25.01.2016 Views

Chapter 124

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

Burns and Post-Burn Care:<br />

Anesthetic Considerations<br />

Marc-André Bernath, Pascal Stucki, and Mette M. Berger<br />

<strong>124</strong><br />

CHAPTER<br />

INTRODUCTION<br />

Burn injury is frequent in the pediatric population. It ranks among<br />

the highest causes of death, usually after motor vehicle accidents,<br />

in both industrialized and developing countries worldwide: 1–6 the<br />

majority of victims are preschool-age males of low socioeconomic<br />

income. Most burn cases are scalds and involve only the skin, but<br />

some patients have an additional inhalation syndrome or multiple<br />

injuries that worsen prognosis.<br />

Although mortality has decreased over the last two decades,<br />

except for one study on a restricted number of patients concluding<br />

that young age is not a predictor of mortality in burns, 7 studies on<br />

larger cohorts found that equivalent burn injuries in children<br />


2050 PART 5 ■ Anesthetic, Surgical, and Interventional Procedures: Considerations<br />

TABLE <strong>124</strong>-1. Physiologic Differences Between Children and Adults Affecting Burn Care<br />

System Characteristics Consequences<br />

Anatomy<br />

Fluid homeostasis<br />

Respiratory<br />

Cardiovascular<br />

Metabolism<br />

Renal<br />

Immunity<br />

Central nervous system<br />

Psychological<br />

Higher body surface to weight ratio<br />

Larger head compared to the rest of the body<br />

(19% in 4 years old vs 9% in adults)<br />

Larger fluid content, with larger extracellular<br />

compartment and larger circulating volume<br />

Narrow airways<br />

Limited respiratory reserve<br />

Large cardiovascular reserve (except for young<br />

infants and in case of previous cardiopathy)<br />

Lower systemic vascular resistances<br />

Higher basal metabolic rate<br />

Growth in process<br />

Greater sensitivity to osmolarity changes<br />

Lower sodium clearance (infancy)<br />

Immune response not mature<br />

Stronger acute phase response (except during the<br />

first months)<br />

Better tolerance to hypoxia<br />

Greater cerebral plasticity<br />

Difficulty to express pain and emotional reactions<br />

Larger evaporation: administration of<br />

fluids must be extremely precise<br />

Different estimation of burned surface<br />

(normograms)<br />

More sensitive to fluid shifts and overload<br />

High risk of airway obstruction<br />

Higher rates of respiratory failure<br />

Good hemodynamic tolerance to fluids<br />

Left ventricle more susceptible to hypertension<br />

Larger energy and nutrient requirements<br />

per kg<br />

Risk of stunting, delayed bone formation<br />

More difficult management of hypernatremia<br />

Greater susceptibility to infection<br />

Fewer neurologic sequelae<br />

Difficulty in pain assessment<br />

Undertreatment of pain<br />

where K f<br />

= capillary filtration coefficient an index of the total<br />

number of filtering pores; P c<br />

and P if<br />

= the hydrostatic pressures of,<br />

respectively, intracapillary and interstitial spaces; s = the osmotic<br />

reflection coefficient, an index of the microvascular permeability<br />

to proteins; and π p<br />

– π if<br />

= the oncotic pressures of, respectively,<br />

plasma and interstitial fluid. Burn injury is unique in that it<br />

significantly modifies all these factors toward increased susceptibility<br />

of edema formation (Figure <strong>124</strong>–1): indeed, K f<br />

, P c<br />

and<br />

π if<br />

increase as s, P if<br />

,andπ p<br />

decrease.<br />

Generalized edema will occur with large burns and will most<br />

often develop over the first 12 to 24 hours following the injury.<br />

The net water accumulation arises from an increased filtration rate<br />

with a decrease in both fluid reabsorption and lymph flow at the<br />

whole body level. 14 This fluid extravasation causes a hypovolemic<br />

shock. At the cellular level, a decrease in skeletal muscle membrane<br />

potential has been documented in patients with burns<br />

involving 20 to 55% BSA, in parallel with decreases in muscle<br />

intracellular sodium levels and increases in intracellular potassium<br />

and water contents. These alterations result from a decreased cell<br />

membrane adenosine triphosphatase activity. 15 The changes affect<br />

nearly any organ, and alterations of membrane potential have been<br />

measured in the hepatocyte, the enterocyte, the heart, and skeletal<br />

muscle. Investigators utilizing animal models and clinical studies<br />

involving human primates have produced a large body of information<br />

suggesting that apoptosis is associated with most of the<br />

tissue damages triggered by severe thermal injuries. 16<br />

Inflammatory and Immune Response<br />

Any aggression induces an inflammatory response, which<br />

constitutes an organized defense mechanisms intended to protect<br />

the body from further damage, to restore homeostasis and to<br />

promote wound repair. It includes a local reaction to injury, a systemic<br />

response (tachycardia, tachypnea, fever), cytokine production,<br />

leukocyte changes, endocrine changes, protein alterations<br />

with a reprioritization of hepatic synthesis, and redistribution of<br />

the micronutrients, including trace elements from the vascular<br />

compartment to the liver and reticuloendothelial system. Cytokine<br />

production is strongly enhanced after major burns, the balance<br />

between proinflammatory and anti-inflammatory mediators in<br />

acute injury being lost; 17 the phenomenon is further increased in<br />

case of infection. The intensity of this reaction is correlated with<br />

mortality. 18 Although the initial acute phase response is perceived<br />

as beneficial, its persistence for prolonged periods of time causes<br />

progressive loss of lean body cell mass, particularly of skeletal<br />

muscle, and an increased susceptibility to infection. It will favor<br />

organ dysfunction, and eventually organ failure.<br />

In clinical settings, the detectable changes are increases in<br />

C-reactive protein, ceruloplasmin, and fibrinogen, associated with<br />

decreases in transferrin, prealbumin, and albumin. It is important<br />

to suspect infection as coresponsible for this unspecific response,<br />

to identify the responsible microorganism, and to treat the infection.<br />

19 During the acute phase, reactive oxygen species (ROS, also<br />

called free radicals) production is markedly increased. ROS are<br />

produced primarily by the mitochondrial respiratory chain, and by<br />

the activated leukocytes. This is a normal phenomenon favoring<br />

bacterial destruction and cell signaling. The endogenous antioxidant<br />

defense mechanisms are usually sufficient to cope with<br />

moderate overproduction. When this defense is overwhelmed, as<br />

in severe burns, the deleterious effects of ROS appear, with<br />

proximity oxidation of nucleotides, proteins, and lipids. Burns are<br />

characterized by an intense lipid peroxidation, due partially to the<br />

direct effect of the burn on the lipids contained in skin, but also<br />

through an increased production of free radicals (mainly the anion


CHAPTER <strong>124</strong> ■ Burns and Post-Burn Care: Anesthetic Considerations 2051<br />

Figure <strong>124</strong>-1. Schematic diagram of edema<br />

formation after burns. Under normal conditions,<br />

there is a slight imbalance of the Starling<br />

equation favoring filtration (flow across the<br />

capillary wall = Jv), which is entirely evacuated<br />

by the lymphatic flow (J L<br />

). After burns, the various<br />

factors of the equation increase, inducing<br />

an increase in net filtration in excess of lymph<br />

flow capacity and resulting in edema formation.<br />

The interstitial proteins undergo transformations<br />

contributing to the increase in oncotic<br />

pressure. (P c<br />

and P if<br />

= capillary and interstitial<br />

hydrostatic pressures).<br />

OH–). Topical antioxidant treatments have been proposed but<br />

have not entered routine protocols. Other therapeutic tools are<br />

under investigation (see “Metabolic and Nutritional support”).<br />

Damage to distant tissues occurring with burns is partly attributable<br />

to the lipid peroxides generated in the burn wound. This<br />

favors occurrence of organ dysfunction. Early wound excision has<br />

been advocated on this basis to reduce the release of lipid peroxide.<br />

Early debridement has contributed to the reduction of burn<br />

mortality.<br />

Immune System<br />

An overall depression of the immune system starts within the first<br />

hours of burn injury. Both humoral and cellular responses are<br />

strongly depressed, 20 with reduced numbers of lymphocytes,<br />

reduced macrophage and neutrophil counts (and activity), and<br />

decreased concentrations of opsonins, immunoglobulins, and<br />

chemotactic factors. These alterations persist for weeks and render<br />

the burned patient particularly sensitive to infections. Aseptic<br />

handling is therefore mandatory, requiring nurses and doctors to<br />

perform strict aseptic techniques. Early and daily hydrotherapy<br />

consists of extensive wound scrubbing in a bath. This is usually<br />

undertaken under heavy sedation or general anesthesia. Many<br />

attempts have been made at restoring immunity. Surgery is an<br />

important contributing factor: early excision has been shown to<br />

accelerate the normalization of antibody synthesis. 21 Trace element<br />

deficiencies have long been known to decrease immune response.<br />

Restoring the copper, selenium, and zinc level using large supplements<br />

during the first week postinjury leads to an improved<br />

neutrophil response and to a reduction in infectious complications,<br />

particularly of pneumonia. 22<br />

Cardiovascular System<br />

Almost immediately after injury, massive fluid shifts occur due to<br />

the loss of vascular and endothelial integrity. The magnitude of<br />

the capillary leak is such that proteins up to 300,000 Kd pass the<br />

endothelial barrier during the first hours postinjury. Impermeability<br />

to albumin-sized molecules is restored only by 36 hours<br />

post-burn. 23 The total amount of fluid lost as exudates is about<br />

1 L/10% BSA in adults, with a protein content reaching 30 g/L. 14<br />

This protein loss via the wound exceeds normal protein intake in<br />

healthy subjects. Some of the exudate evaporates from the wound,<br />

leading to more fluid loss. These fluid losses will rapidly cause<br />

hypovolemia with circulatory failure, which, depending on the<br />

burn size and initial resuscitation, may persist for 72 hours post–<br />

initial burn injury. 24<br />

The hemodynamic evolution is summarized in Table <strong>124</strong>–2.<br />

Burn shock is characterized by a profound reduction in cardiac<br />

output (carbon monoxide, CO) that occurs within minutes of<br />

injury. Initially, the systolic blood pressure and heart rate (HR) are<br />

preserved, while the systemic vascular resistance increases twoor<br />

threefold, the pulmonary vascular resistance increasing even<br />

more. 23 This early vasoconstrictive response is mediated by<br />

catecholamines, vasopressin, and thromboxane A 2<br />

(TXA 2<br />

). The<br />

loss of intravascular volume causes a reduction of ventricular<br />

filling pressure, resulting in low CO. Loss of myocardial con-<br />

TABLE <strong>124</strong>-2. Summary of Hemodynamic and Metabolic<br />

Changes After Major Burns<br />

Variable<br />

0–24 Hours 24–72 Hours* >72 Hours<br />

Hemodynamics<br />

MAP no no, ↓ ↓, no, ↑<br />

Heart rate ↑↑ ↑ no, ↑<br />

CVP ↓ no, ↓ no, ↑<br />

SVR + PVR ↑↑↑ ↑, no no, ↓<br />

Vascular permeability ↑↑↑ ↑ no<br />

Cardiac output + CI ↓↓ no, ↑ no, ↑<br />

SVI + LVSWI ↓↓ no no<br />

Metabolism<br />

VO 2<br />

↓ ↑ ↑↑↑<br />

Energy expenditure ↓ no, ↑ ↑↑↑<br />

Core temperature no, ↓ no, ↑ ↑↑<br />

*Expected changes in case of successful resuscitation. In case of delayed fluid<br />

resuscitation or with heart failure, the picture differs.<br />

MAP = mean arterial pressure; CVP = central venous pressure; CI = cardiac<br />

index; SVR = systemic vascular resistances; PVR = pulmonary vascular<br />

resistance; SVI = stroke volume index; LVSWI = left ventricular stroke work<br />

index; VO 2<br />

= oxygen consumption.


2052 PART 5 ■ Anesthetic, Surgical, and Interventional Procedures: Considerations<br />

tractility, alteration of ventricular compliance, and increase of<br />

afterload further contribute to the reduction of cardiac output.<br />

The reduction in cardiac output is detectable within minutes,<br />

before any decrease in plasma volume is detectable, implicating<br />

some form of direct myocardial depression in addition to the<br />

catecholamine effect. Plasma levels of cardiac troponin l (cTnI), a<br />

marker of nonischemic cardiac injury, was found to be elevated in<br />

burned subjects between the 5th and the 14th day postinjury. 25<br />

Animal studies suggest that humoral factors are involved; 26 a<br />

myocardial depressant factor has indeed been isolated in the<br />

serum of injured animals. Likely depressant humoral mediators<br />

are tumor necrosis factor (TNF), interleukins, vasopressin, and<br />

oxygen free radicals. Mechanisms of burn-related cardiac dysfunction<br />

may involve mitochondrial injury from oxidative stress.<br />

Membrane potential depression in heart and skeletal muscle fibers<br />

initiating shock can indeed be counteracted by the administration<br />

of free radical scavengers, such as glutathione peroxidase, catalase,<br />

and superoxide dismutase. 15,27 Mesenteric lymph from burn<br />

animals used at concentrations varying from 0.1 to 5%, but not<br />

from control sham-burn animals, induced dual positive and<br />

negative inotropic effects depending on the concentrations used.<br />

These burn lymph-induced changes in cardiac myocyte Ca 2+<br />

handling can contribute to burn-induced contractile dysfunction<br />

and ultimately to heart failure. 28 Animal studies have confirmed<br />

clinical findings, that the association of burn, with either smoke<br />

inhalation, sepsis, or exposure to lipopolysaccharides enhance<br />

myocardial dysfunction. 29–33<br />

At the microcirculatory level, loss of plasma causes increased<br />

viscosity with a high hematocrit. This is an additional cause of the<br />

observed reduction in coronary blood flow, shifting the myocardium<br />

towards anaerobic metabolism.<br />

The myocardial depression is mainly left-sided: the cardiac<br />

index (CI), stroke volume index (SVI), and left ventricular stroke<br />

work index (LVSWI) are severely depressed. It has been implicated<br />

in adults as contributing to refractory burns shock, and occurs in<br />

pediatric patients as well. 34 In summary, the acute phase of burns<br />

combines features of both hypovolemic and cardiogenic shock,<br />

which must be considered during resuscitation (see “Fluid Resuscitation”).<br />

Over the next days (2 to 5 days after the initial insult),<br />

depending on the efficiency of the initial resuscitation, the patient<br />

will develop a hypermetabolic state, called the flow phase. This<br />

may result in a two- to threefold increase in CO and oxygen consumption,<br />

which will persist for weeks. It is generally associated<br />

with a moderate reduction in systemic vascular resistances, but<br />

may become very severe in the presence of sepsis. This second<br />

phase has hemodynamic features similar to those of a septic shock,<br />

with increased CI, normal SVI, and low systemic vascular<br />

resistance. In children, episodic or persistent arterial hypertension<br />

is frequent both during the acute and the convalescence phases; it<br />

is frequently associated with hypervolemia and is possibly related<br />

to increased renin and catecholamine production. 35<br />

Respiratory System<br />

Inhalation Injury<br />

Smoke inhalation and respiratory complications are still major<br />

causes of mortality in severely burned patients. Respiratory distress<br />

occurs through one or several of the following mechanisms:<br />

mechanical (airway obstruction), toxic (corrosion and intoxication),<br />

inflammatory (cytokines), and infectious. 36 Inhalation<br />

injury is defined as acute respiratory tract damage of variable<br />

severity caused by inspiration of steam or toxic inhalants such as<br />

fumes (small particles dispersed in air), gases and mists (aerosolized<br />

irritants or cytotoxic liquids; Table <strong>124</strong>–3). 37 The diagnosis<br />

