Interstitial fluid Interstitial fluid

Na+
3° Na+
Water 3° 3°
External (as opposed to internal, see Water
Figure 3) plasma volume losses occur
in subjects who have suffered burns. Loss of whole blood.

it occurs over hours rather minutes, and involves the ECF and the
Volume loss
ICF. Eventually, sufficient fluid is lost from the circulating volume
Fluid depletion may involve total body water, the ECF or the IVV to cause circulatory failure (see below).
(Figure 4). The effect of fluid loss on electrolyte concentrations Loss of isotonic fluid from the ECF compartment has no effect
in blood will depend on the composition of the lost fluid (Figure on the osmolality of ECF. This is the only factor involved in fluid
2), as well as the volume and type of fluid the patient may have movement between the ECF and ICF, hence water does not re-
received. Fluid losses are commonly hypotonic with respect to the distribute out of the intracellular compartment until there is a
ECF (Figure 6), resulting in an increase in osmolality of the ECF, sufficient reduction in the circulating volume to trigger non-osmotic
and are partially compensated by the movement of water out of mechanisms for the retention of salt and water:
the ICF (Figure 4a). A loss of up to 10% of body weight or 17% • release of antidiuretic hormone
of total water in the body (7 l in a 70 kg male) can be tolerated if • suppression of release of atrial natriuretic peptide

Causes of hypokalaemia and hyperkalaemia


Hypokalaemia Hyperkalaemia
Increased renal loss Decreased renal clearance
• Drugs (e.g. diuretics, osmotic diuretics) • Renal failure
• Metabolic acidosis • Drugs (e.g. β-blockers, ACE inhibitors, amiloride)
• Magnesium depletion • Metabolic acidosis
Abnormal gastrointestinal losses • Adrenocortical failure
• Vomiting ICF ECF shift
• Diarrhoea • Tissue destruction (e.g. burns, crush injury, haemolysis, tumour
• Villous adenoma lysis)
ECF ICF shift • Metabolic acidosis
• Drugs (e.g. insulin, β-agonists)
• Metabolic alkalosis

SURGERY 23:12 456 © 2005 The Medicine Publishing Company Ltd


PREOPERATIVE ASSESSMENT

Types of fluid loss


Hypotonic Isotonic
• Vomiting • Tissue oedema (decrease in endothelial basement membrane
• Diarrhoea reflectance co-efficient or loss of plasma protein)
• Burns • Losses from intracavitary drains (pleural, peritoneal)
• Ileus • Haemorrhage
• Inappropriate polyuria (diuretics, hyperglycaemia, mannitol,
diabetes insipidus, renal dysfunction)

• sympathetically-mediated redistribution of renal blood flow


• activation of the renin–angiotensin system. IVVfunctional = Tonevascular× IVVabsolute
Losses of isotonic fluid (Figure 6) are of two main types,
fluid shift from the circulating volume to the interstitial space Functional hypovolaemia can be caused by vasodilation or absolute
(tissue oedema) or body cavities (ascites, pleural effusions; reduction of the IVV. The causes of circulatory failure are not mutu-
Figure 4b), or loss of plasma/whole blood (Figures 4c and d). ally exclusive, and all three may coexist in some patients. Rapid
Compared to hypotonic fluid losses where the burden is shared return of functional euvolaemia is the clinical priority, and therefore
between the ECF and ICF, losses of isotonic fluid predominantly hypotonic crystalloid solutions should be avoided because they
affect the IVV and are less well tolerated. Clinical evidence of are distributed throughout total body water. In general, there is no
circulatory failure becomes apparent after the loss of 15–20% evidence to favour the use of isotonic crystalloids over colloids.
of the circulating volume (700 ml in a 70 kg male or 1.67% of Exceptions include the resuscitation of trauma patients (evidence
total body water) and decompensation occurs with the loss of is in favour of crystalloids) and sepsis (colloids may be preferable).
>30% of IVV. Initial resuscitation with either type of solution provides time to
assess blood biochemistry, haematology and coagulation, and to
crossmatch blood. If blood loss is immediately life-threatening,
Circulatory failure and intravascular volume resuscitation
group O Rhesus negative blood without specific crossmatching
Organ dysfunction, together with proximal (low blood pressure, may be needed, after consultation with the blood bank, if possible.
tachycardia) and distal (metabolic acidosis, low mixed venous Maintenance of IVV and haematocrit must also be accompanied
haemoglobin saturation) markers of circulatory failure may be by correction of coagulopathy with platelet concentrates, fresh
caused by pump failure or ‘functional’ intravascular hypovolaemia frozen plasma and cryoglobulin, if necessary.
(Figures 7 and 8). Functional IVV describes the IVV as perceived by
the body’s volume sensors, and can be considered as the product Synthetic colloids
of vascular tone and absolute IVV: Synthetic colloids consist of isotonic crystalloid solutions containing