is suspected clinically on the basis of history and physical<br />

examination and can be confirmed by bronchoscopy. Respiratory<br />

failure in burned patients occurs through a number of associated<br />

mechanisms. Acute respiratory distress syndrome (ARDS) is a<br />

common early complication. Tracheal stenosis can occur as a late<br />

complication of prolonged mechanical ventilation. Clinical and<br />

experimental studies have shown that damage to the mucosal<br />

barrier and the release of inflammatory mediators are the most<br />

important pathophysiologic events following smoke inhalation.<br />

Manipulation of the inflammatory response following inhalation<br />

may become a future treatment option. 36<br />

Even in the absence of inhalational injury, acute lung injury is<br />

a common cause of morbidity and mortality for patients sustaining<br />

severe burns. A review in 2002 of numerous clinical and<br />

laboratory studies have implicated TNF-α and neutrophils as<br />

important participants in the pathogenesis of burn-induced lung<br />

injury. The mechanism by which these and other proinflammatory<br />

mediators affect the movement of fluid and protein across the<br />

microvascular barrier into the interstitium of lung remained<br />

unclear. 38<br />

A recent study on adult burned patients requiring mechanical<br />

ventilation found a correlation between early onset of ARDS,<br />

changes in white blood cell count and organ dysfunction. Although<br />

multifactorial, the pathogenesis of post-burn respiratory dysfunction<br />

favors an inflammatory process mediated by the effect of<br />

the burn itself, rather than being secondary to sepsis. 39<br />

The lung plays a major role in post injury multiple organ failure<br />

(MOF). In a prospective study on 1344 trauma patients at risk for<br />

postinjury MOF, lung dysfunction was observed in 94% of patients<br />

presenting with one or more organ dysfunctions and in 99% of<br />

patients with two or more organ dysfunctions. The severity of<br />

other organ dysfunction related directly to the severity of respiratory<br />

dysfunction. 40<br />

Renal System<br />

Acute renal failure (ARF) occurring after burn injuries is a threatening<br />

complication, since it is still related with a high mortality<br />

rate. Aggressive fluid resuscitation remains the best preventive tool<br />

of ARF. 41 Before 1983, mortality was 100% in burned children with<br />

concomitant ARF, and has decreased to 56% since 1984. 42 Indeed<br />

ARF occurs in 14.5% of patients with electrical burns 43 (see<br />

“Electrical Injuries”), and a mortality rate of 59% remains similar<br />

to other causes of ARF despite intensive care support including<br />

hemodialysis or peritoneal dialysis. Delayed initiation of intravenous<br />

fluid resuscitation is an important causing factor of ARF<br />

and is related to overall mortality. 41,44<br />

After burn injury, there is an immediate reduction in renal<br />

blood flow mediated by TXA 2<br />

which lasts a few hours. 45 Hypovolemia<br />

and decreased cardiac output further decrease the renal<br />

blood flow and cause filtration failure and tubular dysfunction. In<br />

response to hypovolemia, fluid retention mechanisms are activated,<br />

with elevation of antidiuretic hormone, aldosterone, and<br />

renin activity. 46 Free hemoglobin and myoglobin further contribute<br />

to tubular dysfunction. Rhabdomyolysis occurs to some<br />

extent in any burn patient, but is particularly pronounced in case<br />

of crush syndrome or electrical burns.


TABLE <strong>124</strong>-3. Principal Toxic Gases<br />

Acrolein<br />

Acrylonitriles<br />

Aldehydes<br />

Ammonia (NH 3<br />

)<br />

Chlorine (Cl 2<br />

)<br />

Cyanide<br />

Hydrogen cyanide (HCN)<br />

Formaldehyde<br />

Nitrogen oxides (NO, NO 2<br />

)<br />

Phosgen (COCl 2<br />

)<br />

Sulfide dioxide (SO 2<br />

)<br />

CHAPTER <strong>124</strong> ■ Burns and Post-Burn Care: Anesthetic Considerations 2053<br />

Characteristic Origin Acute Effects<br />

Highly irritating to mucous<br />

membranes<br />

Colorless, highly-water<br />

soluble gas. Forms<br />

ammonium hydroxide:<br />

highly irritating to any<br />

mucous membrane<br />

Heavy, greenish yellow gas,<br />

highly reactive<br />

Colorless, water soluble gas,<br />

bitter almond smell<br />

Colorless, dense, nonflammable<br />

gas<br />

Red-brown, heavy, insoluble,<br />

irritating gas<br />

Colorless, heavy, insoluble gas.<br />

Hydrolyses to form HCl,<br />

extremely irritating<br />

Colorless, heavy, irritating gas,<br />

pungent odor<br />

Cellulose from paper, wood,<br />

cotton, jute. Acrylics in<br />

textiles, wall coverings,<br />

paintings. Polyurethane,<br />

home furnishing<br />

Polyamide in carpets, closing.<br />

Wool, nylon and silk in<br />

blankets, clothing, and<br />

furniture<br />

Mixing of household products<br />

Polyamide and polyurethane<br />

from insulation material<br />

Melamine resins in household<br />

and kitchen goods, foam<br />

insulation<br />

Fabrics and nitrocellulose films<br />

Floor, wall, furniture coverings,<br />

wrappings, wire/pipe coating<br />

Rubber in tires and toys<br />

Severe inflammatory reaction:<br />

tracheobronchitis<br />

Airway obstruction, pulmonary<br />

edema, bronchopneumonia<br />

Airway obstruction, pulmonary<br />

edema, ulcerative tracheobronchitis<br />

Asphyxia<br />

Irritating<br />

Acute pulmonary edema<br />

bronchiolitis<br />

Atelectasis, acute pulmonary<br />

edema, bronchiolitis,<br />

ulcerative bronchiolitis<br />

Bronchoconstriction, mucosal<br />

sloughing, alveolar edema<br />

and hemorrhage<br />

Splanchnic Compartment<br />

The gut has long been identified to be directly affected by burn<br />

injury, with the description of Curling ulcer in the 1970s. 47 The<br />

abdominal compartment syndrome (ACS) has become a matter<br />

of concern in both adult and pediatric burns. 48,49 In one report,<br />

eight out of 48 patients with a mean total BSA (TBSA) burned of<br />

46% developed ACS. All patients with ACS received resuscitation<br />

volumes of 300 mL/kg per day or greater. 49 The other report<br />

compared hypertonic resuscitation with “Parkland” resuscitation<br />

in terms of decreasing the risk for ACS. The hypertonic group<br />

maintained adequate urine output with lower volumes of resuscitation<br />

and had a lower incidence of intra-abdominal hypertension<br />

(2/14 vs 11/22). As in their other report, the critical volume<br />

associated with development of intra-abdominal hypertension was<br />

approximately 300 mL/kg per day—this number is now called the<br />

Ivy index.<br />

Alterations in distribution of blood flow occur in the early postburn<br />

period and are caused by neurogenic and humoral release of<br />

catecholamines and prostanoids. Initially, splanchnic blood flow is<br />

reduced, except for flow to the adrenals and to the liver. TXA 2<br />

is<br />

likely to play a major role in gut dysfunction, promoting mesenteric<br />

vasoconstriction, and decreasing gut blood flow. Poorly<br />

perfused organs shift towards anaerobic glycolysis, promoting<br />

metabolic acidosis. With aggressive fluid resuscitation, perfusion<br />

can be restored to a great extent. The gastrointestinal function,<br />

including the pyloric function, is depressed immediately after<br />

burns. A true paralytic ileus will install for many days if the gastrointestinal<br />

tract is not used; early gastric nutrition is associated<br />

with maintenance of pyloric function. 50 Opiates and sedatives<br />

further depress the gastrointestinal function. Stress ulcer prophylaxis<br />

is mandatory from puberty on (e.g., sucralfate), since<br />

the bleeding risk is elevated in burn injuries and may be lifethreatening.<br />

47<br />

Metabolism and Thermal Regulation<br />

Energy expenditure follows the classical biphasic pattern after<br />

burns: the first 24 to 48 hours, called ebb phase, are characterized<br />

by an intense sideration with depressed energy expenditure and<br />

oxygen consumption. The subsequent flow phase is characterized<br />

by a strong and prolonged increase in resting metabolic rate. 51,52 A<br />

significant proportion of the mortality and morbidity of severe<br />

burns is attributable to this hypermetabolic response, which<br />

can last for as long as 1 year after injury and is associated with<br />

impaired wound healing, increased infection risks, erosion of lean<br />

body mass, hampered rehabilitation, and delayed reintegration of<br />

burn survivors into society. 53<br />

Hypermetabolism is mainly caused by cytokines and stress<br />

hormone release. The stress hormones (i.e., catecholamines,<br />

glucocorticoids and glucagons) are massively released, causing<br />

severe changes in substrate metabolism: ureagenesis, glucogenolysis,<br />

gluconeogenesis, and lipolysis are strongly stimulated, promoting<br />

catabolism. 54 These processes produce extensive body<br />

wasting, resulting in net body-weight loss; this state can persist for<br />

weeks or months, and is particularly deleterious in a growing<br />

child. The occurrence of hyperthermia and sepsis further modify<br />

the metabolic response in an unpredictable direction.


2054 PART 5 ■ Anesthetic, Surgical, and Interventional Procedures: Considerations<br />

Enlargement of the liver with fatty infiltration is a complication<br />

found at autopsy and has been reported repeatedly since the 1970s<br />

in children with extensive burns; 52,55 it is only reported occasionally<br />

in European settings. Liver enlargement starts early<br />

on during the first week, peaking at 2 weeks after burn. The<br />

enlargement is associated with impairment of protein synthesis. 52<br />

This type of complication is associated with hypercaloric nutrition<br />

using large amounts of glucose. This may be related to the more<br />

frequent use of high loads of carbohydrates during parenteral<br />

nutrition in North America. Indeed fatty-liver development is<br />

considered to be secondary to the overload of normal processing<br />

enzymes by the massive peripheral lipolysis, or to the downregulation<br />

of the fatty acid–handling mechanisms as a result of<br />

hormonal or cytokine changes associated with burns. 56 It has been<br />

shown in adult trauma patients that high loads of carbohydrates<br />

cause de novo lipogenesis. 57<br />

Thermal Regulation<br />

After major burns, temperature regulation is seriously altered. The<br />

patients not only lose the insulating properties of the skin, but they<br />

usually strive for a temperature of 38.0 to 38.5°C 58 due to an<br />

increase of their hypothalamic temperature set point. Catecholamine<br />

production contributes to the changes in association with<br />

various cytokines, including interleukin-1 and interleukin-6. Any<br />

attempt to lower the basal temperature by external means will<br />

result in augmented heat loss, thus increasing metabolic rate.<br />

Ambient temperature should be set at 28 to 33°C to limit the<br />

metabolic response. 58 Another cause of increased metabolic rate<br />

is evaporation of exudates from the wounds, which consumes<br />

energy, again causing heat loss. The evaporation causes extensive<br />

fluid losses from the wounds, approximating 4000 mL/m 2 BSA<br />

burns. Every liter of evaporated fluid corresponds to a caloric<br />

expenditure of about 600 kcal. Finally, burned patients are<br />

frequently infected and exhibit highly febrile states. Extensively<br />

burned patients frequently experience hypothermia (defined as<br />

core temperature below 35°C). Surgery under general anesthesia,<br />

which inhibits the heat-conserving and heat-generating mechanisms,<br />

frequently results in hypothermia. Time to recover from<br />

hypothermia has been shown to be predictive of outcome in<br />

adults, 59 time to revert to normothermia being longer in nonsurvivors.<br />

Considering that hypothermia favors infections and<br />

delays wound healing, the maintenance of perioperative normothermia<br />

is of utmost importance. 60<br />

Platelets<br />

The early phase is characterized by a fall in platelet count secondary<br />

to dilution, consumption, increased platelet aggregation, and<br />

to lung trapping. This is followed by thrombocytosis starting<br />

during the 2nd week after injury. This elevation depends on burn<br />

size and may persist for weeks or months.<br />

Clotting Factors<br />

The alterations observed after burns are complex and can be<br />

summarized as follows. During the early phase after burns, fibrin<br />

split products increase. Dilution and consumption explain low<br />

prothrombin time (PT) values. Thereafter, as part of the acute<br />

phase response, fibrin increases, as well as factors V and VIII;<br />

these alterations may last 2 to 3 months.<br />

Central Nervous System<br />

Neurologic disturbances are commonly observed in burned<br />

patients. Several pathophysiologic mechanisms are involved 62<br />

including cerebral glucose metabolism alterations as shown in<br />

animal models. 63,64 Inhalation of neurotoxic chemicals, of carbon<br />

monoxide, or hypoxic encephalopathy may adversely affect the<br />

central nervous system as well as arterial hypertension. 62 Other<br />

factors include hypo- and hypernatremia, hypovolemic shock,<br />

sepsis, antibiotic overdosage (e.g., penicillin), and possible oversedation<br />

or withdrawal effects of sedative drugs. The possibility<br />

of cerebral edema and raised intracranial pressure must be<br />

considered during the early resuscitation phase, especially in the<br />

case of associated brain injury.<br />

ELECTRICAL BURN INJURIES<br />

Electrical injury may be extremely devastating and destructive<br />

(Figure <strong>124</strong>–2). It encompasses different types of injury, depending<br />

on the level of energy involved, and can result in severe<br />

surface and deep tissue injury associated with the passage of<br />

electricity. 65 Associated lesions to the spine must be considered.<br />

High-voltage current (>1000 V) results in the most extensive<br />

Hematologic Effects<br />

Hematologic alterations are extensive, appear during the first<br />

hours after injury, and will persist for weeks. 61<br />

Erythrocytes<br />

Anemia invariably occurs in the course of burns, arising from<br />

multiple factors (enhanced erythrocyte destruction or decreased<br />

bone marrow production). Thermal or electrical injuries induce<br />

direct and delayed destruction of erythrocytes. Other factors such<br />

as blood sampling for laboratory tests, surgery, or gastrointestinal<br />

bleeding play an important role. Reduced or delayed production<br />

of erythrocytes is caused by the inflammatory response, infection,<br />

iron deficiency, and other nutritional deficiencies such as copper<br />

deficiency.<br />

Figure <strong>124</strong>-2. Child with an oral electrical burn injury (courtesy<br />

B. Bissonnette).


CHAPTER <strong>124</strong> ■ Burns and Post-Burn Care: Anesthetic Considerations 2055<br />

damage. In high-voltage injury, three types of injuries are observed:<br />

entry and exit wounds, arc burns, and surface burns resulting from<br />

ignition of clothes or objects in the environment (usually deep<br />

burns). Low-voltage current (domestic accidents) causes<br />

physiologic alterations due to the passage of current flow through<br />

the cardiovascular system, and especially the heart. Alternating<br />

current (50–60 Hz) is more dangerous than direct current, because<br />

it causes tetanic muscle contractions that “freeze” the victim to the<br />

source of the current. Electrical injuries are deep by nature,<br />

involving muscle and other fluid-rich structures. Their demarca -<br />

tion is slow, rendering full assessment difficult. Cardiac arrhythmia<br />

(ventricular fibrillation) and cardiac arrest are common, 66 especi -<br />

ally in injuries from low-voltage alternating current. Vascular<br />

complications include delayed hemorrhage and thrombosis.<br />

Neurologic complications are also frequent (loss of consciousness,<br />

seizures, spinal cord lesions, deafness, peripheral nerve injury).<br />

Gastrointestinal complications include bowel perforations, as well<br />

as ulcers at various levels of the gut. Acute renal failure is a<br />

common complication of electrical injury, due to rhabdomyolysis<br />

or to renal injury directly related to the electrical current.<br />

RESUSCITATION<br />

Initial Evaluation<br />

On the site of an accident, the initial evaluation must be quick and<br />

address life support. As with all traumatic injuries, the first<br />

concern is the patency of the airway (the ABC [airway, breathing,<br />

circulation] of advanced trauma life support). The immediate<br />

institution of intravenous fluid resuscitation is the next high<br />

priority: delays in resuscitation are predictors of poor outcome in<br />

massive burns. A secondary assessment will be carried out in the<br />

hospital emergency department or, better, in the burns facility. As<br />

in adults, the assessment of the burn injury involves the determination<br />

of the total burned surface, using the normograms<br />

adapted for age. It also involves the recognition of associated<br />

injuries and of comorbidity. Burn victims require a detailed and<br />

thorough examination to appreciate the extent of the damage, as<br />

well as a complete history for determination of adequate therapy.<br />

Airway and Inhalation Injury<br />

General Management of the Patient<br />

In children, the risk of acute and rapid airway obstruction is<br />

particularly high, due to the very rapid development of laryngeal<br />

edema. Any sign that the airway is threatened or compromised is<br />

an indication for immediate endotracheal intubation. Otherwise<br />

it is appropriate to provide oxygen through a face mask until the<br />

resuscitation phase is completed. An initial bronchoscopy is<br />

required for the diagnosis of inhalation. In case of mucosal lesions,<br />

sloughing may require repeated suctioning. Total tracheal tube<br />

obstruction by debris (occurring more frequently in small<br />

children) must be recognized immediately, with prompt removal<br />

and replacement of the tube. If acute respiratory failure occurs,<br />

consensus treatment strategies for acute lung injury (ALI)/ARDS<br />

should be applied.<br />

Carbon Monoxide Poisoning<br />

Measurement of the peripheral oxygen saturation (SpO2) is not<br />

helpful for the detection of CO poisoning. Carboxyhemoglobin<br />

TABLE <strong>124</strong>-4. Symptoms of Carbon Monoxide (CO)<br />

Poisoning<br />

CO Hemoglobin, %<br />

0–10<br />

10–20<br />

20–30<br />

30–40<br />

40–50<br />

50–60<br />

60–70<br />

70–80<br />

Clinical Signs<br />

None (angina pectoris in patients with<br />

ischemic coronaropathy)<br />

Headache, cutaneous vasodilatation<br />

(flushing), dyspnea on vigorous<br />

exercise<br />

Pulsatile headache, dyspnea on<br />

moderate exercise<br />

Intense headache, nausea, vomiting,<br />

irritability<br />

Generalized weakness, dizziness,<br />

blurred vision, confusion<br />

Fainting on exertion<br />

Tachycardia, dyspnea at rest, loss of consciousness<br />

Tachycardia, tachypnea, Coma, con -<br />

vulsions, Cheyne–Stokes breathing<br />

pattern<br />

Coma, convulsions, cardiovascular and<br />

respiratory depression<br />

Possible death<br />

Refractory shock, death<br />

(COHb) levels must be measured directly. Symptoms are not<br />

specific (Table <strong>124</strong>–4). Because COHb elimination half-life is<br />

dependent on oxygen tension and time, 100% oxygen should be<br />

provided to accelerate the dissociation of CO from hemoglobin<br />

and to increase the amount of dissolved O 2<br />

in the blood,<br />

improving oxygenation. In adults, hyperbaric oxygen may reduce<br />

the incidence of neurologic sequelae 67 by shortening the halflife<br />

of carboxyhemoglobin from 320 minutes to approximately<br />

60 minutes. The therapeutic target is to reduce COHb below 5%.<br />

In children with neurological symptoms or COHb concentration<br />

greater than 40%, hyperbaric oxygen should be considered despite<br />

the lack of clear data on its effectiveness in children. 68<br />

Cyanide Poisoning<br />

Cyanide uncouples oxidative phosphorylation at the mitochondrial<br />

level. Cyanide binds reversibly to the ferric ion (Fe 3+ ) of<br />

cytochrome oxidase, the terminal oxidase of the mitochondrial<br />

electron transport chain, inhibiting this enzyme and halting<br />

aerobic metabolism. This binding blocks the major pathway of<br />

high-energy phosphate production, resulting in anaerobic<br />

metabolism, decreased adenosine triphosphate (ATP) production<br />

and thus, rapid depletion of cellular energy stores. Glycolysis<br />

continues with further pyruvate production, since it can no<br />

longer be incorporated in the tricarboxylic acid cycle. Instead, it<br />

is reduced to lactate, which accumulates rapidly. This causes<br />

systemic hyperlactatemia and metabolic acidosis with an increased<br />

anion gap. Cyanide poisoning causes tissue hypoxia by decreasing<br />

extraction of the transported oxygen and by inhibition of the<br />

central respiratory centers, which causes hypoventilation. Normally,<br />

the body’s natural defense is the enzyme rhodanese, which<br />

catalyses the complexing of cyanide with sulfur, forming the much<br />

less toxic thiocyanate ion (SCN–). The sulfur pool is limited and,


2056 PART 5 ■ Anesthetic, Surgical, and Interventional Procedures: Considerations<br />

therefore, is the rate-limiting factor for endogenous cyanide<br />

detoxification.<br />

Inhalation of high airborne cyanide concentrations may result<br />

in loss of consciousness after only a few breaths. Patients with<br />

acute poisoning present with headache, vomiting, hypotension,<br />

and cardiac arrhythmias, which rapidly progress to coma, convulsions,<br />

shock, respiratory failure, and death. Specific therapy for<br />

cyanide poisoning is controversial, because of absence of consensus<br />

on the incidence of clinically important exposure. 37<br />

Antidotes are indicated in patients with suspected important<br />

exposure, or in case of severe lactic acidosis (>10 mmol/L). 69 In<br />

adults, hydroxocobalamin was shown to reduce mortality among<br />

victims of cyanide inhalation poisoning and is well tolerated with<br />

few side effects. 70 It may be safely administered very early even by<br />

the rescue team. 71 Other antidotes may be used depending on their<br />

specific risk-benefit ratio (e.g., thiosulfate, cobalt, amyl nitrite, or<br />

sodium nitrite, methemoglobin forming agents). 69<br />

In smoke inhalation, carbon monoxide and cyanide poisoning<br />

may be combined; the treatment should target both toxic agents,<br />

combining cyanide antidotes and normobaric or hyperbaric oxygen.<br />

Circumferential Chest Burns<br />

Extensive third-degree burns to the thorax, especially if circumferential,<br />

may result in impaired thoracic wall motion with<br />

reduced compliance. If early escharotomy is not performed, precipitation<br />

into respiratory failure may occur even in the absence of<br />

any other risk factor.<br />

Fluid Resuscitation and<br />

Hemodynamic Support<br />

General Principles of Fluid<br />

and Drug Administration<br />

Fluid resuscitation is a major determinant of outcome in children<br />

with burn. 41 It requires special care in children, because some<br />

formulas may underestimate their requirements, especially in<br />

small patients (40% BSA). Pediatric<br />

patients require more fluid for resuscitation than adults (Table<br />

<strong>124</strong>–5, Figure <strong>124</strong>–3): this results form the high volume-to-surface<br />

area ratio in infants and children. In children, the separate calculation<br />

of daily maintenance fluids provided, in addition to the<br />

burns resuscitation fluids, is recommended. 72 Current formulas,<br />

which are derived from experimental studies, are successful in<br />

preventing or reversing burn shock in 70 to 95% of cases. 41,44<br />

The principles of fluid resuscitation were developed in the early<br />

1950s. It was shown that exudates and edema fluid found in burn<br />

wounds was isotonic, containing same amounts of electrolytes and<br />

protein as plasma does. Studying hemodynamic effects of various<br />

regimes, using different proportions of colloids and crystalloids<br />

resulted finally in the development of the Parkland Hospital<br />

formula. 73 No single fluid resuscitation formula has proven to be<br />

superior, but the Parkland formula probably remains the easiest<br />

and therefore the safest for extended use.<br />

In burns below 5% BSA, fluid resuscitation can be done by the<br />

oral route. Above 5% BSA, an intravenous resuscitation is required<br />

under normal civilian condition. The first half of the fluids<br />

calculated with the Parkland formula is usually administered over<br />

8 hours, the remainder being given in 16 hours. Other treatment<br />

strategies recommend even a faster infusion rate in the first<br />

4 hours. The crystalloid resuscitation results in large increase of<br />

total body sodium and water contents, with fluid retention and<br />

massive interstitial edema. Nevertheless, liberal fluid administration<br />

is associated with several possible complications such as an<br />

increasing need for tracheostomies, pulmonary edema, and ACS<br />

with raised intra-abdominal pressure. Therefore, a strict control of<br />

fluid administration is required. After the first 24 hours, fluid and<br />

sodium administration is drastically reduced by 50 to 70% compared<br />

to the first day to cover daily maintenance requirements.<br />

The use of colloids is controversial. It is generally accepted that<br />

liberal colloid use results in increased morbidity and delayed<br />

healing. Colloid use should be avoided as much as possible.<br />

Albumin 5% may be considered if severe hypoalbuminemia is<br />

present (albuminemia 15–20g/L). Indication to fresh frozen<br />

plasma should be limited to correction of coagulation defects.<br />

Below burns 30–40% BSA, a central venous catheter is usually<br />

sufficient to guide hemodynamic resuscitation. In larger burns,<br />

a arterial catheter is helpful to adjust vasopressor therapy. In<br />

recent years, minimally invasive hemodynamic monitoring has<br />

been made available. Pulse contour analysis associated with<br />

TABLE <strong>124</strong>-5. Resuscitation Formulas for Pediatric Patients<br />

Author Formula Time Partition Fluid Pediatric Specific<br />

Evans,1952<br />

Carvajal<br />

Shriners’ Burns Unit,<br />

Galveston<br />

Shriners’ Burns Unit,<br />

Cincinnati<br />

Baxter, 1968,<br />

Parkland<br />

Sick Kids Hospital,<br />

Toronto<br />

1 mL/kg/1% BSA +<br />

1 mL/kg/1% BSA<br />

5000 mL/m 2 BSA burn<br />

+ 2000 mL/m 2<br />

5000 mL/m 2 BSA burn<br />

+ 2000 mL/m 2<br />

4 mL/kg/1% BSA burn<br />

+ 1500 mL/m 2<br />

4 mL/kg/1% BSA burn<br />

6 mL/kg/% BSA burn<br />

0–24 h<br />

0–24 h<br />

0–24 h<br />

0–24 h<br />

0–24 h<br />

0–8 h 50%<br />

8–16 h 25%<br />

16–24 h 25%<br />

0–24 h<br />

0–8 h 50%<br />

8–24h 50%<br />

0–8 h 50%<br />

8–24 h 50%<br />

RL<br />

colloid<br />

RL<br />

RL<br />

+ 12.5 g albumin<br />

RL + 50 mmol NaHCO 3<br />

RL<br />

RL + 12.5 g albumin<br />

RL<br />

RL<br />

No<br />

Yes<br />

Yes<br />

Yes<br />

No<br />

Yes<br />

RL = Ringer lactate.