Features and differential diagnosis of circulatory failure

Pump failure Hypovolaemia Increased capacity


Features
Heart rate ↑ ↑
Blood pressure ↓ ↓
Confusion/agitation
Oliguria
Metabolic acidosis

Differential diagnosis
CVP High/normal Low Low
Cardiac output Low Low High/normal
SVR High High Low
Capillary refill Slow Slow Fast
Diastolic High High Low

CVP: Central venous pressure; SVR: Systemic vascular resistance.

SURGERY 23:12 457 © 2005 The Medicine Publishing Company Ltd


PREOPERATIVE ASSESSMENT

Circulatory failure

Circulatory failure is manifested by increased heart rate,


Hypovolaemia decreased blood pressure, and functional evidence of
organ hypoperfusion (e.g. confusion, agitation, oliguria).
CVP Hypovolaemia, increased circulatory capacity, and pump
Heart
failure are the main causes and they are not mutually
exclusive. Hypovolaemia and increased capacity (sepsis)
are associated with a low central venous pressure, but
can be distinguished because hypovolaemia is
Venous Pump characterized by a low cardiac output and a high
capacitance failure systemic vascular resistance (cold peripheries, slow
capillary return, narrow pulse pressure), while increased
capacity is characterized by a normal or high cardiac
Increased output and a low systemic vascular resistance (warm
capacity peripheries, bounding pulses, wide pulse pressure).
Renal
function

Third-space loss

a suspension of macromolecules of semi-synthetic polypeptides or by α-1,4 linkages with occasional α-1,6 branching linkages.
polysaccharides. The crystalloid solvent may be 0.9% sodium chlo- Hydroxyethyl groups are attached by ether linkage (to reduce
ride, 5% dextrose or have an electrolyte profile similar to plasma metabolic degradation) primarily at C2 of the glucose unit and to
(Figure 2). The colloids may have specific effects with respect to a lesser extent at C3 and C6. The resultant material is hydrolysed
coagulation, crossmatching and the risk of adverse reactions. and fractionated to yield polydisperse colloids with the desired
ranges of molecular weight. The physicochemical properties of
Gelatins (semi-synthetic polypeptides): two forms of gelatin solu- hydroxyethyl starches are characterized by the size range of the
tion are available in the UK. Both are derived from cattle bone, but molecules and the proportion of hydroxyethyl groups to glucose
by slightly different processes. Succinylated gelatin (e.g. Gelofusine units (molar substitution), indicated in parentheses after the
modified fluid gelatin, Physiogel™) is prepared by heating followed product’s name (Mw/molar substitution, e.g. hydroxyethyl starch,
by alkaline hydrolysis of the raw material and chemical linkage 200/0.7). Higher-weight hydroxyethyl starches interfere with the
of the resulting polymers with succinic anhydride as a coupling action of factor VIII and von Willebrand factor—an effect magni-
agent. Urea-linked gelatin (Haemaccel™) is also extracted by heat- fied by higher degrees of substitution, higher C2:C6 ratios, and
ing, but the resulting molecules are crosslinked using methylene infusion volumes of >1500 ml. Hydroxyethyl starches polymers
di-isocyanate. Both solutions contain molecules with a range of are thought to reduce capillary leak by ‘plugging’ holes in the
sizes (i.e. polydisperse rather than monodisperse) which can be endothelial basement membrane, but their use in patients with
summarized as the weight-average molecular weight (Mw) or the sepsis or septic shock is an independent risk factor for acute
number-average molecular weight (Mn). Most of the administered renal failure. Molecules of hydroxyethyl starches 50 kDa in size
dose is excreted unchanged in the urine, with only 10–15% excreted are rapidly eliminated by glomerular filtration. Larger molecules
from the bowel and 3% metabolized. There is no long-term reten- are metabolized by naturally occurring amylases found in liver,
tion. The high calcium content of Haemaccel™ can precipitate clots kidney, spleen and lung, with some of the polymers being taken
when mixed with citrated blood in the same giving set. This can be up by the reticuloendothelial system where they may persist for
prevented by flushing the giving set with a calcium-free crystalloid prolonged periods. Administration of total doses of >3 g/kg are
or colloid. The raw material for the manufacture of these products associated with pruritus.
comes from the USA, where the incidence of bovine spongiform
encephalopathy is low, but the theoretical risk of transmitting new Dextrans (synthetic polysaccharides) are polymers of D-glucose
variant Creutzfeld–Jacob disease remains. synthesized from sucrose by the bacterium Leuconostoc mesenter-
oides, in which 90% of the glucose molecules are linearly linked
Starches (semi-synthetic polysaccharides): hydroxyethyl by α bonds (1–6), with a very small degree of branching. Further
starches are derived from a waxy starch composed almost entirely processing by partial hydrolysis and fractionation produces poly-
of amylopectin extracted from maize or sorghum. Resembling disperse solutions with different weight-average molecular weights,
glycogen, the polymerized D-glucose units are joined primarily commonly 40 kDa (dextran 40) and 70 kDa (dextran 70). As with

SURGERY 23:12 458 © 2005 The Medicine Publishing Company Ltd


PREOPERATIVE ASSESSMENT

hydroxyethyl starches, the extent and speed of renal elimination and prevent clot formation) with adenine, phosphate and dextrose
is a function of molecular weight, with 50% of a dextran 40 solu- (CAPD) as preservatives, giving the blood a shelf-life of 35 days.
tion being renally excreted within a few hours of administration. Unprocessed, a donor unit contains 450–500 ml of whole blood,
Many tissues also contain naturally occurring dextranases, and which is screened for antibodies against syphilis, HIV-1, HIV-2,
metabolic degradation accounts for up to 10% of their elimina- hepatitis B and hepatitis C, and then typed for ABO blood group
tion, with a further 10% being eliminated in the gut. Dextrans and Rhesus D. Most donations are further processed by centrifuga-
improve microcirculatory flow by reducing erythrocyte aggregation tion to yield 250 ml of packed red blood cells, one unit of platelets
and leukocyte plugging, but interfere with platelet aggregation (about 70 ml) and 200 ml of plasma. The packed red blood cells
(reducing circulating concentrations of von Willebrand factor), are diluted to a volume of about 350 ml with a mixture of saline,
and enhance fibrinolysis. The risk of a severe allergic reaction is adenine, glucose and mannitol to a haematocrit of 0.6–0.7, and
slightly greater for dextrans than for gelatins, but can be substan- can then be frozen for long-term storage or refrigerated at 2–6°C
tially reduced by pre-administration of a small dose of dextran 1 for use within the shelf-life. Since 1998 in the UK, blood has been
(Mw 1 kDa), which acts as a hapten-binding agent. Dextran 1 is depleted of leukocytes by filtration, reducing the risk of sensitiza-
not available in the UK. tion to leukocyte antigens.
Except in extreme circumstances (see above), blood for trans-
Natural colloids fusion is matched to the recipient’s blood group and Rhesus type;
Human albumin solution is available as a dilute (4.5% or 5%) in the UK, red cells from potentially suitable units of blood are
or concentrated (20% or 25%) monodisperse solution containing incubated with the serum of the intended recipient at 37°C to
>90% albumin (Mn 66 kDa) that is free of clotting factors, blood check for compatibility. Subsequent transfusion requirements are
group antibodies or plasma cholinesterase. Although derived from likely to require fresh serum for compatibility testing because of the
the plasma of multiple donors in very large quantities, the purifica- potentially rapid generation of red cell antibodies. Procedures for
tion steps (which include cold ethanol extraction, heat-inactivation crossmatching, labelling, collection, checking and administration
and, in some cases, chromatography) are very effective at removing of blood must be extremely rigorous because of the potentially
viruses. In the UK, human albumin solution is derived from human fatal consequences of error.
plasma sourced from the USA to reduce the risk of transmitting Moderate degrees of chronic anaemia (haemoglobin of ≥8 g/dl)
new variant Creutzfeld–Jacob disease. Albumin solutions do not may be correctable (≥10 g/dl) before surgery by giving supplements
have a specific effect on blood coagulation, although there is some (e.g. folic acid, iron, vitamin B12) rather than blood transfusion.
evidence that human albumin solution may interfere with platelet In the postoperative setting, the transfusion threshold is a hae-