CHAPTER <strong>124</strong> ■ Burns and Post-Burn Care: Anesthetic Considerations 2057<br />

Time<br />

Variable<br />

Therapeutic options<br />

Result<br />

adequate<br />

not achieved<br />

Admission<br />

RL 4 ml x kg -1 x burned %BSA-1 in 24 hr<br />

50% in 4 hours<br />

Antioxidant micronutrients (IV)<br />

At 4 hours<br />

Assess<br />

Urine output<br />

MAP<br />

pHa<br />

1ml<br />

stable<br />

7.3<br />

< 1ml<br />

unstable<br />

< 7.3<br />

achieved<br />

continue<br />

Start EN<br />

not ok<br />

RL: 1 ml x kg -1 x %BSA -1<br />

At 8 hours<br />

Re-Assess<br />

As above + CVP<br />

Albumin<br />

10 mmHg<br />

15 ml x kg -1<br />

> 10 mmHg<br />

< 15 ml x kg-1<br />

achieved<br />

not ok<br />

continue<br />

RL: 1 ml x kg -1 x %BSA -1<br />

colloids 10 ml x kg -1<br />

At 12 hours<br />

As above<br />

achieved<br />

Re-Assess<br />

not ok<br />

continue<br />

dobutamine 5 µg x kg -1 /min<br />

consider PAC<br />

At 18 hours<br />

Re-Assess<br />

At 24 hours<br />

As above + SvO 2<br />

PaO 2 /FIO 2<br />

65%<br />

200<br />

achieved<br />

continue<br />

< 65%<br />

< 200<br />

not ok<br />

Shift fluid composition<br />

to glucose 5%, or glucosaline<br />

prescribe 50% of first 24 hrs' fluid intake<br />

use PAC<br />

dobutamine<br />

Re-Assess<br />

As above<br />

achieved<br />

continue<br />

not ok<br />

dobutamine<br />

colloids 10 ml x kg-1<br />

Figure <strong>124</strong>-3. Resuscitation algorithm (see legend for Figure <strong>124</strong>–1).<br />

thermodilution cardiac output measurement (PiCCO) provides<br />

accurate cardiac output assessment even in small children and<br />

gives important parameters to guide hemodynamic resuscitation. 74<br />

Some laboratory routines are useful particularly during the<br />

first weeks after a major burn injuries; these are summarized in<br />

Table <strong>124</strong>–6.<br />

Mass Casualty<br />

When fire or burn disasters cause mass fatalities, most or all<br />

of the fatalities occur on-scene, during transit, or soon after<br />

hospital arrival. Civilian accidents generally involve a constant<br />

proportion of children and adolescents, particularly when


2058 PART 5 ■ Anesthetic, Surgical, and Interventional Procedures: Considerations<br />

TABLE <strong>124</strong>-6. Proposed Minimum Laboratory Monitoring Routines in Patients With Major Burns (>40% Body Surface Area)<br />

During Acute Phase (First 2 Weeks)<br />

Hemoglobin, hematocrit,<br />

Na, K, glucose<br />

Mg (ionized), P<br />

Ca (ionized)<br />

pH, anion gap, lactate,<br />

blood gases<br />

Albumin<br />

Prealbumin<br />

Leukocyte count<br />

C-reactive protein<br />

ASAT, ALAT, γ-GT,<br />

alkaline phosphatase<br />

Frequency<br />

6 hourly during the first 72 h, or 24 h after the<br />

surgical sessions<br />

Daily during the first week<br />

Then, 2–3 times weekly<br />

After transfusion<br />

Weekly during parenteral nutrition and with<br />

prolonged bed rest<br />

Twice per day<br />

Every 12 hours during the first 72 h, then daily<br />

during first week<br />

Once weekly<br />

Daily during first 72 h<br />

Daily in case of fever<br />

Weekly with parenteral or enteral nutrition, or<br />

in case of suspicion of overfeeding<br />

Comment<br />

Very low levels are very common<br />

In respiratory failure, may require more<br />

frequency<br />

Helps assess response to feeding<br />

After resuscitation according to clinical<br />

signs of infection<br />

Remains elevated during the first 10 days.<br />

Changes help to assess presence of<br />

infection<br />

Often slightly increased without any<br />

clinical relevance<br />

Beware of increasing values with artificial<br />

nutrition<br />

ASAT = aspartate transaminase; ALAT = alanine aminotransferase; γ-GT = gamma glutamyl transferase.<br />

discotheques are involved. Burn patients requiring hospitalization<br />

are usually distributed across several hospitals on the day of<br />

a disaster. 75<br />

It is important to keep in mind that in case of mass causality,<br />

oral fluid resuscitation is an alternative with burns up to 40%<br />

BSA; 76 the volume of fluid required to achieve stability is about<br />

15 to 20% of body weight during the first 24 hours, which corresponds<br />

to 10.5 to 14 liters per day in a 70-kg patient—it is enormous.<br />

Salt water must be avoided, because it generates nausea and<br />

vomiting. Practically, salt tablets are added to water or any other<br />

fluid (5–7.5 g/L). 77<br />

Hospitalized burn patients are resource- intensive and have<br />

longer lengths of stay when compared with other disaster victims.<br />

In addition to patients received directly on the day of the disaster,<br />

burn centers should anticipate additional admissions in the first<br />

week following the disaster, as other hospitals appropriately<br />

request transfer of admitted thermally injured patients.<br />

ANESTHETIC MANAGEMENT<br />

General Considerations<br />

Successful anesthesia for hydrotherapy, excision, and grafting of a<br />

burn requires planning. Patients with burns involving


CHAPTER <strong>124</strong> ■ Burns and Post-Burn Care: Anesthetic Considerations 2059<br />

Tracheal Intubation<br />

An important airway management decision must be made as soon<br />

as the patient arrives in the hospital. Edema associated with<br />

massive fluid resuscitation may compromise the airway and make<br />

delayed tracheal intubation difficult. As a general rule, it is better<br />

to tracheally intubate the burn patient early rather than late. For<br />

children, an inhalational induction with oxygen and a volatile<br />

agent such as sevoflurane before airway manipulation is probably<br />

the safest technique. 72 Nasotracheal intubation is the preferred<br />

route for prolonged tracheal intubation in infants and small<br />

children, because it is more secure and less irritant to the trachea<br />

with head movement. Uncuffed tubes have been and may remain<br />

the first choice in children and infants. Classically, the tube size<br />

is chosen to permit an air leak as of an airway pressure of 18 to<br />

25 cmH 2<br />

O. On the rare occasions with concomitant ARDS, cuffed<br />

tubes may be needed to ensure high airway pressure ventilation<br />

for adequate oxygenation. During the first 72 hours postinjury,<br />

edema of the tracheal mucosa may be severe, and may require<br />

reduction in tube diameter. However, changing the tracheal tube<br />

at this stage may be life-threatening, as usually the face and upper<br />

airway are also extensively swollen, rendering visibility and access<br />

to intubation very difficult. If children are critically burned and<br />

expected to require more than transient mechanical ventilation<br />

support, low-pressure cuffed endotracheal tubes should be placed,<br />

regardless of the child’s age. 82 Because accidental extubation may<br />

be fatal at this stage, particular care should be taken to secure<br />

properly the tracheal tube (lace around the head) associated to<br />

deep sedation. In particular cases, utilizing a suture to maintain<br />

the tube can be considered. At any stage in the course of a burn<br />

injury involving the face or neck, the airway may be significantly<br />

compromised. In patients with burns involving the neck, the upper<br />

thorax, and the inferior part of the face, scarring with associated<br />

tissue retraction may lead to difficult debridement. It affects<br />

airway management; intubation of a previously easy airway may<br />

become extremely difficult after a few weeks. Ketamine anesthesia<br />

is an alternative for the release of neck contractures before<br />

intubation. Even when available, fiberoptic-aided intubation may<br />

fail; a laryngeal mask can offer a temporary rescue solution.<br />

Monitoring<br />

Standard monitoring may be extremely difficult to obtain in major<br />

burns. A useful means of monitoring may be by esophageal<br />

stethoscope, as it practically always allows hearing both heart and<br />

ventilation sounds. Peripheral pulse oximetry may be difficult to<br />

obtain from a finger, toe, or ear, but alternative sites such as, nose,<br />

or tongue may be helpful. Monitoring temperature is mandatory,<br />

as heat loss may be massive. Capnography is mandatory with all<br />

forms of ventilation, whether mechanical or spontaneous, especially<br />

for patients with their trachea intubated or for those<br />

breathing through a laryngeal mask. Blood pressure is measured<br />

by cuffs, adapted to any limb for minor burns, or monitored<br />

invasively in major burn patients. In children, arrhythmias are<br />

mainly of three types: sinus tachycardia, almost universal, and<br />

(rarely) ventricular fibrillation or cardiac arrest. An echocardiogram<br />

(ECG) is sometimes very difficult to obtain, especially when<br />

the patient is rotated during surgery. Alternative electrode sites<br />

should be tried. If no ECG trace is obtainable, consider placement<br />

of sterile subcutaneous or intradermal pace maker wires in the<br />

surgical field or needles with crocodile grips; extensively burned<br />

patients are at high risk of hemodynamic instability, with an<br />

increased risk of problems presented by arrhythmias. As mentioned<br />

above, severely burned patients with hemodynamic<br />

instability are monitored with central lines and may even require<br />

pulse contour cardiac output monitoring 74 to help guide the fluid<br />

and pharmacologic therapy. Urine output should be measured and<br />

maintained >0.5 mL/kg/h throughout the surgical and anesthetic<br />

procedure.<br />

Heat Loss<br />

Temperature control is mandatory, and maintaining temperature<br />

is a serious concern. A study of patients with a mean burn size<br />

of 44% TBSA showed that patients at thermoneutral ambient<br />

temperature (28–32°C) had metabolic rates 1.5 times those of<br />

nonburned controls. 83 However, when ambient temperature was<br />

decreased to 22–28°C, the metabolic rate increased in proportion<br />

to burn size. Thus, ambient temperatures less than the thermoneutral<br />

range should be avoided, whether in the burn unit or in the<br />

operating room. The four classical routes for temperature loss are<br />

convection, conduction, evaporation, and radiation. During burn<br />

excision/grafting sessions, large areas of skin are exposed, leading<br />

to extensive evaporative and convective heat losses. The main<br />

factors affecting heat loss are the burned area surface, the donor<br />

site area for grafts, wet packs, a cool operating room, and anesthesia,<br />

which causes vasodilatation. Heat loss can hence be<br />

reduced by raising room temperature and closing doors to limit<br />

draughts, using a warming blanket or an overhead heater, covering<br />

the nonoperated areas, warming blood and infused solutions,<br />

warming and humidifying inspired gases, and using warm packs.<br />

Using a forced-air convection system is usually not possible with<br />

extensive burns, as the remaining available surface after surgical<br />

preparation is very limited. However, it is recommended for<br />

surgery limited to the extremities or small burned surface areas.<br />

Pharmacology and Choice<br />

of Anesthetic Agents<br />

Burns involving >10% BSA cause large changes in fluid compartments<br />

and alter pulmonary, hepatic, and renal functions considerably.<br />

Uptake, volume of distribution, and clearance of many drugs<br />

are affected. 84 The major changes in plasma proteins will affect the<br />

pharmacokinetics of the medications such as benzodiazepines<br />

with strong protein binding. The two most important proteins<br />

in this respect are alpha1-acid glycoprotein and albumin: the<br />

first increases, and albumin, the most important quantitatively,<br />

decreases after major burns, modifying the proportion of free<br />

drugs in an unpredictable way. Because most anesthetic drugs are<br />

not highly protein bound, the impact of these changes is minimal.<br />

Inhalation Agents<br />

The pharmacokinetics of inhalation anesthetics are least altered<br />

amongst anesthetic drugs. All halogenated agents have been used<br />

for anesthetizing burn patients with no major problems. Many<br />

anesthesiologists however, prefer total intravenous anesthesia<br />

(TIVA), considering inhalational techniques unfit for burns<br />

because of their undesired side effects. For instance, halogenated<br />

agents cause peripheral vasodilatation with enhanced cooling and<br />

increased bleeding, postoperative rigidity, and shivering (source<br />

of graft displacement and pain), They are contraindicated with<br />

certain vasoconstrictor regimes used for surgery (halothane


2060 PART 5 ■ Anesthetic, Surgical, and Interventional Procedures: Considerations<br />

combined with epinephrine). Halothane has been extensively used<br />

in burned children. There is no evidence that its repeated use is<br />

associated with additional risk of halothane hepatitis. 84 The lower<br />

solubility of sevoflurane, combined with minimal airway irritation,<br />

offers the advantage of a more rapid induction. The same<br />

mentioned undesired effects apply to this inhalation agent except<br />

that it is less feared in presence of epinephrine. Isoflurane has no<br />

advantage on either halothane or sevoflurane for induction.<br />

Moreover of the three halogenated agents, it is the most potent<br />

vasodilator. Entonox, a mixture of oxygen and nitrous oxide, has<br />

been used extensively for dressings in burned children. Although<br />

very effective, long-term use is associated with bone marrow<br />

depression. 85<br />

Intravenous Agents<br />

KETAMINE: Some procedures like hydrotherapy or bandaging can<br />

be performed using ketamine anesthesia or sedation. This agent is<br />

particularly versatile, having both analgesic and anesthetic effects.<br />

The most important adverse effects are hallucinations and excessive<br />

increases in blood pressure and heart rate. These reactions<br />

can be attenuated or avoided by combining ketamine with sedative<br />

or hypnotic drugs like midazolam and/or propofol. 86 Dreams and<br />

psychic problems seem to be less common in patients with severe<br />

burns, 78 possibly from the frequent use of benzodiazepines for<br />

their basal sedation in the intensive care unit (ICU). It can be<br />

administered by both intramuscular and oral routes, making it<br />

advantageous when venous access is difficult. Following intravenous<br />

administration, a rapid onset of action is seen within 1 minute<br />

lasting for about 10 minuts. The use of the pure S-ketamine<br />

enantiomer, which is more potent than the racemic solution,<br />

allows the dose to be reduced. Anesthesia can be initiated by<br />

incremental I.V. doses of midazolam up to about 0.1 mg/kg<br />

or until the child looks sleepy, followed by I.V. injection of<br />

S-ketamine at a dose of 0.5-1 mg/kg, repeated every 10 to 15<br />

minutes (representing a 50% dose reduction compared to racemic<br />

ketamine). Ketamine with propofol is hemodynamically neutral.<br />

Combining (S)-ketamine to midazolam for analgosedation in the<br />

ICU reduces exogenous catecholamine requirements. Moreover,<br />

the effects on intestinal motility are superior to opiates. 86 In a<br />

randomized, double blind trial on burned children, propofolketamine<br />

combination was superior to propofol-fentanyl because<br />

of more restlessness in patients given propofol–fentanyl. 87<br />

Ketamine is proposed as first choice for anesthesia in burned<br />

patients, for its many advantages: 88 rapid onset and short duration<br />

of action, its wide safety margin, its direct stimulation effect on<br />

central sympathetic tone which is threefold in burned patients, its<br />

faculty of retaining protective airway reflexes, its prolonged<br />

analgesic effect, and its capacity to reduce systemic inflammation<br />

and ischemia–reperfusion damage.<br />

PROPOFOL: The use of propofol has gained wide acceptance<br />

among pediatric anesthesiologists over the last decade. 89,90 Clinical<br />

studies have shown that infants and young children require larger<br />

doses than older children and adults for both induction and<br />

maintenance. A pharmacokinetic study done on children 1 to<br />

3 years of age with minor burns (48 hours) at high doses (>4 mg/kg/h) may cause a rare<br />

but frequently fatal complication known as propofol infusion<br />

syndrome (PRIS). PRIS is characterized by metabolic acidosis,<br />

rhabdomyolysis of both skeletal and cardiac muscle, arrhythmias<br />

(bradycardia, atrial fibrillation, ventricular and supraventricular<br />

tachycardia, bundle branch block and asystole), myocardial failure,<br />

renal failure, hepatomegaly and death. PRIS must be kept in mind<br />

as a rare, but highly lethal, complication of propofol use, not<br />

necessarily confined to its prolonged use. If PRIS is suspected,<br />

propofol must be stopped immediately and cardiocirculatory<br />

stabilization and correction of metabolic acidosis initiated. 92 In<br />

view of the poor prognosis and availability of alternative forms of<br />

sedation, propofol is not recommended for long-time infusion. 93<br />

Prolonged infusions should also be avoided considering the<br />

possibility of direct neurotoxicity through an effect on the<br />

γ-aminobutyric acid (GABA)ergic neurons. 94 However, propofol<br />

remains suitable for short-term perioperative sedation or anesthesia<br />

associated with high-dose opiate analgesia or with ketamine.<br />

87,95 In an animal model of burn injury propofol anesthesia<br />

offered a possible protection against apoptosis of enterocytes<br />

and reduced serum TNF-α levels, when compared to ketamine<br />

anesthesia. 96 This would be an argument to use propofol for the<br />

first anesthetic post burn injury, ignoring the viewpoint that its<br />

use should be avoided in the first 48 hours, period of major<br />

hemodynamic instability. 89 Patient-controlled sedation with<br />

propofol has been validated as safe and effective for burn dressings<br />

provided no lockout interval was used. 97 This may well be applied<br />

to older children.<br />

Muscle Relaxants<br />

Burned patients respond abnormally to both depolarizing and<br />

nondepolarizing muscle relaxants. This is related to changes in the<br />

muscle membrane observed in burns of less than 10% BSA.<br />

Neuromuscular dysfunction is proportional to burn size and to the<br />

degree of hypermetabolism: it is related to changes in the nicotinic<br />

acetylcholine receptors. Immature nicotinic receptors appear in<br />

the neuromuscular junction and are generalized to the entire<br />

skeletal muscles. The absolute number of receptors increases, and<br />

they differ from mature receptors with respect to their half-life, the<br />

duration of their ionic canal permeability, their affinity for agonists,<br />

and their sensitivity to antagonists. 98 Immobilization further<br />

contributes to these changes in acetylcholine receptor subunit<br />

mRNA, but the changes after burns differ from those seen after<br />

denervation. 99 A decrease in plasma cholinesterase activity is also<br />

observed.<br />

DEPOLARIZING AGENTS: The response to succinylcholine is<br />

increased, with a marked increase in kalemia. Cardiac arrest<br />

following normal doses of succinylcholine has repeatedly been<br />

reported. 100–103 The rise in kalemia is related to the burn size, the<br />

dose of succinylcholine, and the time elapsed since the injury. It<br />

appears after 3 to 4 days; duration of the phenomenon is related to<br />

the burn size and to the duration of immobilization in bed. The


CHAPTER <strong>124</strong> ■ Burns and Post-Burn Care: Anesthetic Considerations 2061<br />

acute sensitivity to succinylcholine was demonstrated on electromyographic<br />

investigations using doses as small as 0.1 mg/kg.<br />

Patients may become paralyzed with very small doses at the time<br />

of maximum sensitivity. Succinylcholine can be used in the<br />

immediate postburn period (


2062 PART 5 ■ Anesthetic, Surgical, and Interventional Procedures: Considerations<br />

TABLE <strong>124</strong>-7. Simplified Sedation Score<br />

Level<br />

Clinical Sign<br />

0 Agitated<br />

1 Awake<br />

2 Drowsy, roused by voice<br />

3 Roused by strong stimuli (tracheal suction)<br />

4 Not arousable<br />

5 Anesthetized or paralyzed<br />

Adapted from Addenbrooke’s Sedation Score. 153<br />

the perception of pain. Assessment can be done by using questionnaires<br />

or visual analogue scales (VASs) in adults. In children, the<br />

Children’s Hospital of Eastern Ontario’s Pain Score (CHEOPS)<br />

score, a visual analog scale, and the pain thermometer developed<br />

in Montreal can be used. 111 Frequent pain assessment with valid<br />

patient self-report measures should be the basis for documenting<br />

pain treatment. Whenever sedation is used in the ICU, there is a<br />

risk of oversedation, because it is easier to nurse a sleeping patient.<br />

Oversedation has a cost and must be avoided. Therefore, the depth<br />

of sedation must be regularly assessed (Table <strong>124</strong>–7), and patients<br />

should always remain arousable.<br />

Hypnosis<br />

This form of pain control has been increasingly included in the<br />

multimodal pain management of burned patients. 123,<strong>124</strong> Case<br />

reports in children are generally positive, but authors report it to<br />

be more difficult in preschool-aged children, whereas children<br />

aged 6 years and older respond very well.<br />

Regional Anesthesia<br />

Limited burns to limbs may be managed by an anesthetic<br />

technique that associates regional blocks with either propofol<br />

sedation or light inhalational anesthesia. This technique is very<br />

useful for skin grafting. The following blocks for the lower limb are<br />

easy to perform and often useful. The femoral nerve block will<br />

provide analgesia for the anterior aspect of the thigh and leg. The<br />

lateral cutaneous block will provide analgesia to the lateral aspect<br />

of the thigh. For the upper limb, the most performed block is the<br />

axillary plexus block. Central blocks such as caudals, epidurals, or<br />

spinals are seldom used in the initial post-burn phase, because of<br />

the loss of sympathetic tone exacerbating hypotension and heat<br />

loss. Although continuous peripheral blocks such as the facia iliaca<br />

compartment block prove to be efficacious for pain relief on donor<br />

sites after skin grafting in burned patients, 125 the same but singleblock<br />

technique has the same morphine sparing-effect during a<br />

72-hour postoperative period with limited side effects, compared<br />

to the continuous technique. 126<br />

Hematologic Management Strategies<br />

Morbidity with homologous blood transfusion is low but well<br />

documented. Transfusion of blood products should be limited.<br />

Surgical blood losses are maximal during the first 2 weeks postburn<br />

injury, 127 during which the inflammatory response is<br />

maximal. The losses range from 4 to 15% of the patient’s blood<br />

volume for every percent of skin debrided. In a more recent<br />

retrospective chart review of consecutive pediatric burn surgeries<br />

the average blood loss per percent TBSA treated was 15 mL (range:<br />

0.7–37 mL) and the average percent of total blood volume loss per<br />

percent of TBSA treated was only 0.76%. The protocol to reduce<br />

intraoperative blood loss consisted of the debridement of fullthickness<br />

burns with electrocautery and partial-thickness burns<br />

with dermabrasion. All debrided or harvested surgical sites were<br />

treated immediately with epinephrine solution-soaked pads. All<br />

graft harvest sites were injected with an epinephrine solution<br />

before harvesting split-thickness skin grafts. 128 The advantages of<br />

both early tangential excision and split-skin grafting for surgical<br />

burns are well established. In a cohort of pediatric patients with<br />

massive burns delays in excision were associated with longer<br />

hospitalization and delayed wound closure, as well as increased<br />

rates of invasive wound infection and sepsis.<br />

Early excision within 48 hours was considered optimal. 129<br />

However, because of the important associated blood loss, this<br />

technique requires an experienced team which may only be<br />

available in a few centers worldwide: other centers may have to<br />

limit the excision to as little as 10 to 30% BSA per session. The<br />

estimation of blood loss is quite difficult and requires clinical<br />

expertise. It is based practically on the close observation of the<br />

surgical field, associated with the integration of each of the values<br />

provided by the various monitoring devices mentioned above. A<br />

rising of both diastolic blood pressure and heart rate associated<br />

with a drop in the pulse oximeter amplitude is an early sign<br />

of hypovolemia and is usually treated with administration of<br />

crystalloid solutions. A drop in systolic blood pressure with a<br />

disappearing pulse oximeter trace denotes severe hypovolemia<br />

requiring the administration of colloids. Very severe hypovolemia<br />

must be suspected when capnography reveals a drop in end tidal<br />

CO 2<br />

. At this stage, if the previous mentioned filling strategies have<br />

been provided, only blood transfusion will permit restoration of<br />

hemodynamic stability. Characteristic blood clotting changes are<br />

also clinically observable; diffuse bleeding usually reflects massive<br />

hemodilution and, less frequently, a clotting disorder.<br />

What is the acceptable hemoglobin level? There is yet no<br />

definitive answer to this question, although levels of 70 to 80 g/L<br />

of hemoglobin appear safe in children and healthy adults; much<br />

lower levels have been associated with survival in Jehovah‘s<br />

Witnesses. 60 A large trial in critically ill adult patients showed<br />

that a conservative blood transfusion policy as above is safe and<br />

is possibly even associated with lower mortality than a more<br />

liberal transfusion policy. 130 Early administration of recombinant<br />

erythropoietin (300 U/kg of recombinant erythropoietin within<br />

72 hours of admission, and daily for 7 days, followed by 150 U/kg<br />

for 2 weeks) does not prevent post-burn anemia, nor does it<br />

decrease transfusion requirements. 67 Moreover, failure to provide<br />

iron supplementation in patients receiving recombinant erythropoietin<br />

can lead to a rapid depletion of iron stores and may<br />

contribute to an immune dysfunction. 131 The different treatment<br />

strategies are summarized in Table <strong>124</strong>–8.<br />

In large burns (>20 to 30% BSA; depending on site and degree<br />

of burn), it is mandatory to monitor blood pressure invasively;<br />

indeed, rapid changes in volemia are diagnosed at a glance, correlated<br />

with changes in the surface area under the pressure curve.<br />

Moreover, left ventricular contractility can be estimated on the<br />

upstroke limb of the pressure curve. A second transducer committed<br />

to on-line central venous pressure measurement is<br />

mandatory in infants and small children, since even small volumes<br />

may lead to cardiac dilatation through overload. Changes in<br />

heart rate may be the only available observation. Repeated


CHAPTER <strong>124</strong> ■ Burns and Post-Burn Care: Anesthetic Considerations 2063<br />

TABLE <strong>124</strong>-8. Treatment Strategies for Hematologic Disorders<br />

Hematologic Disorder Suggested Management Comment<br />

Anemia<br />

Prevention during surgery<br />

Established anemia:<br />

Ht


2064 PART 5 ■ Anesthetic, Surgical, and Interventional Procedures: Considerations<br />

is highly recommended. General strategies of lung protective<br />

ventilation and treatment should be applied, including positive<br />

end-expiratory pressure (PEEP), low tidal volume target, permissive<br />

hypercapnia, and permissive hypoxemia.<br />

Prolonged intubation and ventilatory support may be required<br />

with severe burns, especially those involving the face with inhalation<br />

injury. The risks involved are associated with the small size<br />

of the pediatric tubes and the difficulty in suctioning secretions, as<br />

well as the risk of accidental extubation and long-term sequelae<br />

(subglottic stenosis). In patients requiring long term ventilatory<br />

support, tracheostomy may be a way to avoid these complications.<br />

134<br />

Metabolic and Nutritional Support<br />

During the last 3 decades, awareness of the importance of nutritional<br />

support in burn patients has grown rapidly. This aspect is<br />

particularly critical in patients with prolonged hospital stays. One<br />

half of the children admitted to a burn center will stay up to<br />

1 month, and 25% will require up to 3 months. 135<br />

Antioxidant Micronutrient Therapy—<br />

Reinforcing Endogenous Defenses<br />

The normally effective endogenous antioxidant defense system is<br />

overwhelmed after major burns. In experimental burns, antioxidant<br />

vitamin supplements have been shown to limit the endothelial<br />

injury and capillary leak. Large supraphysiologic doses of<br />

vitamin C administered over the first 24 hours reduce post-burn<br />

fluid requirement by an antioxidant mechanism. 136 In animals,<br />

selenium supplements limit the peroxidative damage caused by<br />

burns. 137 In adults, supplementation with trace elements, particularly<br />

with selenium, reduces the lipid peroxidation process<br />

during the first week and reinforces endogenous antioxidant and<br />

immune defenses, 138 but has no impact on early fluid requirements.<br />

Data in children are still missing, although trace element<br />

deficiency has been shown to be caused by cutaneous exudative<br />

losses as in adults. 22<br />

Energy Expenditure<br />

Energy expenditure undergoes major changes over time after burn<br />

injury, in both adult and pediatric patients. The increase in resting<br />

energy expenditure starts around the 5th day, peaking between<br />

the 2nd and 4th week depending on burn size, then reverting<br />

progressively towards normal about 2 years after injury. 52 In<br />

children, the changes have been shown to be sex dependent, with<br />

a significantly lower energy expenditure in girls at any time after<br />

injury. 52 The endocrine response was also gender linked: girls had<br />

significantly higher levels of insulin-like growth factor 1, insulinlike<br />

growth factor binding protein 3, free thyroxin index, T4, and<br />

insulin compared with boys (P < .05).<br />

Post-burn oxandrolone has become part of the supportive<br />

metabolic treatment in major burns. In a randomized trial<br />

including 235 children with burns of more than 40% BSA, length<br />

of ICU stay was significantly decreased in the oxandrolone-treated<br />

patients (0.48 ± 0.02 days/1% burn) compared with controls (0.56<br />

± 0.02 days/1% burn) (P < .05). 52 Control patients lost 8 ± 1% of<br />

their lean body mass, whereas oxandrolone-treated patients had<br />

preserved lean body mass (9 ± 4%, P < .05). Oxandrolone significantly<br />

increased serum prealbumin, total protein, testosterone, and<br />

aspartate transaminase (ASAT)/alanine aminotransferase (ALAT),<br />

whereas it significantly decreased alpha 2-macroglobulin and<br />

complement C3 (P < .05). Oxandrolone had no adverse effect on<br />

the endocrine and inflammatory response (no significant dif -<br />

ference was observed between groups).<br />

For nearly 50 years, standard clinical management of burned<br />

patients included high-calorie and high-protein input to promote<br />

healing and maintain body weight. Overfeeding however, should<br />

be avoided as strongly as underfeeding. Recent evidence confirms<br />

the inadequacy of the hypercaloric formulas: all overestimate<br />

requirements and none take into account the evolution of energy<br />

expenditure over time. The strongest recommendations are produced<br />

by the Galveston center. They propose the Galveston formula<br />

to initiate nutrition according to age as a first step, followed<br />

by targeted adaptations according to indirect calorimetry measurements.<br />

The outdated Currerri formula should no longer be<br />

used. The variability of energy expenditure is such, that all<br />

respectable centers now recommend using indirect calorimetry<br />

and accordingly adapt feeding for prolonged periods.<br />

Nutrient Delivery<br />

Pediatric patients require artificial nutrition whenever burns<br />

exceed 20% BSA, even less with face or inhalation involvement.<br />

Early enteral nutrition has become a routine in most burn centers.<br />

Gastric or postpyloric feeding is started within 12 hours of injury<br />

by a nasoenteric tube. It is widely and successfully used in North<br />

American Centers. 139 Maintaining continuous enteral nutrition<br />

during surgical procedures contributes to increase energy intake,<br />

but this must be weighed against the risk of bronchoaspiration. In<br />

severe burns, percutaneous endoscopic gastrostomy may be a<br />

good alternative to nasoenteric tubes to facilitate nursing. The<br />

diets are those used in other pediatric trauma patients, i.e., highenergy<br />

and high-protein diets. Total parenteral nutrition is<br />

indicated for patients presenting contraindications to enteral<br />

nutrition or in whom the gut function is compromised. Parenteral<br />

nutrition requires giving particular attention to the energy target,<br />

since it is frequently easier to administer the nutrients by this route<br />

than by the enteral approach. A study in burned adults reported a<br />

higher rate of infectious complications in patients receiving<br />

parenteral nutrition supplements compared with those on<br />

pure enteral nutrition (63% vs 26%, respectively). 140 These data<br />

should be regarded with caution, since the parenteral supplements<br />

resulted in very large energy intakes, i.e., overfeeding. The<br />

deleterious effects may actually be related to this excessive caloric<br />

intake and not to the intravenous technique. Enteral nutrition is<br />

not always easy in the severely critically ill patient. It may result in<br />

insufficient energy intakes, with a risk of developing malnutrition.<br />

Combining parenteral nutrition with enteral nutrition may<br />

actually be required to reach a reasonable energy target in a<br />

minority of patients. Daily close monitoring of energy delivery is<br />

therefore required, and modern computerized systems facilitate<br />

the metabolic management as shown in Figure <strong>124</strong>–4.<br />

Impact on Liver Function<br />

Hepatic homeostasis and metabolism are essential for survival in<br />

critically ill and severely burned patients. The liver undergoes<br />

hypertrophy after burn. In a trial including 102 children with<br />

major burns, liver size was measured by means of ultrasound, and<br />

liver weight was calculated weekly during hospital stay and at 6, 9,<br />

and 12 months after burn. The liver size was then compared with


CHAPTER <strong>124</strong> ■ Burns and Post-Burn Care: Anesthetic Considerations 2065<br />