aggregation. Human albumin solution does not interfere with moglobin concentration of 10 g/dl. In the absence of frailty or
blood crossmatching, and has a risk of adverse reaction similar cardiorespiratory disease, haemoglobin concentrations down to
to gelatin solutions and hydroxyethyl starches, but is considerably 8 g/dl are well tolerated, and transfusion in these patients may be
more expensive. Use of human albumin solution in critically ill associated with increased mortality. There are additional consid-
adults declined dramatically in the UK after the publication of a erations with large-volume transfusion for major haemorrhage.
meta-analysis in 1988 that concluded it was associated with excess • Rapid infusion of blood at 4°C (which can be avoided by using
mortality. Criticized as methodologically flawed, this conclusion a blood warmer) quickly reduces body temperature, exacerbat-
has not been confirmed by a subsequent meta-analysis, and a ing any coagulopathy, impairing the delivery of oxygen and
large randomized comparison of albumin versus saline to main- increasing the risk of infection.
tain intravascular volume in ICU patients detected no difference • The concentration of 2,3-diphosphoglycerate within the
in morbidity, mortality or length of stay. Hypoalbuminaemia and erythrocytes of stored blood falls with time, shifting the oxyhae-
low colloid oncotic pressure are associated with poorer outcomes moglobin dissociation curve to the left and reducing the capacity
(although causality has not been established). In terms of main- of blood to release oxygen to the tissues (see ‘Physiology of
taining colloid oncotic pressure (rather than intravascular volume), breathing II’, page 430). Normal concentrations of 2,3-diphos-
albumin has the advantage that resultant changes in albumin phoglycerate are restored within 24 hours.
concentration in serum can be measured. However, the association • Over time, intracellular potassium leeches out of the erythro-
between concentrations of albumin in plasma and colloid oncotic cytes of stored blood, and the extracellular concentration of
pressure in serum is weakened in critically ill patients by changes potassium may rise to 30 mmol/l.
in the concentrations of other plasma proteins (e.g. acute-phase • Stored blood is deficient in factors V and VIII (corrected
proteins, immunoglobulins). Nevertheless, infusions of albumin with fresh frozen plasma) and fibrinogen (corrected with
produce increases in albumin concentration in serum and colloid cryoprecipitate).
oncotic pressure and, in some studies, improve outcome. In the • The risk of subsequent development of acute respiratory distress
UK, dilute human albumin solution corrects a plasma–volume syndrome or transfusion-related lung injury are related to the
deficit in patients with salt and water retention and oedema, volume of transfused blood.
but not hypoalbuminaemia per se; concentrated human albumin Other risks associated with blood transfusion include:
solution may be used to obtain a diuresis in hypoalbuminaemic • acute haemolytic transfusion reaction
patients (e.g. in hepatic cirrhosis). • infusion of contaminated blood
• reactions due to non-ABO antibodies
Blood • delayed haemolytic transfusion reactions
In the UK, blood is collected from unpaid volunteers, and put into • anaphylaxis
bags usually containing a mixture of citrate (to chelate calcium ions • alloimmunization.