Figure <strong>124</strong>-4. Monitoring of fluid and energy balances with help of computerized systems facilitates decision making. For every<br />

24 hours (24 h per column), the figure shows energy target, enteral and intravenous energy delivery, energy balances, indirect<br />

calorimetry determinations, and protein intakes. The upper graph show daily energy balances and plasma albumin values, and<br />

the lower shows the fluid balance (full line) and evolution of weight curve.<br />

the predicted value for each individual. 52 Liver size and weight<br />

increased significantly during the first week after burn (mean<br />

85%), peaking at 2 weeks after burn (mean 126%): at discharge,<br />

the increase was still 89%. At 6, 9, and 12 months, the liver weight<br />

remained elevated by 40 to 50% compared with the predicted liver<br />

weight. The hepatic protein synthesis was affected up to 9 months<br />

after burn.<br />

Liver dysfunction is clearly multifactorial—high dose insulin<br />

and glucose have been blamed, as well as peripheral lipolysis. 54<br />

Some years back an isotopic investigation in 6 patients with severe<br />

burns showed that despite a glucose delivery rate similar to 100%<br />

in excess of energy requirements, did not alter total hepatic very<br />

low-density protein (VLDL) triglyceride (TG) secretion rate,<br />

plasma TG concentration, nor plasma VLDL TG concentration. 141<br />

Instead, the high-carbohydrate delivery in conjunction with<br />

insulin therapy nearly tripled the proportion of de novo–<br />

synthesized palmitate in VLDL TG front, with a corresponding<br />

decreased amount of palmitate from lipolysis. A 15-fold increase<br />

in plasma insulin concentration did not change the rate of release<br />

of FA into plasma. The peripheral release of FA represents a far<br />

greater potential for hepatic lipid accumulation in burn patients<br />

than the endogenous hepatic fat synthesis, even during excessive<br />

carbohydrate intake in conjunction with insulin therapy<br />

Control of hyperglycemia has been shown to decrease<br />

mortality in some categories of surgical critically ill adults, 142<br />

whereas data in children remain few but encouraging. A study<br />

in burned children with a before and after design including<br />

64 children, 143 targeted glucose 90 to 120 mg/dL in the interven -<br />

tion group showed that intensive insulin therapy was safe: it was<br />

associated with lower infection rates (urinary tract infections) and<br />

improved survival.<br />

Anabolic Agents<br />

A continued catabolic state results in weight loss, lean muscle loss,<br />

immunologic compromise, and poor or delayed wound healing<br />

with prolonged recovery times. Various efforts have been made to


2066 PART 5 ■ Anesthetic, Surgical, and Interventional Procedures: Considerations<br />

promote anabolism. Recombinant human growth hormone<br />

(rhGH) therapy has been extensively investigated: in GH-deficient<br />

children, rhGH therapy improved nitrogen balance, increased<br />

body cell mass, and promoted bone formation. 144 These effects<br />

make GH a candidate for anabolism stimulation. Supplementation<br />

studies in burned pediatric patients have shown a decrease in<br />

donor-site healing times and length of hospital stay per 1% burned<br />

BSA, and attenuation of hypermetabolism and of inflammation<br />

particularly when used in combination with propranolol. 145<br />

Although safe in children, the use of rhGH in critically ill adult<br />

patients is not warranted, because it doubles mortality, mainly<br />

from multiple organ dysfunction and septic shock. 146<br />

Insulin has well-known anabolic properties, as it promotes<br />

protein synthesis. In a group of adolescent patients, high-dose<br />

intravenous glucose along with insulin was associated with a<br />

2-day reduction in the donor-site healing time. 147 However, recent<br />

data show that high loads of glucose promote de novo lipogenesis<br />

in the critically ill. 57 In combination with data published regarding<br />

liver fat deposits on postmortem pathologic evaluation in patients<br />

receiving large glucose loads, 55 the de novo lipogenesis data<br />

indicate that using high glucose loads along with insulin should be<br />

restricted until further studies prove its safe use.<br />

Micronutrients<br />

Trace element deficiencies (particularly copper, iron, and zinc)<br />

have been known to occur in burned children since the 1970s. 138<br />

They are largely explained by exudative cutaneous losses which<br />

have now been confirmed in children. 22 Copper, which plays an<br />

essential role in collagen synthesis, wound repair, and immunity,<br />

is strongly depleted in burns. 138 Early trace element supplementation<br />

is associated with improved wound healing, decreased<br />

protein catabolism, decreased pulmonary infection, and shortened<br />

ICU stay. 138 During artificial nutrition, special attention should be<br />

given to all micronutrients; trace element and vitamin contents of<br />

enteral nutrition or of standard parenteral nutrition additives are<br />

insufficient to cover the increased requirements after burns. The<br />

standard recommendations for pediatric parenteral supplements<br />

are provided in Table <strong>124</strong>–9. 148 Additional intravenous vitamin C,<br />

vitamin E, copper, selenium, and zinc supplements should be<br />

provided. 22 Doses and combinations of micronutrients used in our<br />

institution, calculated by adapting the adult doses to the weight of<br />

the pediatric patients, are proposed in the same Table 9. Some<br />

micronutrient supplements may be provided by either the enteral<br />

or intravenous routes. On the other hand, because there is competition<br />

and antagonism for intestinal transmucosal transport,<br />

copper and zinc are best provided by the intravenous route.<br />

Calcium and Bone Metabolism<br />

Calcium, magnesium, and phosphorus homeostasis are severely<br />

altered after trauma and particularly after burns. 149 The changes<br />

are long-lasting and continue into the recovery period, many years<br />

after the injury. Bone formation is reduced both in adults and<br />

children when burns exceed 40% BSA. Bone mineral density is<br />

significantly lower in burned children compared with the same<br />

age-related normal children. Girls have improved bone mineral<br />

content and percentage fat compared with boys. 52 The consequences<br />

are increased risk of fractures, decreased growth velocity<br />

and, finally, stunting. 144 The bone is affected for multiple reasons,<br />

such as alteration of mineral metabolism, elevated cytokine and<br />

corticosteroid levels, decreased GH, nutritional deficiencies, and<br />

interoperative immobilization. Cytokines contribute to the<br />

alterations, particularly interleukin-1β and interleukin-6, which<br />

are greatly increased in burns, and stimulate osteoblast-mediated<br />

bone resorption. Cortisol production is increased in burns,<br />

leading to decreased bone formation. Low GH levels fail to<br />

promote bone formation. 144 Various studies suggest that immobilization<br />

plays a primary role in the pathogenesis of burnassociated<br />

bone disease. 150 Alterations of magnesium and calcium<br />

homeostasis constitute another cause. Hypocalcemia and hypomagnesemia<br />

are constant findings, and ionized calcium remains<br />

low for weeks 35 ; the alterations are partly explained by large<br />

TABLE <strong>124</strong>-9. Recommended Daily Intravenous Micronutrient Supplements After Burn Injury Versus in Patients<br />

Without Burns<br />

Daily Intravenous Supplements<br />

Micronutrient Dose: Major Burns (Nonburned Patient) Comment<br />

Magnesium<br />

Multitrace element solution<br />

Additional trace element<br />

supplement<br />

Multivitamin preparation<br />

Additional vitamin E<br />

Additional ascorbic acid<br />

0.25 mmol/kg<br />

Many brands available<br />

Administer daily requirements according to label<br />

4 mL per kg of a 250 mL solution containing:<br />

• Copper 3.75 mg → 60 mg/kg (20 mg/kg)<br />

• Selenium 375 µg → 6 µg/kg(1.5 µg/kg)<br />

• Zinc 37.5 mg → 600 µg/kg (100 µg/kg)<br />

• Glu-1-P 1200 mg → 19 mg/kg<br />

Many brands available<br />

Administer daily requirement according to label<br />

1.5 mg/kg 60 kg: maximum 100 mg<br />

15 mg/kg maximum 40 kg: 500 mg<br />

from 60 kg: 1000 mg<br />

BSA = body surface area; Glu-1-P = glucose-1-phosphate; arrow = yield.<br />

Both basic requirements and supplements should be administered as of admission.<br />

Partially adapted from Greene et al. 148<br />

May be given by the enteral route<br />

Start on admission<br />

Start on admission and pursue<br />

according to burns for:<br />

BSA 30%: 15 days<br />

Start on admission<br />

Start on admission<br />

May be given by the enteral route<br />

Start on admission


CHAPTER <strong>124</strong> ■ Burns and Post-Burn Care: Anesthetic Considerations 2067<br />

exudative magnesium and phosphorus losses. 149 A close followup<br />

of ionized calcium, magnesium, and inorganic phosphate is<br />

mandatory, since burn patients usually require substantial supplementation<br />

by intravenous or enteral routes.<br />

Heterotopic ossification, which is encountered only occasionally<br />

in children, is an alteration of calcium metabolism<br />

associated with an invalid state. It has not been entirely elucidated;<br />

new bone is formed in tissues that do not normally ossify. The<br />

condition is favored by immobilization. Nutritional supplements<br />

have been implicated and reported to mobilize calcium and to<br />

increase calciuria. 151 Overzealous joint manipulation causing<br />

microtrauma to the musculotendinous apparatus is also probably<br />

involved. 152<br />

Septic Complications<br />

Due to an overall depression of the immune defense system, burn<br />

patients have a high incidence of septic complications. Pulmonary<br />

and cutaneous infections are the most frequent, followed by<br />

urinary and blood stream infections. The use of prophylactic<br />

antibiotherapy in acute burns enhances the development of<br />

resistant organisms, although only minimally reducing the<br />

incidence of sepsis. There are no data showing a reduction of<br />

wound infections due to prophylaxis. Therefore, antibiotic treatment<br />

must be directed against identified germs. Children frequently<br />

have other sources of infections (e.g., upper respiratory<br />

tract infections, otitis media) that require appropriate treatment.<br />

Ethical Issues<br />

Therapy withdrawal or withholding is a sensitive question all over<br />

the world. It becomes even more difficult when children are<br />

concerned. Very little literature exists in this area. The national<br />

legislations vary from complete interdiction to tolerance of therapy<br />

withholding or withdrawal. There are, in addition, important<br />

cultural, religious, and individual factors involved that explain the<br />

large variability. Whatever the attitude toward these issues, any<br />

intensivist would agree that some victims of burns are identified<br />

early as probable nonsurvivors. Major burns involving more than<br />

80% BSA with extensive destruction of face and limbs, extremes of<br />

age, and unprecedented survival in an institution may call for such<br />

reflection. Under certain circumstances, the decision to withhold<br />

curative treatment and to focus on the patient’s comfort may be<br />

considered a treatment option, applying to both children and<br />

adults. Some institutions have communication protocols that<br />

facilitate the discussion of the topic, but most do not. Implementing<br />

“do not resuscitate” order guidelines must be based on<br />

the particular center’s experience as well as on its cultural and<br />

legislative background. When legally possible, decisions not to<br />

resuscitate or to withhold further treatment should be based on a<br />

multidisciplinary approach.<br />

CONCLUSION<br />

Care of the burned patients involves detailed knowledge of the<br />

pleiomorphic effects of burn injuries on all the organ systems. The<br />

extensive alterations will modify pharmacokinetics and pharmacodynamics<br />

of anesthetic agents. Burns are a devastating event<br />

in a child’s life: adequate analgesia and sedation and safe repeated<br />

anesthetic care, together with concern and understanding of the<br />

psychological aspects, maintenance of metabolic support to limit<br />

stunting observed after major burns, all contribute to limit the<br />

negative impact of severe burn injury on a young life. The management<br />

of these patients therefore requires a multidisciplinary<br />

approach and the constitution of a true burn team.<br />

REFERENCES<br />

1. Franco MA, Gonzales NC, Diaz ME, et al. Epidemiological and clinical<br />

profile of burn victims: Hospital Universitario San Vicente de Paul,<br />

Medellin, 1994–2004. Burns. 2006;32:1044–1051.<br />

2. Holland AJ. Pediatric burns: the forgotten trauma of childhood. Can J<br />

Surg. 2006;49:272–277.<br />

3. Langer S, Hilburg M, Drucke D, et al. Analysis of burn treatment for<br />

children at Bochum University Hospital. Unfallchirurg. 2006;109:862–866.<br />

4. Rawlins JM, Khan AA, Shenton AF, et al. Epidemiology and outcome<br />

analysis of 208 children with burns attending an emergency department.<br />

Pediatr Emerg Care. 2007;23:289–293.<br />

5. Roudsari BS, Shadman M, Ghodsi M. Childhood trauma fatality and<br />

resource allocation in injury control programs in a developing country.<br />

BMC Public Health. 2006;6:117.<br />

6. Xin W, Yin Z, Qin Z, et al. Characteristics of 1494 pediatric burn patients<br />

in Shanghai. Burns. 2006;32:613–618.<br />

7. Sheridan RL, Weber JM, Schnitzer JJ, et al. Young age is not a predictor<br />

of mortality in burns. Pediatr Crit Care Med. 2001;2:223–224.<br />

8. Thombs BD, Singh VA, Milner SM. Children under 4 years are at greater<br />

risk of mortality following acute burn injury: evidence from a national<br />

sample of 12,902 pediatric admissions. Shock. 2006;26:348–352.<br />

9. Young AE, Chambers TL. Standards for care of critically ill children with<br />

burns. Lancet. 2005;365:1911–1913.<br />

10. Finnerty CC, Herndon DN, Przkora R, et al. Cytokine expression profile<br />

over time in severely burned pediatric patients. Shock. 2006;26:13–19.<br />

11. Demling R, LaLonde C, Knox J, et al. Fluid resuscitation with deferoxamine<br />

prevents systemic burn-induced oxidant injury. J Trauma. 1991;<br />

31:538–544.<br />

12. Lund T, Wiig H, Reed RK. Acute postburn edema: role of strongly<br />

negative interstitial fluid pressure. Am J Physiol. 1988;255:H1069–1074.<br />

13. Kramer GC, Nguyen TT. Pathophysiology of burn shock and burn edema.<br />

In: Herndon D, editor. Total Burn Care. London: Saunders;1996. p. 44–52.<br />

14. Arturson G. Fluid therapy of thermal injury. Acta Anaesthesiol Scand.<br />

1985;29:55–59.<br />

15. Barber AE, Shires GT. Cell damage after shock. New Horizons. 1996;4:<br />

161–166.<br />

16. Gravante G, Delogu D, Sconocchia G. “Systemic apoptotic response” after<br />

thermal burns. Apoptosis. 2007;12:259–270.<br />

17. Carlson DL, Horton JW. Cardiac molecular signaling after burn trauma.<br />

J Burn Care Res. 2006;27:669–675.<br />

18. Marano MA, Fong Y, Moldawer LL, et al. Serum cachectin: tumor<br />

necrosis factor in critically ill patients with burns correlates with infection<br />

and mortality. Surg Gynecol Obstet. 1990;170:32–38.<br />

19. Marshall JC, Vincent JL, Fink MP, et al. Measures, markers, and mediators:<br />

toward a staging system for clinical sepsis. A report of the Fifth<br />

Toronto Sepsis Roundtable, Toronto, Ontario, Canada, October 25–26,<br />

2000. Crit Care Med. 2003;31:1560–1567.<br />

20. Allgöwer M, Schoenenberger GA, Sparkes BG. Burning the largest<br />

immune organ. Burns. 1995;21:S7–S47.<br />

21. Yamamoto H, Siltharm S, deSerres S, et al. Immediate burn wound excision<br />

restores antibody synthesis to bacterial antigen. J Surg Res. 1996;63:157–162.<br />

22. Berger MM. Acute copper and zinc deficiency due to exudative losses—<br />

substitution versus nutritional requirements. Burns. 2006;32:393.<br />

23. Demling RH. Fluid replacement in burned patients. Surg Clin North Am.<br />

1987;67:15–30.<br />

24. Carleton SC. Cardiac problems associated with burns. Cardiol Clin.<br />

1995;13:257–262.<br />

25. Zeng L, Chen Y, Wu M. Cardiac troponin I: a marker for detecting nonischemic<br />

cardiac injury. Zhonghua Yi Xue Za Zhi. 2001;81:393–395.<br />

26. Raffa J, Trunkey DD. Myocardial depression in acute thermal injury.<br />

J Trauma. 1978;18:90–93.<br />

27. Zang Q, Maass DL, White J, et al. Cardiac mitochondrial damage and loss<br />

of ROS defense after burn injury: the beneficial effects of antioxidant<br />

therapy. J Appl Physiol. 2007;102:103–112.


2068 PART 5 ■ Anesthetic, Surgical, and Interventional Procedures: Considerations<br />

28. Yatani A, Xu DZ, Irie K, et al. Dual effects of mesenteric lymph isolated<br />

from rats with burn injury on contractile function in rat ventricular<br />

myocytes. Am J Physiol Heart Circ Physiol. 2006;290:H778–785.<br />

29. Goto M, Samonte V, Ravindranath T, et al. Burn injury exacerbates<br />

hemodynamic and metabolic responses in rats with polymicrobial sepsis.<br />

J Burn Care Res. 2006;27:50–59.<br />

30. Niederbichler AD, Westfall MV, Su GL, et al. Cardiomyocyte function<br />

after burn injury and lipopolysaccharide exposure: single-cell contraction<br />

analysis and cytokine secretion profile. Shock. 2006;25:176–183.<br />

31. Quinn DA, Moufarrej R, Volokhov A, et al. Combined smoke inhalation<br />

and scald burn in the rat. J Burn Care Rehabil. 2003;24:208–216.<br />

32. Ren J, Wu S. A burning issue: do sepsis and systemic inflammatory<br />

response syndrome (SIRS) directly contribute to cardiac dysfunction?<br />

Front Biosci. 2006;11:15–22.<br />

33. White J, Thomas J, Maass DL, et al. Cardiac effects of burn injury<br />

complicated by aspiration pneumonia-induced sepsis. Am J Physiol Heart<br />

Circ Physiol. 2003;285:H47–58.<br />

34. Reynolds EM, Ryan DP, Sheridan RL, et al. Left ventricular failure<br />

complicating severe pediatric burn injuries. J Pediatr Surg. 1995;30:264–<br />

269; discussion 269–270.<br />

35. Szyfelbein SK, Drop LJ, Martyn JAJ. Persistent ionized hypocalcemia in<br />

patients during resuscitation and recovery phases of body burns. Crit Care<br />

Med. 1981;9:454–458.<br />

36. Bargues L, Vaylet F, Le Bever H, et al. Respiratory dysfunction in burned<br />

patients. Rev Mal Respir. 2005;22:449–460.<br />

37. Fitzpatrick JC, Cioffi WG. Inhalation injury. Trauma Quart. 1994;11:114–<br />

126.<br />

38. Turnage RH, Nwariaku F, Murphy J, et al. Mechanisms of pulmonary<br />

microvascular dysfunction during severe burn injury. World J Surg.<br />

2002;26:848–853.<br />

39. Steinvall I, Bak Z, Sjoberg F. Acute respiratory distress syndrome is as<br />

important as inhalation injury for the development of respiratory<br />

dysfunction in major burns. Burns. 2008;.<br />

40. Ciesla DJ, Moore EE, Johnson JL, et al. The role of the lung in postinjury<br />

multiple organ failure. Surgery. 2005;138:749–757; discussion 757–748.<br />

41. Barrow RE, Jeschke MG, Herndon DN. Early fluid resuscitation improves<br />

outcomes in severely burned children. Resuscitation. 2000;45(2):91–96.<br />

42. Jeschke MG, Barrow RE, Wolf SE, et al. Mortality in burned children with<br />

acute renal failure. Arch Surg. 1998;133:752–756.<br />

43. Haberal MA. An eleven-year survey of electrical burn injuries. J Burn<br />

Care Rehabil. 1995;16:43–48.<br />

44. Gore DC, Hawkins HK, Chinkes DL, et al. Assessment of adverse events<br />

in the demise of pediatric burn patients. J Trauma-Injury Infect Crit Care.<br />

2007;63:814–818.<br />

45. Myers SI, Minei JP, Casteneda A, et al. Differential effects of acute thermal<br />

injury on rat splanchnic and renal blood flow and prostanoid release.<br />

Prostaglandins Leukotrienes Essential Fatty Acids. 1995;53:439–444.<br />

46. Gore DC, Dalton JM, Gehr TWB. Colloid infusions reduce glomerular<br />

filtration in resuscitated burn victims. J Trauma. 1996;40:356–360.<br />

47. Pruitt BA Jr, Foley FD, Moncrief JA. Curling’s ulcer: a clinical-pathology<br />

study of 323 cases. Ann Surg. 1970;172:523–539.<br />

48. Namias N. Advances in burn care. Curr Opin Crit Care. 2007;13(4):405–<br />

410.<br />

49. Ivy ME, Atweh NA, Palmer J, et al. Intra-abdominal hypertension and<br />

abdominal compartment syndrome in burn patients. J Trauma.<br />

2000;49:387–391.<br />

50. Raff T, Hartmann B, Germann G. Early intragastric feeding of seriously<br />

burned and long-term ventilated patients: a review of 55 patients. Burns.<br />

1997;23:19–25.<br />

51. Cunningham JJ. Factors contributing to increase energy expenditure in<br />

thermal injury: a review of studies employing indirect calorimetry. JPEN.<br />

1990;14:649–656.<br />

52. Jeschke MG, Finnerty CC, Suman OE, et al. The effect of oxandrolone on<br />

the endocrinologic, inflammatory, and hypermetabolic responses during<br />

the acute phase postburn. Ann Surg. 2007;246:351–360; discussion 360–<br />

352.<br />

53. Pereira CT, Murphy KD, Herndon DN. Altering metabolism. J Burn Care<br />

Rehabil. 2005;26(3):194–199.<br />

54. Aarsland A, Chinkes D, Wolfe RR, et al. Beta-blockade lowers peripheral<br />

lipolysis in burn patients receiving growth hormone. Ann Surg.<br />

1996;223:777–789.<br />

55. Burke JF, Wolfe RR, Mullany CJ, et al. Glucose requirements following<br />

burn injury. Ann Surg. 1979;190:274–285.<br />

56. Wolfe RR, Klein S, Herndon DN, et al. Substrate cycling in thermogenesis<br />

and amplification of net substrate flux in human volunteers and burned<br />

patients. J Trauma. 1990;30:S6–S9.<br />

57. Tappy L, Schwarz JM, Schneiter P, et al. Effects of isoenergetic glucosebased<br />

or lipid-based parenteral nutrition on glucose metabolism, de novo<br />

lipogenesis, and respiratory gas exchanges in critically ill patients. Crit<br />

Care Med. 1998;26:860–867.<br />

58. Wolf SE, Debroy M, Herndon DN. The cornerstones and directions of<br />

pediatric burn care. Pediatr Surg Int. 1997;12:312–320.<br />

59. Shiozaki T, Kishikawa M, Hiraide A, et al. Recovery from postoperative<br />

hypothermia predicts survival in extensively burned patients. Am J Surg.<br />

1993;165:326–330.<br />

60. Donner B, Tryba M, Kurz-Muller K, et al. Anesthesia and intensive care<br />

management of severely burned children of Jehovah’s Witnesses.<br />

Anaesthesist. 1996;45:171–517.<br />

61. Harries RHC, Phillips LG. Hematologic and acute phase response. In:<br />

Herndon D, editor. Total Burn Care. London: Saunders;1996. p. 293–301.<br />

62. Belmonte Torras JA, Marin de la Cruz D, Sune Garcia JM, et al. Burns<br />

produced by lighters. An Pediatr (Barc). 2006;64:468–473.<br />

63. Carter EA, Tompkins RG, Babich JW, et al. Decreased cerebral glucose<br />

utilization in rats during the ebb phase of thermal injury. J Trauma.<br />

1996;40:930–935.<br />

64. Zhang Q, Carter EA, Ma B, et al. Burn-related metabolic and signaling<br />

changes in rat brain. J Burn Care Res. 2008;29:346–352.<br />

65. Hettiaratchy S, Dziewulski P. ABC of burns: pathophysiology and types of<br />

burns. BMJ. 2004;328:1427–1429.<br />

66. Chen EH, A Sareen. Do children require ECG evaluation and inpatient<br />

telemetry after household electrical exposures? Ann Emerg Med. 2007;<br />

49:64–67.<br />

67. Still JM Jr, Belcher K, Law EJ, et al. A double-blinded prospective<br />

evaluation of recombinant human erythropoietin in acutely burned<br />

patients. J Trauma. 1995;38:233–236.<br />

68. Scalfaro P, Haenggi MH, Roulet E, et al. Carbon monoxide poisoning in<br />

children: never trivialize. Rev Med Suisse Romande. 2000;120:259–262.<br />

69. Megarbane B, Delahaye A, Goldgran-Toledano D, et al. Antidotal<br />

treatment of cyanide poisoning. J Chin Med Assoc. 2003;66:193–203.<br />

70. Borron SW, Baud FJ, Barriot P, et al. Prospective study of hydroxocobalamin<br />

for acute cyanide poisoning in smoke inhalation. Ann Emerg<br />

Med. 2007;49:794–801, 801 e791–792.<br />

71. Fortin JL, Giocanti JP, Ruttimann M, et al. Prehospital administration of<br />

hydroxocobalamin for smoke inhalation-associated cyanide poisoning:<br />

8 years of experience in the Paris Fire Brigade. Clin Toxicol (Phila). 2006;44<br />

Suppl 1:37–44.<br />

72. MacLennan N, Heimbach DM, Cullen BF. Anesthesia for major thermal<br />

injury. Anesthesiology. 1998;89:749–770.<br />

73. Warden GD. Fluid resuscitation and early management. In: Herndon D,<br />

editor. Total Burn Care. London: Saunders;1996. p. 53–60.<br />

74. Fakler U, Pauli Ch, Balling G, et al. Cardiac index monitoring by pulse<br />

contour analysis and thermodilution after pediatric cardiac surgery.<br />

J Thorac Cardiovasc Surg. 2007;133(1):224–228.<br />

75. Barillo DJ. Planning for burn mass casualty incidents. J Trauma. 2007;62<br />

(suppl):S68.<br />

76. Sørensen B. Management of burns occurring as mass casualties after<br />

nuclear explosion. Burns. 1980;6.<br />

77. Berger MM, Eggimann P, Heyland DK, et al. Reduction of nosocomial<br />

pneumonia after major burns by trace element supplementation: aggregation<br />

of two randomised trials. Crit Care. 2006;10:R153:e-pub 2 Nov.<br />

78. Brown TCK, Fisk GC. Burns. In: Brown T, Fisk G, editors. Anesthesia for<br />

Children. 2nd ed. London: Blackwell Scientific Publication; 1992. p. 281–290.<br />

79. Pearson KS, From RP, Symreng T, et al. Continuous enteral feeding and<br />

short fasting periods enhance perioperative nutrition in patients with<br />

burns. J Burn Care Rehabil. 1992;13:477–481.<br />

80. Snelling CFT. Quantitative evaluation of blood loss during debridement<br />

and grafting in burns. Can J Surg. 1982;25:416–417.<br />

81. Goldstein B, Doody D, Briggs S. Emergency intraosseous infusion in<br />

severely burned children. Pediatr Emerg Care. 1990;6:195–197.<br />

82. Sheridan RL. Uncuffed endotracheal tubes should not be used in seriously<br />

burned children. Pediatr Crit Care Med. 2006;7:258–259.<br />

83. Milner EA, Cioffi WG, Mason AD, et al. A longitudinal study of resting<br />

energy expenditure in thermally injured patients. J Trauma. 1994;37:167–<br />

170.<br />

84. Martyn J. Clinical pharmacology and drug therapy in the burned patient.<br />

Anesthesiology. 1986;65:67–75.