SURGERY 23:12 459 © 2005 The Medicine Publishing Company Ltd


PREOPERATIVE ASSESSMENT

Some groups of patients will not (Jehovah’s Witnesses) or should to provide an appropriate intravascular volume for the ambient
not (awaiting bone marrow transplantation, previous multiple vascular tone to maintain functional euvolaemia.
transfusions, multiple antibodies) receive blood; other techniques Intravascular volume (and functional euvolaemia) can be
to avoid blood transfusion should be considered (e.g. intraoperative maintained by daily weights in patients whose homeostatic
cell salvage, acute normovolaemic haemodilution). mechanisms are intact and who do not suffer changes in vascular
tone or vascular endothelial reflectance coefficient. This would
Crystalloids apply to patients undergoing day-case surgery or clean elective
Simple electrolyte solutions that do not contain macromolecules are surgery of short to moderate length. Most of these patients are
termed crystalloids. They are iso-osmolar with respect to plasma rendered mildly hypovolaemic before surgery because of prolonged
when used for volume replacement (Figure 2). Hyperosmolar solu- preoperative fasting and fluid restriction; restoration of euvolae-
tions are also available, but these are for specific purposes (e.g. mia with fluid loading (up to 40 ml/kg) pre- or intraoperatively
provision of glucose, alkalinization, rapid resuscitation). Depend- improves subjective outcome from day-case and minor surgery,
ing on their tonicity, these solutions are distributed throughout the and is not controversial.
ECF (isotonic) or total body water (hypotonic). Sodium chloride In more substantial elective surgery where third-space losses
0.9% (‘normal’ saline) is isotonic and iso-osmolar, but the abnor- are minimal and the principal source of morbidity is from fluid
mally high concentration of chloride can cause a hyperchloraemic overload (upper and lower gastrointestinal tract and thoracic sur-
acidosis which has been associated with impaired renal function, gery), limiting perioperative or intraoperative fluids to compensate
poorer gastric perfusion and subjective discomfort. Haemodilution for preoperative dehydration and intraoperative loss of fluid is
with normal saline results in modest prolongation of the prothrom- also beneficial. Equally, studies in vascular surgery have failed to
bin and activated partial thromboplastin times, an exaggerated show benefit from fluid loading. This conservative approach to
reduction of antithrombin III and enhanced platelet aggregation, fluid management in these patients contradicts current anaesthetic
suggesting in vivo activation of the coagulation cascade. practice, despite evidence that fluid overload is associated with
increased morbidity and mortality. However, there are two stud-
Endpoints ies that paradoxically show benefit from fluid loading in patients
Clinical estimation of absolute IVV is impractical and, given the undergoing major elective surgery. These studies were conducted
confounding effect of vascular tone (Figure 5), would be a dis- in a heterogeneous groups of patients and did not specify pre-
appointing exercise. In health, homeostatic mechanisms maintain operative bowel preparation or fluid management, and resulted
the circulating volume (and total body water) within a narrow in smaller differences in fluid balance between the groups. It is
range, and volume excursions below, or significantly above, this possible that in these studies patients were less well hydrated
normal range are poorly tolerated. As discussed above, ‘functional’ before surgery and the intervention groups were ‘fluid loaded’ to
hypovolaemia may be caused by a decrease in volume or tone, functional euvolaemia, leaving the ‘standard care’ groups relatively
and culminates in the signs and symptoms of circulatory failure. hypovolaemic.
Conversely, modest loading of fluid is a normal physiological In the context of a significant preoperative fluid deficit or acti-
response. Increased demand for substrate supply (e.g.stress, exer- vation of a systemic inflammatory response with alterations in
tion, pregnancy) is met by an increased cardiac output, generated vascular tone and capillary integrity (major blood loss, prolonged
by an increase in end-diastolic ventricular volume, as well as surgery, sepsis), the principal source of morbidity is from hypo-
increases in heart rate and cardiac contractility. In the short term, volaemia (absolute and relative). In these patients, attainment of
increased ventricular filling pressure (an important determinant of ‘functional euvolaemia’ is difficult when titrating fluid therapy
end-diastolic volume), is achieved by the sympathetically-mediated against the traditional endpoints of heart rate, blood pressure or
redistribution of intravascular volume from the abdomen, to the urine output because they are compromised by the confounding
heart and lungs. In the longer term, chronically increased demands effects of pain (increase in antidiuretic hormone), vasodilation
are met, in health and disease, by an increase in circulating volume. (epidural analgesia, sepsis), cardiac dysfunction (ischaemic heart
Even in untrained individuals, fluid loading increases maximum disease, sepsis), intrinsic renal disease and absolute hypovolaemia

aerobic capacity (VO2 max), but not endurance. (bleeding, third-space losses). In these more complex cases, early
Surgery and associated complications present a metabolic recourse to more sophisticated measures of intravascular volume
challenge through the effects of pain, inflammation, substrate status (e.g. monitoring of central venous pressure, pulmonary
mobilization and tissue repair. The magnitude of the metabolic artery catheterization, oesophageal Doppler cardiac output moni-
challenge is primarily determined by the procedure (e.g. hernia toring, arterial pulse-contour analysis (PiCCO and LiDCO)), are
repair versus radical cystectomy), surgical technique (skill, speed, beneficial. u
minimal damage to tissue) and any associated complications aris-
ing (e.g. sepsis, haemorrhage). Secondary demands may arise in
the process of trying to meet the primary metabolic burden of
surgery (myocardial infarction, heart failure, ischaemia-related
dysrhythmias). Failure to meet these demands results in morbidity
and mortality. Optimizing functional intravascular volume (fIVV)

allows each patient to reach his own VO2max, but this does not

guarantee that this VO2 (which depends on pre-existing cardiac CROSS REFERENCE
and pulmonary function) will be sufficient to meet the demands Denman E S. Osmolarity and partitioning of fluids. Surgery 2005; 23(6):
of surgery. The goal of perioperative fluid therapy, therefore, is 190–4.

SURGERY 23:12 460 © 2005 The Medicine Publishing Company Ltd

Potrebbero piacerti anche