CHAPTER <strong>124</strong> ■ Burns and Post-Burn Care: Anesthetic Considerations 2069<br />

85. Fee JP, Thompson GH. Comparative tolerability profiles of the inhaled<br />

anaesthetics. Drug Saf. 1997;16:157–170.<br />

86. Adams HA, Werner C. From the racemate to the eutomer: (S)-ketamine.<br />

Renaissance of a substance? Anaesthesist. 1997;46:1026–1042.<br />

87. Tosun Z, Esmaoglu A, Coruh A. Propofol-ketamine vs propofol-fentanyl<br />

combinations for deep sedation and analgesia in pediatric patients<br />

undergoing burn dressing changes. Paediatr Anaesth. 2008;18:43–47.<br />

88. Ceber M, Salihoglu T. Ketamine may be the first choice for anesthesia in<br />

burn patients. J Burn Care Res. 2006;27:760–762.<br />

89. Carsin H. Use of Diprivan in burn patients. Ann Fr Anesth Reanim.<br />

1994;13:541–544.<br />

90. Murat I, Billard V, Vernois J, et al. Pharmacokinetics of propofol after a<br />

single dose in children aged 1–3 years with minor burns. Comparison of<br />

three data analysis approaches. Anesthesiology. 1996;84:526–532.<br />

91. Han TH, Lee JH, Kwak IS, et al. The relationship between bispectral<br />

index and targeted propofol concentration is biphasic in patients with<br />

major burns. Acta Anaesthesiol Scand. 2005;49:85–91.<br />

92. Fodale V, La Monaca E. Propofol infusion syndrome: an overview of a<br />

perplexing disease. Drug Saf. 2008;31:293–303.<br />

93. Bray RJ. Propofol infusion syndrome in children. Paediatr Anaesth.<br />

1998;8:491–499.<br />

94. Honnegger P, Matthieu JM. Selective toxicity of the general anesthetic<br />

propofol for GABAergic neurons in rat brain cell cultures. J Neuroscience<br />

Res. 1996;45:631–636.<br />

95. Escarment J, Cantais E, Le Dantec P, et al. Propofol and ketamine for<br />

dressing in burnt patients. Can Anesthesiol. 1995;43:31–34.<br />

96. Yagmurdur H, Akca G, Aksoy M, et al. The effects of ketamine and<br />

propofol on bacterial translocation in rats after burn injury. Acta<br />

Anaesthesiol Scand. 2005;49:177–182.<br />

97. Coimbra C, Choiniere M, Hemmerling TM. Patient-controlled sedation<br />

using propofol for dressing changes in burn patients: a dose-finding<br />

study. Anesth Analg. 2003;97:839–842.<br />

98. Martyn JA, White DA, Gronert GA, et al. Up-and-down regulation of<br />

skeletal muscle acetylcholine receptors. Effects on neuromuscular<br />

blockers. Anesthesiology. 1992;76:822–843.<br />

99. Ward JM, Rosen KM, Martyn JAJ. Acetylcholine receptor subunit<br />

mRNA changes in burns are different from those seen after denervation:<br />

The 1993 Lindberg award. J Burn Care Rehabil. 1993;14:595–601.<br />

100. Belin RP, Karleen CI. Cardiac arrest in the burned patient following<br />

succinyldicholine administration. Anesthesiology. 1966;27:516–518.<br />

101. Bush GH, Graham HA, Littlewood AH, et al. Danger of suxamethonium<br />

and endotracheal intubation in anaesthesia for burns. Br Med J. 1962;<br />

2:1081–1085.<br />

102. Forrest T. A report on two cases of cardiac arrest. Brit J Anaesth. 1959;31:<br />

277–279.<br />

103. Tolmie JD, Joyce TH, Mitchell GD. Succinylcholine danger in the burned<br />

patient. Anesthesiology. 1967;28:467–470.<br />

104. Moore GP, Pace SA, Busby W. Comparison of intraosseous, intramuscular,<br />

and intravenous administration of succinylcholine [Letters]. 1989;<br />

5:209–210.<br />

105. Mills AK, Martyn JAJ. Neuromuscular blockade with vecuronium in<br />

paediatric patients with burn injury. Br J Clin Pharmacol. 1989;28:155–<br />

159.<br />

106. Badetti C, Pascal L, Bernini V, et al. Resistance to vecuronium in burnt<br />

patients. Influence of the burnt surface on the effectiveness of the dose.<br />

Ann Fr Anesth Reanim. 1996;15:135–141.<br />

107. Uyar M, Hepaguslar H, Ugur G, et al. Resistance to vecuronium in<br />

burned children. Paediatr Anaesth. 1999;9:115–118.<br />

108. Han T, Kim H, Bae J, et al. Neuromuscular pharmacodynamics of rocuronium<br />

in patients with major burns. Anesth Analg. 2004;99:386–392.<br />

109. Werba AE, Neiger FX, Bayer GS, et al. Pharmacodynamics of mivacurium<br />

in severely burned patients. Burns. 1995;22:62–64.<br />

110. Martyn JAJ, Szyfelbein SK, Sheridan R, et al. Burned patients are not<br />

resistant to the neuromuscular effetcs of mivacurium. Europ J Anaesth.<br />

1997;14 (Suppl.6):1.<br />

111. Choinière M. Pain of burns. In: Wall P, Melzack R, editors. Textbook of<br />

Pain. 3rd ed. London: Churchill Livingstone; 1994.<br />

112. Ashburn M. A. Burn pain: the management of procedure-related pain.<br />

J Burn Care Rehabil. 1995;16:365–371.<br />

113. Friedland LR, Pancioli AM, Duncan KM. Pediatric emergency<br />

department analgesic practice. Pediatr Emerg Care. 1997;13:103–106.<br />

114. Zernikow B, Burk G, Michel E. Pain therapy in acute pediatric diseases<br />

and vaccination. Schmerz. 2000;14:319–323.<br />

115. Jellish WS, Gamelli RL, Furry PA, et al. Effect of topical local anesthetic<br />

application to skin harvest sites for pain management in burn patients<br />

undergoing skin-grafting procedures. Ann Surg. 1999;229:115–120.<br />

116. Beushausen T, Mucke K. Anesthesia and pain management in pediatric<br />

burn patients. Pediatr Surg Int. 1997;12:327–333.<br />

117. Foyatier JL, Latarjet J, Comparin JP, et al. Treatment of burns in infants.<br />

Arch Pediatr. 1995;2:1000–1006.<br />

118. Hughes J, Leach HJ, Choonara I. Hallucinations on withdrawal of<br />

isoflurane used as sedation. Acta Paediat. 1993;82:885–886.<br />

119. Cederholm I, Bengtsson M, Björkman S, et al. Long term high dose<br />

morphine, ketamine and midazolam infusion in a child with burns.<br />

Br J Clin Pharmacol. 1990;30:901–905.<br />

120. Martyn J, Greenblatt DJ. Lorazepam conjugation is unimpaired in burn<br />

trauma. Clin Pharmacol Ther. 1988;43:250–255.<br />

121. Rice TL, Kyf JV. Intranasal administration of midazolam to a severely<br />

burned child. Burns. 1990;16:307–308.<br />

122. Choinière M, Grenier R, Paquette C. Patient-controlled analgesia: a<br />

double-blind study in burn patients. Anaesthesia. 1992;47:467–472.<br />

123. Patterson DR, Hoffman HG, Weichman SA, et al. Optimizing control of<br />

pain from severe burns: a literature review. Am J Clin Hypn. 2004;47:43–54.<br />

<strong>124</strong>. de Jong AE, Middelkoop E, Faber AW, et al. Non-pharmacological<br />

nursing interventions for procedural pain relief in adults with burns: a<br />

systematic literature review. Burns. 2007;33:811–827.<br />

125. Cuignet O, Pirson J, Boughrouph J, et al. The efficacy of continuous<br />

fascia iliaca compartment block for pain management in burn patients<br />

undergoing skin grafting procedures. Anesth Analg. 2004;98:1077–1081.<br />

126. Cuignet O, Mbuyamba J, Pirson J. The long-term analgesic efficacy of a<br />

single-shot fascia iliaca compartment block in burn patients undergoing<br />

skin-grafting procedures. J Burn Care Rehabil. 2005;26:409–415.<br />

127. Steadman PB, Pegg SP. A quantitative assessment of blood loss in burn<br />

wound excision and grafting. Burns. 1992;18:490–491.<br />

128. Losee J.E, Fox I, Hua LB, et al. Transfusion-free pediatric burn surgery:<br />

techniques and strategies. Ann Plast Surg. 2005;54:165–171.<br />

129. Xiao-Wu W, Herndon DN, Spies M, et al. Effects of delayed wound excision<br />

and grafting in severely burned children. Arch Surg. 2002;137:1049–1054.<br />

130. Hébert PC, Blajchman MA, Marshall J, et al. A multicenter, randomized,<br />

controlled clinical trial of transfusion requirements in critical care. New<br />

Engl J Med. 1999;340:409–417.<br />

131. Fleming RY, Herndon DN, Vaidya S, et al. The effect of erythropoietin<br />

in normal healthy volunteers and pediatric patients with burn injuries.<br />

Surgery. 1992;112:424–431; discussion 431–422.<br />

132. Cartotto R, Kadikar N, Musgrave MA, et al. What are the acute<br />

cardiovascular effects of subcutaneous and topical epinephrine for<br />

hemostasis during burn surgery? J Burn Care Rehabil. 2003;24:297–305.<br />

133. McGill V, Kowal-Vern A, Gamelli RL. A conservative thermal injury<br />

treatment protocol for the appropriate Jehovah’s Witness candidate.<br />

J Burn Care Rehabil. 1997;18:133–138.<br />

134. Barret JP, Desai MH, Herndon DN. Effects of tracheostomies on<br />

infection and airway complications in pediatric burn patients. Burns.<br />

2000;26:190–193.<br />

135. Murphy J, Purdue G, Hunt J, et al, editors. Burn Injury. 2nd ed. New<br />

York: Churchill Livingstone; 1997.<br />

136. Tanaka H, Matsuda T, Miyagantani Y, et al. Reduction of resuscitation<br />

fluid volumes in severely burned patients using ascorbic acid administration.<br />

Arch Surg. 2000;135:326–331.<br />

137. Agay D, Sandre C, Ducros V, et al. Optimization of selenium status by a<br />

single intra-peritoneal injection of Se in Se deficient rat: possible application<br />

to burned patient treatment. Free Rad Biol Med. 2005;39:762–768.<br />

138. Berger MM, Baines M, Raffoul W, et al. Trace element supplements after<br />

major burns modulate antioxidant status and clinical course by way of<br />

increased tissue trace element concentration. Am J Clin Nutr. 2007;85:<br />

1293–1300.<br />

139. Greenhalgh DG, Housinger TA, Kagan RJ, et al. Maintenance of serum<br />

albumin levels in pediatric burn patients: a prospective randomized trial.<br />

J Trauma. 1995;39:67–74.<br />

140. Herndon DN, Barrow RE, Stein M, et al. Increased mortality with<br />

intravenous supplemental feeding in severely burned patients. J Burn<br />

Care Rehabil. 1989;10:309–313.<br />

141. Aarsland A, Chinkes DL, Sakurai Y, et al. Insulin therapy in burn patients<br />

does not contribute to hepatic triglyceride production. J Clin Invest.<br />

1998;101:2233–2239.<br />

142. Van den Berghe G, Wouters P, Weekers F, et al. Intensive insulin therapy<br />

in critically ill patients. New Engl J Med. 2001;345:1359–1367.


2070 PART 5 ■ Anesthetic, Surgical, and Interventional Procedures: Considerations<br />

143. Pham TN, Warren AJ, Phan HH, et al. Impact of tight glycemic control<br />

in severely burned children. J Trauma. 2005;59:1148–1154.<br />

144. Klein GL, Wolf SE, Langman CB, et al. Effect of therapy with recombinant<br />

human growth hormone on insulin-like growth factor system<br />

components and serum levels of biochemical markers of bone formation<br />

in children after severe burn injury. J Clin Endocrinol Metab. 1998;83:<br />

21–24.<br />

145. Jeschke MG, Finnerty CC, Kulp GA, et al. Combination of recombinant<br />

human growth hormone and propranolol decreases hypermetabolism<br />

and inflammation in severely burned children. Pediatr Crit Care Med.<br />

2008;9:209–216.<br />

146. Takala J, Ruokonen E, Webster NR, et al. Increased mortality associated<br />

with growth hormone treatment in critically ill adults. New Engl J Med.<br />

1999;341:785–792.<br />

147. Pierre EJ, Barrow RE, Hawkins HK, et al. Effects of insulin on wound<br />

healing. J Trauma. 1998;44:342–345.<br />

148. Greene HL, Hambidge KM, Schanler R, et al. Guidelines for the use<br />

of vitamins, trace elements, calcium, magnesium, and phosphorus in<br />

infants and children receiving total parenteral nutrition: Report of the<br />

subcommittee pn pediatric parenteral nutrition from the Committee on<br />

Clinical Practice Issues of the American Society for Clinical Nutrition.<br />

Am J Clin Nutr. 1988;48:1324.<br />

149. Berger MM, Rothen C, Cavadini C, et al. Exudative mineral losses after<br />

serious burns: a clue to the alterations of magnesium and phosphate<br />

metabolism. Am J Clin Nutr. 1997;65:1473–1481.<br />

150. Klein GL, Kikuchi Y, Sherrard DJ, et al. Burn-associated bone disease in<br />

sheep: roles of immobilization and endogenous corticosteroids. J Burn<br />

Care Rehabil. 1996;17:518–521.<br />

151. Evans B. Heterotopic bone formation in thermal burns. Clin Orthop<br />

Related Res. 1991;263:94–101.<br />

152. Peterson SL, Mani MM, Crawford CM, et al. Postburn heterotopic<br />

ossification: insights for management decision making. J Trauma. 1989;<br />

29:365–369.<br />

153. Weinbroum AA, Helpern P, Rudick Sorkine, P, et al. Midazolam versus<br />

proprofol for long-term sedation in the ICU: a randomized prospective<br />

comparison. Intensive Care Med. 1997;23:1258–1263.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!